Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Immunity ; 39(2): 259-71, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973222

RESUMEN

Stable Foxp3 expression is required for the development of functional regulatory T (Treg) cells. Here, we demonstrate that the expression of the transcription factor Foxp3 can be regulated through the polyubiquitination of multiple lysine residues, resulting in proteasome-mediated degradation. Expression of the deubiquitinase (DUB) USP7 was found to be upregulated and active in Treg cells, being associated with Foxp3 in the nucleus. Ectopic expression of USP7 decreased Foxp3 polyubiquitination and increased Foxp3 expression. Conversely, either treatment with DUB inhibitor or USP7 knockdown decreased endogenous Foxp3 protein expression and decreased Treg-cell-mediated suppression in vitro. Furthermore, in a murine adoptive-transfer-induced colitis model, either inhibition of DUB activity or USP7 knockdown in Treg cells abrogated their ability to resolve inflammation in vivo. Our data reveal a molecular mechanism in which rapid temporal control of Foxp3 expression in Treg cells can be regulated by USP7, thereby modulating Treg cell numbers and function.


Asunto(s)
Colitis/inmunología , Factores de Transcripción Forkhead/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Línea Celular , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Endopeptidasas/metabolismo , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño , Ubiquitina Tiolesterasa/genética , Peptidasa Específica de Ubiquitina 7 , Ubiquitinación
2.
Immunity ; 38(2): 275-84, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23333074

RESUMEN

Epidermal growth factor receptor (EGFR) is known to be critically involved in tissue development and homeostasis as well as in the pathogenesis of cancer. Here we showed that Foxp3(+) regulatory T (Treg) cells express EGFR under inflammatory conditions. Stimulation with the EGF-like growth factor Amphiregulin (AREG) markedly enhanced Treg cell function in vitro, and in a colitis and tumor vaccination model we showed that AREG was critical for efficient Treg cell function in vivo. In addition, mast cell-derived AREG fully restored optimal Treg cell function. These findings reveal EGFR as a component in the regulation of local immune responses and establish a link between mast cells and Treg cells. Targeting of this immune regulatory mechanism may contribute to the therapeutic successes of EGFR-targeting treatments in cancer patients.


Asunto(s)
Receptores ErbB/inmunología , Glicoproteínas/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Linfocitos T Reguladores/inmunología , Anfirregulina , Animales , Anticuerpos Neutralizantes/farmacología , Comunicación Celular/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Familia de Proteínas EGF , Receptores ErbB/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/genética , Glicoproteínas/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Activación de Linfocitos/efectos de los fármacos , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Mastocitos/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/inmunología , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
3.
Immunity ; 39(2): 298-310, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23954131

RESUMEN

Foxp3 is crucial for both the development and function of regulatory T (Treg) cells; however, the posttranslational mechanisms regulating Foxp3 transcriptional output remain poorly defined. Here, we demonstrate that T cell factor 1 (TCF1) and Foxp3 associates in Treg cells and that active Wnt signaling disrupts Foxp3 transcriptional activity. A global chromatin immunoprecipitation sequencing comparison in Treg cells revealed considerable overlap between Foxp3 and Wnt target genes. The activation of Wnt signaling reduced Treg-mediated suppression both in vitro and in vivo, whereas disruption of Wnt signaling in Treg cells enhanced their suppressive capacity. The activation of effector T cells increased Wnt3a production, and Wnt3a levels were found to be greatly increased in mononuclear cells isolated from synovial fluid versus peripheral blood of arthritis patients. We propose a model in which Wnt produced under inflammatory conditions represses Treg cell function, allowing a productive immune response, but, if uncontrolled, could lead to the development of autoimmunity.


Asunto(s)
Artritis/inmunología , Colitis/inmunología , Factores de Transcripción Forkhead/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Células HEK293 , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Líquido Sinovial/citología , Linfocitos T Reguladores/metabolismo , Vía de Señalización Wnt , Proteína Wnt3A/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
4.
Int J Mol Sci ; 21(18)2020 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-32899721

RESUMEN

The pathogenesis of many inflammatory diseases is associated with the uncontrolled activation of nuclear factor kappa B (NF-κB) in macrophages. Previous studies have shown that in various cell types, heat shock protein 70 (Hsp70) plays a crucial role in controlling NF-κB activity. So far, little is known about the role of Hsp70 in canine inflammatory processes. In this study we investigated the potential anti-inflammatory effects of Hsp70 in canine macrophages as well as the mechanisms underlying these effects. To this end, a canine macrophage cell line was stressed with arsenite, a chemical stressor, which upregulated Hsp70 expression as detected by flow cytometry and qPCR. A gene-edited version of this macrophage cell line lacking inducible Hsp70 was generated using CRISPR-Cas9 technology. To determine the effects of Hsp70 on macrophage inflammatory properties, arsenite-stressed wild-type and Hsp70 knockout macrophages were exposed to lipopolysaccharide (LPS), and the expression of the inflammatory cytokines IL-6, IL-1ß and tumor necrosis factor-α (TNF-α) and levels of phosphorylated NF-κB were determined by qPCR and Western Blotting, respectively. Our results show that non-toxic concentrations of arsenite induced Hsp70 expression in canine macrophages; Hsp70 upregulation significantly inhibited the LPS-induced expression of the pro-inflammatory mediators TNF-α and IL-6, as well as NF-κB activation in canine macrophages. Furthermore, the gene editing of inducible Hsp70 by CRISPR-Cas9-mediated gene editing neutralized this inhibitory effect of cell stress on NF-κB activation and pro-inflammatory cytokine expression. Collectively, our study reveals that Hsp70 may regulate inflammatory responses through NF-κB activation and cytokine expression in canine macrophages.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Inflamación/metabolismo , FN-kappa B/metabolismo , Animales , Arsenitos/farmacología , Línea Celular , Citocinas/metabolismo , Perros , Proteínas HSP70 de Choque Térmico/inmunología , Proteínas I-kappa B/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/inmunología , Óxido Nítrico/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
5.
Eur J Immunol ; 46(5): 1109-18, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26909514

