Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Immunol ; 264: 110217, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38621471

RESUMEN

Common variable immunodeficiency (CVID) is an immune defect characterized by hypogammaglobulinemia and impaired development of B cells into plasma cells. As follicular helper T cells (TFH) play a central role in humoral immunity, we examined TFH cells in CVID, and investigated whether an inducible T cell co-stimulator (ICOS) agonist, vopratelimab, could modulate TFH, B cell interactions and enhance immunoglobulin production. CVID subjects had decreased TFH17 and increased TFH1 subsets; this was associated with increased transitional B cells and decreased IgG+ B and IgD-IgM-CD27+ memory B cells. ICOS expression on CVID CD4+ T cells was also decreased. However, ICOS activation of CD4+ T cells by vopratelimab significantly increased total CVID TFH, TFH2, cell numbers, as well as IL-4, IL-10 and IL-21 secretion in vitro. Vopratelimab treatment also increased plasma cells, IgG+ B cells, reduced naïve & transitional B cells and significantly increased IgG1 secretion by CVID B cells. Interestingly, vopratelimab treatment also restored IgA secretion in PBMCs from several CVID patients who had a complete lack of endogenous serum IgA. Our data demonstrate the potential of TFH modulation in restoring TFH and enhancing B cell maturation in CVID. The effects of an ICOS agonist in antibody defects warrants further investigation. This biologic may also be of therapeutic interest in other clinical settings of antibody deficiency.


Asunto(s)
Linfocitos B , Inmunodeficiencia Variable Común , Proteína Coestimuladora de Linfocitos T Inducibles , Células T Auxiliares Foliculares , Humanos , Inmunodeficiencia Variable Común/inmunología , Inmunodeficiencia Variable Común/tratamiento farmacológico , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Linfocitos B/inmunología , Linfocitos B/efectos de los fármacos , Femenino , Masculino , Persona de Mediana Edad , Adulto , Células T Auxiliares Foliculares/inmunología , Células T Auxiliares Foliculares/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Inmunoglobulina G/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Anciano , Adulto Joven
2.
Mol Cancer Ther ; 22(4): 471-484, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36780212

RESUMEN

Tumor-associated macrophages (TAM) play an important role in maintaining the immunosuppressive state of the tumor microenvironment (TME). High levels of CD163+ TAMs specifically are associated with poor prognosis in many solid tumor types. Targeting TAMs may represent a key approach in development of the next generation of cancer immune therapeutics. Members of the leukocyte immunoglobulin-like receptor B (LILRB) family, including LILRB2 (ILT4), are known to transmit inhibitory signals in macrophages and other myeloid cells. Leveraging bulk and single cell RNA-sequencing datasets, as well as extensive immunophenotyping of human tumors, we found that LILRB2 is highly expressed on CD163+ CD11b+ cells in the TME and that LILRB2 expression correlates with CD163 expression across many tumor types. To target LILRB2, we have developed JTX-8064, a highly potent and selective antagonistic mAb. JTX-8064 blocks LILRB2 binding to its cognate ligands, including classical and nonclassical MHC molecules. In vitro, JTX-8064 drives the polarization of human macrophages and dendritic cells toward an immunostimulatory phenotype. As a result, human macrophages treated with a LILRB2 blocker are reprogrammed to increase the activation of autologous T cells in co-culture systems. Furthermore, JTX-8064 significantly potentiates the activity of anti-PD-1 in allogeneic mixed lymphocyte reaction. In a human tumor explant culture, pharmacodynamic activity of JTX-8064 was observed in monotherapy and in combination with anti-PD-1. Collectively, our work provides strong translational and preclinical rationale to target LILRB2 in cancer.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Macrófagos/metabolismo , Activación de Linfocitos , Técnicas de Cocultivo , Linfocitos T , Microambiente Tumoral , Glicoproteínas de Membrana/genética , Receptores Inmunológicos
3.
Genes Dev ; 35(13-14): 1035-1054, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34168040

RESUMEN

G9a is a histone methyltransferase responsible for the dimethylation of histone H3 at lysine 9 (H3K9me2). G9a plays key roles in transcriptional silencing of developmentally regulated genes, but its role in X-chromosome inactivation (XCI) has been under debate. Here, we uncover a female-specific function of G9a and demonstrate that deleting G9a has a disproportionate impact on the X chromosome relative to the rest of the genome. G9a deficiency causes a failure of XCI and female-specific hypersensitivity to drug inhibition of H3K9me2. We show that G9a interacts with Tsix and Xist RNAs, and that competitive inhibition of the G9a-RNA interaction recapitulates the XCI defect. During XCI, Xist recruits G9a to silence X-linked genes on the future inactive X. In parallel on the future Xa, Tsix recruits G9a to silence Xist in cis Thus, RNA tethers G9a for allele-specific targeting of the H3K9me2 modification and the G9a-RNA interaction is essential for XCI.


Asunto(s)
Cromosomas Humanos X , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Metiltransferasas , ARN Largo no Codificante , Femenino , Histonas/metabolismo , Humanos , Metiltransferasas/genética , ARN Largo no Codificante/genética , Inactivación del Cromosoma X/genética
4.
Blood Adv ; 2(21): 2904-2916, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30396910

RESUMEN

The main complication of replacement therapy with factor in hemophilia A (HemA) is the formation of inhibitors (neutralizing anti-factor VIII [FVIII] antibodies) in ∼30% of severe HemA patients. Because these inhibitors render replacement FVIII treatment essentially ineffective, preventing or eliminating them is of top priority in disease management. The extended half-life recombinant FVIII Fc fusion protein (rFVIIIFc) is an approved therapy for HemA patients. In addition, it has been reported that rFVIIIFc may induce tolerance to FVIII more readily than FVIII alone in HemA patients that have developed inhibitors. Given that the immunoglobulin G1 Fc region has the potential to interact with immune cells expressing Fc receptors (FcRs) and thereby affect the immune response to rFVIII, we investigated how human macrophages, expressing both FcRs and receptors reported to bind FVIII, respond to rFVIIIFc. We show herein that rFVIIIFc, but not rFVIII, uniquely skews macrophages toward an alternatively activated regulatory phenotype. rFVIIIFc initiates signaling events that result in morphological changes, as well as a specific gene expression and metabolic profile that is characteristic of the regulatory type Mox/M2-like macrophages. Further, these changes are dependent on rFVIIIFc-FcR interactions. Our findings elucidate mechanisms of potential immunomodulatory properties of rFVIIIFc.


Asunto(s)
Factor VIII/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Células Cultivadas , Factor VIII/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Hemofilia A/tratamiento farmacológico , Hemofilia A/patología , Humanos , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Leucocitos Mononucleares/citología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/efectos de los fármacos
5.
PLoS One ; 12(10): e0186073, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29020082

RESUMEN

Genome-wide linkage analysis studies (GWAS) studies in systemic lupus erythematosus (SLE) identified the 1q23 region on human chromosome 1, containing the Signaling Lymphocytic Activation Molecule Family (SLAMF) cluster of genes, as a lupus susceptibility locus. The SLAMF molecules (SLAMF1-7) are immunoregulatory receptors expressed predominantly on hematopoietic cells. Activation of cells of the adaptive immune system is aberrant in SLE and dysregulated expression of certain SLAMF molecules has been reported. We examined the expression of SLAMF1-7 on peripheral blood T cells, B cells, monocytes, and their respective differentiated subsets, in patients with SLE and healthy controls in a systematic manner. SLAMF1 levels were increased on both T cell and B cells and their differentiated subpopulations in patients with SLE. SLAMF2 was increased on SLE CD4+ and CD8+ T cells. The frequency of SLAMF4+ and SLAMF7+ central memory and effector memory CD8+ T cells was reduced in SLE patients. Naïve CD4+ and CD8+ SLE T cells showed a slight increase in SLAMF3 levels. No differences were seen in the expression of SLAMF5 and SLAMF6 among SLE patients and healthy controls. Overall, the expression of various SLAMF receptors is dysregulated in SLE and may contribute to the immunopathogenesis of the disease.


Asunto(s)
Perfilación de la Expresión Génica , Lupus Eritematoso Sistémico/genética , Células Madre de Sangre Periférica/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Adulto , Anciano , Estudios de Casos y Controles , Diferenciación Celular/genética , Humanos , Lupus Eritematoso Sistémico/metabolismo , Subgrupos Linfocitarios/metabolismo , Persona de Mediana Edad , Monocitos/metabolismo , Células Madre de Sangre Periférica/patología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Adulto Joven
6.
Arthritis Rheumatol ; 69(5): 1035-1044, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28076903

RESUMEN

OBJECTIVE: Effector CD8+ T cell function is impaired in systemic lupus erythematosus (SLE) and is associated with a compromised ability to fight infections. Signaling lymphocytic activation molecule family member 7 (SLAMF7) engagement has been shown to enhance natural killer cell degranulation. This study was undertaken to characterize the expression and function of SLAMF7 on CD8+ T cell subsets isolated from the peripheral blood of SLE patients and healthy subjects. METHODS: CD8+ T cell subset distribution, SLAMF7 expression, and expression of cytolytic enzymes (perforin, granzyme A [GzmA], and GzmB) on cells isolated from SLE patients and healthy controls were analyzed by flow cytometry. CD107a expression and interferon-γ (IFNγ) production in response to viral antigenic stimulation in the presence or absence of an anti-SLAMF7 antibody were assessed by flow cytometry. Antiviral cytotoxic activity in response to SLAMF7 engagement was determined using a flow cytometry-based assay. RESULTS: The distribution of CD8+ T cell subsets was altered in the peripheral blood of SLE patients, with a decreased effector cell subpopulation. Memory CD8+ T cells from SLE patients displayed decreased amounts of SLAMF7, a surface receptor that characterizes effector CD8+ T cells. Ligation of SLAMF7 increased CD8+ T cell degranulation capacity and the percentage of IFNγ-producing cells in response to antigen challenge in SLE patients and healthy controls. Moreover, SLAMF7 engagement promoted cytotoxic lysis of target cells in response to stimulation with viral antigens. CONCLUSION: CD8+ T cell activation in response to viral antigens is defective in SLE patients. Activation of SLAMF7 through a specific monoclonal antibody restores CD8+ T cell antiviral effector function to normal levels and thus represents a potential therapeutic option in SLE.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/inmunología , Lupus Eritematoso Sistémico/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Subgrupos de Linfocitos T/inmunología , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Citometría de Flujo , Granzimas/metabolismo , Humanos , Activación de Linfocitos/inmunología , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Persona de Mediana Edad , Perforina/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Adulto Joven
7.
Arthritis Rheumatol ; 69(4): 808-813, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27992687

RESUMEN

OBJECTIVE: Imbalanced cytokine production by T cells characterizes both patients with systemic lupus erythematosus (SLE) and lupus-prone mice and contributes to immune dysregulation. This study was undertaken to further investigate in detail the production of interleukin-2 (IL-2), interferon-γ (IFNγ), IL-4, and IL-17A by CD4+ cell subsets in healthy subjects and patients with SLE, and the signaling response of CD4+ T cells in response to exogenous IL-2. METHODS: Cytokine production by differentiated subsets of CD4+ T cells was assessed by intracellular staining following stimulation with phorbol myristate acetate and ionomycin and by enzyme-linked immunosorbent assay after anti-CD3/anti-CD28 stimulation. The IL-2 signaling pathway was examined by assessing JAK-3/STAT-5 phosphorylation. Cell proliferation in response to IL-2 was examined by carboxyfluorescein succinimidyl ester dilution. RESULTS: Production of IL-2 was defective primarily among naive CD4+ T cells, whereas the production of IFNγ, IL-4, and IL-17A was not significantly different between patients with SLE and healthy subjects. JAK-3/STAT-5 phosphorylation and proliferation of CD4+ T cells from SLE patients in response to exogenous IL-2 were impaired compared to cells from healthy subjects. CONCLUSION: These data suggest that altered IL-2 production, as well as impaired IL-2-mediated signaling and proliferative responses, characterize SLE CD4+ T cells. Our data demonstrate the need for caution in designing IL-2 treatment trials for patients with SLE. Approaches to restore CD4+ T cell sensitivity to IL-2 should be considered.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Interleucina-2/biosíntesis , Interleucina-2/uso terapéutico , Interleucina-4/biosíntesis , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Células Cultivadas , Humanos
8.
Proc Natl Acad Sci U S A ; 113(33): 9321-6, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27482100

RESUMEN

Signaling lymphocytic activation molecule family 3 (SLAMF3/Ly9) is a coregulatory molecule implicated in T-cell activation and differentiation. Systemic lupus erythematosus (SLE) is characterized by aberrant T-cell activation and compromised IL-2 production, leading to abnormal regulatory T-cell (Treg) development/function. Here we show that SLAMF3 functions as a costimulator on CD4(+) T cells and influences IL-2 response and T helper cell differentiation. SLAMF3 ligation promotes T-cell responses to IL-2 via up-regulation of CD25 in a small mothers against decapentaplegic homolog 3 (Smad3)-dependent mechanism. This augments the activation of the IL-2/IL-2R/STAT5 pathway and enhances cell proliferation in response to exogenous IL-2. SLAMF3 costimulation promotes Treg differentiation from naïve CD4(+) T cells. Ligation of SLAMF3 receptors on SLE CD4(+) T cells restores IL-2 responses to levels comparable to those seen in healthy controls and promotes functional Treg generation. Taken together, our results suggest that SLAMF3 acts as potential therapeutic target in SLE patients by augmenting sensitivity to IL-2.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Interleucina-2/farmacología , Lupus Eritematoso Sistémico/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/fisiología , Linfocitos T Reguladores/fisiología , Adulto , Anciano , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Polaridad Celular , Femenino , Humanos , Interleucina-2/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/análisis , Subunidad alfa del Receptor de Interleucina-2/genética , Masculino , Persona de Mediana Edad
9.
J Immunol ; 196(12): 4915-24, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183584

RESUMEN

T cells from patients with systemic lupus erythematosus (SLE) display a number of abnormalities, including increased early signaling events following engagement of the TCR. Signaling lymphocytic activation molecule family cell surface receptors and the X-chromosome-defined signaling lymphocytic activation molecule-associated protein (SAP) adaptor are important in the development of several immunocyte lineages and modulating the immune response. We present evidence that SAP protein levels are decreased in T cells and in their main subsets isolated from 32 women and three men with SLE, independent of disease activity. In SLE T cells, SAP protein is also subject to increased degradation by caspase-3. Forced expression of SAP in SLE T cells normalized IL-2 production, calcium (Ca(2+)) responses, and tyrosine phosphorylation of a number of proteins. Exposure of normal T cells to SLE serum IgG, known to contain anti-CD3/TCR Abs, resulted in SAP downregulation. We conclude that SLE T cells display reduced levels of the adaptor protein SAP, probably as a result of continuous T cell activation and degradation by caspase-3. Restoration of SAP levels in SLE T cells corrects the overexcitable lupus T cell phenotype.


Asunto(s)
Interleucina-2/biosíntesis , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos , Transducción de Señal , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/metabolismo , Linfocitos T/inmunología , Adulto , Anciano , Calcio/metabolismo , Caspasa 3/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Inmunoglobulina G/inmunología , Interleucina-2/inmunología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/fisiopatología , Masculino , Persona de Mediana Edad , Fosforilación , Receptores de Antígenos de Linfocitos T/inmunología , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Linfocitos T/metabolismo , Tirosina/metabolismo , Adulto Joven
10.
Arthritis Rheumatol ; 68(1): 164-73, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26314831

RESUMEN

OBJECTIVE: Engagement of signaling lymphocytic activation molecule family member 4 (SLAMF4; CD244, 2B4) by its ligand SLAMF2 (CD48) modulates the function and expansion of both natural killer cells and a subset of cytotoxic CD8+ T cells. Because the cytotoxicity of CD8+ T lymphocytes isolated from patients with systemic lupus erythematosus (SLE) is known to be impaired, the aim of this study was to assess whether the expression and function of the checkpoint regulator SLAMF4 are altered on CD8+ T cells from patients with SLE. METHODS: The expression of SLAMF4 by T cells from healthy donors and patients with SLE was determined by quantitative polymerase chain reaction and flow cytometry. T cells were activated with anti-CD3 antibody, and degranulation activity was monitored by the surface expression of lysosome-associated membrane protein 1 (LAMP-1; CD107a). The SLAMF4+ and SLAMF4- CD8+ T cell subpopulations were characterized by LAMP-1, perforin, and granzyme B expression and viral peptide-induced proliferation. RESULTS: SLAMF4 gene and surface protein expression was down-regulated in CD8+ T cells from SLE patients compared with that in cells obtained from healthy donors. Importantly, SLE patients had significantly fewer SLAMF4+ CD8+ T cells compared with healthy donors. SLAMF4- CD8+ T cells from SLE patients had a decreased cytotoxic capacity and decreased proliferative responses to viral peptides. The loss of memory SLAMF4+ CD8+ T cells in SLE patients was linked to the fact that these cells have an increased propensity to lose CD8 expression and become double-negative T cells. CONCLUSION: A selective loss of SLAMF4+ CD8+ T cells contributes to the compromised ability of T cells from patients with SLE to fight infection.


Asunto(s)
Antígenos CD/inmunología , Lupus Eritematoso Sistémico/inmunología , ARN Mensajero/metabolismo , Receptores Inmunológicos/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos CD/genética , Western Blotting , Antígeno CD48 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Estudios de Casos y Controles , Proliferación Celular , Citometría de Flujo , Expresión Génica , Granzimas/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Lupus Eritematoso Sistémico/genética , Activación de Linfocitos , Perforina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Inmunológicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Linfocitos T Citotóxicos/metabolismo , Proteínas Virales
11.
Stem Cells Dev ; 24(15): 1805-16, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25808140

RESUMEN

The major reservoir of human multipotent mesenchymal stem/stromal cells (MSCs) is the bone marrow (BM) with the capability to control hematopoietic stem cell development. The regenerative potential of MSCs is associated with enhanced endogenous repair and healing mechanisms that modulate inflammatory responses. Our previous results revealed that MSC-like (MSCl) cells derived from pluripotent human embryonic stem cells resemble BM-derived MSCs in morphology, phenotype, and differentiating potential. In this study, we investigated the effects of MSCl cells on the phenotype and functions of dendritic cells (DCs). To assess how antiviral immune responses could be regulated by intracellular pattern recognition receptors of DCs in the presence of MSCl cells, we activated DCs with the specific ligands of retinoic acid-inducible gene-I (RIG-I) helicases and found that activated DCs cocultured with MSCl cells exhibited reduced expression of CD1a and CD83 cell surface molecules serving as phenotypic indicators of DC differentiation and activation, respectively. However, RIG-I-mediated stimulation of DCs through specific ligands in the presence of MSCl cells resulted in significantly higher expression of the costimulatory molecules, CD80 and CD86, than in the presence of BM-MSCs. In line with these results, the concentration of IL-6, IL-10, and CXCL8 was increased in the supernatant of the DC-MSCl cocultures, while the secretion of TNF-α, CXCL10, IL-12, and IFNγ was reduced. Furthermore, the concerted action of mechanisms involved in the regulation of DC migration resulted in the blockade of cell migration, indicating altered DC functionality mediated by MSCl cell-derived signals and mechanisms resulting in a suppressive microenvironment.


Asunto(s)
Comunicación Celular/inmunología , Células Dendríticas/inmunología , Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Transducción de Señal/inmunología , Antígenos CD/inmunología , Línea Celular , Citocinas/inmunología , Células Dendríticas/citología , Humanos , Células Madre Mesenquimatosas/citología , Monocitos/citología
13.
J Immunol ; 192(9): 4436-42, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24670806

RESUMEN

Signaling lymphocyte activation molecule family (SLAMF)2/CD48 is a coactivator and adhesion molecule on cells with hematopoietic origin. It ligates mainly SLAMF4 on effector/memory CD8(+) T cells and NK cells, suggesting a potential role during viral infection, with SLAMF2 acting as a ligand to activate SLAMF4-bearing cells. The ability of SLAMF2 to signal on its own after it is engaged and the functional consequences are largely unknown. We found that cytosolic DNA-activated dendritic cells (DCs) upregulate the expression of SLAMF2 molecules. Using anti-SLAMF2 Ab and SLAMF4 recombinant protein, we found that SLAMF2 engagement activates immature DCs and, more interestingly, prolongs the survival of DNA-activated DCs by inhibiting IFN-ß production and IFN-ß-induced apoptosis and promotes the production of the granzyme B inhibitor protease inhibitor-9. Thus, SLAMF2 can serve as a survival molecule for DNA-activated DCs during their interaction with SLAMF4-expressing cytotoxic T cells. Based on our results, we propose that SLAMF2 engagement regulates adaptive immune responses by providing longer access of putative APCs to virus-specific effector T cells by prolonging the time frame of effective stimulation.


Asunto(s)
Antígenos CD/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa/inmunología , Antígeno CD48 , Supervivencia Celular , Células Dendríticas/citología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
14.
Clin Immunol ; 150(2): 192-200, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24434273

RESUMEN

Systemic Lupus Erythematosus (SLE) remains a challenging disease to diagnose and follow, as no reliable biomarkers are known to date. We designed a gene expression panel with 40 genes known to play a role in SLE pathogenesis. We found that the combined expression of these genes in SLE T cells can accurately differentiate SLE from healthy individuals and patients with other autoimmune diseases. The accuracy of the test increased further (83%) when only three out of the initial genes (OAS2, CD70 and IL10) were used. A T cell score, calculated from the combined expression levels of these genes, correlated positively with various SLE activity markers in a cross-sectional cohort and in a few patients that were followed prospectively. These data showcase the usefulness of measuring mRNA levels of key molecules in diagnosing and following patients with SLE.


Asunto(s)
Regulación de la Expresión Génica , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , Adulto , Anciano , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Biomarcadores , Ligando CD27/genética , Estudios de Casos y Controles , Estudios Transversales , Femenino , Humanos , Interleucina-10/genética , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/diagnóstico , Masculino , Persona de Mediana Edad , Transcriptoma
15.
Proc Natl Acad Sci U S A ; 110(50): 20194-9, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24282294

RESUMEN

MicroRNA-155 (miR-155) regulates antibody responses and subsequent B-cell effector functions to exogenous antigens. However, the role of miR-155 in systemic autoimmunity is not known. Using the death receptor deficient (Fas(lpr)) lupus-prone mouse, we show here that ablation of miR-155 reduced autoantibody responses accompanied by a decrease in serum IgG but not IgM anti-dsDNA antibodies and a reduction of kidney inflammation. MiR-155 deletion in Fas(lpr) B cells restored the reduced SH2 domain-containing inositol 5'-phosphatase 1 to normal levels. In addition, coaggregation of the Fc γ receptor IIB with the B-cell receptor in miR-155(-/-)-Fas(lpr) B cells resulted in decreased ERK activation, proliferation, and production of switched antibodies compared with miR-155 sufficient Fas(lpr) B cells. Thus, by controlling the levels of SH2 domain-containing inositol 5'-phosphatase 1, miR-155 in part maintains an activation threshold that allows B cells to respond to antigens.


Asunto(s)
Autoanticuerpos/inmunología , Lupus Eritematoso Sistémico/prevención & control , MicroARNs/genética , MicroARNs/inmunología , Animales , Autoanticuerpos/biosíntesis , Northern Blotting , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Técnicas Histológicas , Inmunohistoquímica , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Noqueados , Urinálisis
16.
Proc Natl Acad Sci U S A ; 110(35): 14348-53, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23942120

RESUMEN

Inflammation and vascular injury triggered by ischemia/reperfusion (I/R) represent a leading cause of morbidity and mortality in a number of clinical settings. Wnt and its homolog partners R-spondins, in addition to regulating embryonic development have recently been demonstrated to serve as wound-healing agents in inflammation-associated conditions. Here we ask whether R-spondins could prevent inflammation-associated tissue damage in ischemic disorders and thus investigate the role of R-spondin3 (R-spo3) in a mouse model of mesenteric I/R. We demonstrate that R-spo3 ameliorates mesenteric I/R-induced local intestinal as well as remote lung damage by suppressing local and systemic cytokine response and deposition of IgM and complement in intestinal tissues. We also show that decreased inflammatory response is accompanied by tightening of endothelial cell junctions and reduction in vascular leakage. We conclude that R-spo3 stabilizes endothelial junctions and inhibits vascular leakage during I/R and thereby mitigates the inflammatory events and associated tissue damage. Our findings uniquely demonstrate a protective effect of R-spo3 in I/R-related tissue injury and suggest a mechanism by which it may have these effects.


Asunto(s)
Endotelio Vascular/metabolismo , Isquemia Mesentérica/metabolismo , Trombospondinas/metabolismo , Animales , Endotelio Vascular/patología , Uniones Intercelulares/efectos de los fármacos , Isquemia Mesentérica/tratamiento farmacológico , Ratones , Unión Proteica , Daño por Reperfusión/prevención & control , Trombospondinas/farmacología
17.
Immunobiology ; 218(11): 1361-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23870824

RESUMEN

Matrix metalloproteinases (MMPs) are endopeptidases with the potential to cleave extracellular matrix, support tissue renewal and regulate cell migration. Functional activities of MMPs are regulated by tissue inhibitors of MMPs (TIMPs) and disruption of the MMP-TIMP balance has pathological consequences. Here we studied the expression and secretion of MMPs and TIMPs in CD1a(-) and CD1a(+) monocyte-derived dendritic cell (DC) subpopulations. Our results showed that monocytes express TIMPs but lack MMPs, whereas upon differentiation to moDCs and in response to activation signals the expression of MMPs is increased and that of TIMPs is decreased. MMP-9 is expressed dominantly in the CD1a(-) subpopulation, while MMP-12 is preferentially expressed in CD1a(+) cells. Experiments performed with the synthetic MMP inhibitor GM6001 revealed that this drug efficiently inhibits the migration of moDCs through inactivation of MMPs. We conclude that modulation of MMP activity by GM6001 emerges as a novel approach to manipulate DC migration under inflammatory conditions.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Dendríticas/inmunología , Dipéptidos/farmacología , Inflamación/inmunología , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Antígenos CD1/metabolismo , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Humanos , Metaloproteinasa 12 de la Matriz/biosíntesis , Metaloproteinasa 12 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/metabolismo , Monocitos , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
18.
FASEB J ; 27(8): 3123-31, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23629864

RESUMEN

The costimulatory receptor Slamf6 partially controls lupus-related autoimmunity in congenic Sle1b mice; for instance, the presence of the protein isoform Slamf6-H1 in Sle1b.Slamf6-H1 mice mitigates disease. Here, we report that young Sle1b mice, but not Sle1b.Slamf6-H1 or B6 mice, contain a memory T-helper cell subset identified by ]mt]2-fold increase in expression of 17 genes, chief among which is Spp1, encoding the cytokine osteopontin (OPN). These T follicular helper (TFH) cells, including OPN(+) TFH cells, expand concomitantly with severity of the disease. By contrast, Sle1b.Slamf6-H1 or Sle1b.SAP(-)/(-) mice do not develop autoantibodies and the number of T(FH) cells is 5 times lower than in age-matched Sle1b mice. By comparing Sle1b and Sle1b.OPN(-)/(-) mice, we find that the lack of OPN expression impedes early autoantibody production. Furthermore, on the adoptive transfer of Sle1b.OPN(-)/(-) CD4(+) T cells into bm12 recipients autoantibody production and germinal center formation is reduced compared to recipients of Sle1b.OPN(+/+) CD4(+) T cells. We propose a model in which OPN provides a survival signal for a precursor T(FH) cell subset, which is a key factor in autoimmunity.


Asunto(s)
Antígenos CD/inmunología , Autoinmunidad/inmunología , Osteopontina/inmunología , Receptores de Superficie Celular/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Autoinmunidad/genética , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Femenino , Citometría de Flujo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Masculino , Ratones , Ratones Noqueados , Osteopontina/genética , Osteopontina/metabolismo , Receptor de Muerte Celular Programada 1 , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Receptores CXCR5/genética , Receptores CXCR5/inmunología , Receptores CXCR5/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Transcriptoma/inmunología
19.
Arthritis Rheum ; 65(7): 1882-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23575983

RESUMEN

OBJECTIVE: To identify microRNAs (miRNAs) in human T cells that can explain known antiinflammatory properties of steroids. METHODS: Activated human CD4+ T cells from healthy donors were exposed to 1 µM methylprednisolone (MP) in vitro and then subjected to miRNA and messenger RNA microarray analyses. Changes in expression profiles were recorded. Using quantitative polymerase chain reaction (qPCR), flow cytometry, and enzyme-linked immunosorbent assay (ELISA), we confirmed the suppression of predicted targets, and through miRNA transfection experiments, we could suggest mechanistic links. RESULTS: We identified numerous steroid-responsive genes and miRNAs-many known and some novel-including multiple previously unknown proinflammatory genes suppressed by MP. Further studies using qPCR, flow cytometry, and ELISA demonstrated that methylprednisolone increased the expression of miRNA-98 (miR-98) and suppressed the levels of predicted targets, including interleukin-13 and 3 tumor necrosis factor receptors (TNFRs): Fas, FasL, and TNFR superfamily member 1B. Forced expression of miR-98 in T cells resulted in suppression of the same targets. CONCLUSION: The findings of this study demonstrate a link between miR-98 expression and the effects of MP and provide evidence suggesting that MP acts through miR-98 to inhibit specific proinflammatory targets. Identification of this antiinflammatory mechanism of glucocorticoids is important, since it may pave the way toward the elusive goal of dissociating adverse effects from therapeutic effects.


Asunto(s)
Glucocorticoides/farmacología , Metilprednisolona/farmacología , MicroARNs/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , MicroARNs/metabolismo , Linfocitos T/metabolismo , Regulación hacia Arriba
20.
PLoS One ; 8(2): e55264, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23405128

RESUMEN

The aim of this study was to characterize and identify the mode of action of IC31®, a two-component vaccine adjuvant. We found that IC31® was accumulated in human peripheral blood monocytes, MHC class II positive cells and monocyte-derived DCs (moDCs) but not in plasmacytoid DCs (pDCs). In the presence of IC31® the differentiation of inflammatory CD1a(+) moDCs and the secretion of chemokines, TNF-α and IL-6 cytokines was inhibited but the production of IFNß was increased. Sustained addition of IC31® to differentiating moDCs interfered with IκBα phosphorylation, while the level of phospho-IRF3 increased. We also showed that both IC31® and its KLK component exhibited a booster effect on type I IFN responses induced by the specific ligands of TLR3 or TLR7/8, whereas TLR9 ligand induces type I IFN production only in the presence of IC31® or ODN1. Furthermore, long term incubation of moDCs with IC31® caused significantly higher expression of IRF and IFN genes than a single 24 hr treatment. The adjuvant activity of IC31® on the IFN response was shown to be exerted through TLRs residing in the vesicular compartment of moDCs. Based on these results IC31® was identified as a moDC modulatory adjuvant that sets the balance of the NF-κB and IRF3 mediated signaling pathways to the production of IFNß. Thus IC31® is emerging as a potent adjuvant to increase immune responses against intracellular pathogens and cancer in future vaccination strategies.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Endosomas/inmunología , Interferón Tipo I/biosíntesis , Leucocitos Mononucleares/efectos de los fármacos , Oligodesoxirribonucleótidos/farmacología , Oligopéptidos/farmacología , Receptores Toll-Like/inmunología , Adyuvantes Inmunológicos/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Quimiocinas/inmunología , Células Dendríticas/inmunología , Combinación de Medicamentos , Endosomas/efectos de los fármacos , Humanos , Proteínas I-kappa B/biosíntesis , Proteínas I-kappa B/inmunología , Factor 3 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Interferón beta/inmunología , Interleucina-6/inmunología , Leucocitos Mononucleares/inmunología , Ligadura , Inhibidor NF-kappaB alfa , FN-kappa B/inmunología , Oligodesoxirribonucleótidos/inmunología , Oligopéptidos/inmunología , Fosforilación , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...