RESUMEN

CD8(+) T cells responding to infection recognize pathogen-derived epitopes presented by MHC class-I molecules. While most of such epitopes are generated by proteasome-mediated antigen cleavage, analysis of tumor antigen processing has revealed that epitopes may also derive from proteasome-catalyzed peptide splicing (PCPS). To determine whether PCPS contributes to epitope processing during infection, we analyzed the fragments produced by purified proteasomes from a Listeria monocytogenes polypeptide. Mass spectrometry identified a known H-2K(b) -presented linear epitope (LLO296-304 ) in the digests, as well as four spliced peptides that were trimmed by ERAP into peptides with in silico predicted H-2K(b) binding affinity. These spliced peptides, which displayed sequence similarity with LLO296-304 , bound to H-2K(b) molecules in cellular assays and one of the peptides was recognized by CD8(+) T cells of infected mice. This spliced epitope differed by one amino acid from LLO296-304 and double staining with LLO296-304 - and spliced peptide-folded MHC multimers showed that LLO296-304 and its spliced variant were recognized by the same CD8(+) T cells. Thus, PCPS multiplies the variety of peptides that is processed from an antigen and leads to the production of epitope variants that can be recognized by cross-reacting pathogen-specific CD8(+) T cells. Such mechanism may reduce the chances for pathogen immune evasion.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Empalme de Proteína , Animales , Presentación de Antígeno/inmunología , Simulación por Computador , Epítopos de Linfocito T/química , Antígenos de Histocompatibilidad Clase I/inmunología , Evasión Inmune , Listeria monocytogenes/química , Espectrometría de Masas , Ratones , Péptidos/química , Péptidos/inmunología , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/química
6.
J Immunol ; 189(5): 2333-7, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22821961

RESUMEN

NK cells play an important role in the early defense against invading pathogens. Although it is well established that infection leads to a substantial, local increase in NK cell numbers, little is known about the mechanisms that trigger their proliferation and migration. In this study, we investigated the dynamics of NK cell responses after intranasal respiratory virus infection. We show that NK cell numbers increased in the airways after influenza virus infection but find no evidence of proliferation either at the site of infection or in the draining lymph nodes. Instead, we find that the bone marrow (BM) is the primary site of proliferation of both immature and mature NK cells during infection. Using an adoptive transfer model, we demonstrate that peripheral, long-lived and phenotypically mature NK cells migrate back to the BM and proliferate there, both homeostatically and in response to infection. Thus, the BM is not only a site of NK cell development but also an important site for proliferation of long-lived mature NK cells.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Proliferación Celular , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Traslado Adoptivo , Animales , Células de la Médula Ósea/patología , Supervivencia Celular/inmunología , Virus de la Influenza A/inmunología , Células Asesinas Naturales/patología , Trasplante de Hígado/inmunología , Trasplante de Hígado/patología , Trasplante de Pulmón/inmunología , Trasplante de Pulmón/patología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Bazo/citología , Bazo/inmunología , Bazo/trasplante
7.
Hum Vaccin Immunother ; 20(1): 2330768, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38517203

RESUMEN

Chlamydia trachomatis is an obligate intracellular pathogen responsible for the most prevalent bacterial sexually transmitted disease globally. The high prevalence of chlamydial infections underscores the urgent need for licensed and effective vaccines to prevent transmission in populations. Bacterial outer membrane vesicles (OMVs) have emerged as promising mucosal vaccine carriers due to their inherent adjuvant properties and the ability to display heterologous antigens. In this proof-of-concept study, we evaluated the immunogenicity of Salmonella OMVs decorated with C. trachomatis MOMP-derived CTH522 or HtrA antigens in mice. Following a prime-boost intranasal vaccination approach, two OMV-based C. trachomatis vaccines elicited significant humoral responses specific to the antigens in both systemic and vaginal compartments. Furthermore, we demonstrated strong antigen-specific IFN-γ and IL17a responses in splenocytes and cervical lymph node cells of vaccinated mice, indicating CD4+ Th1 and Th17 biased immune responses. Notably, the OMV-CTH522 vaccine also induced the production of spleen-derived CD8+ T cells expressing IFN-γ. In conclusion, these results highlight the potential of OMV-based C. trachomatis vaccines for successful use in future challenge studies and demonstrate the suitability of our modular OMV platform for intranasal vaccine applications.


Asunto(s)
Infecciones por Chlamydia , Vacunas , Femenino , Animales , Ratones , Chlamydia trachomatis , Linfocitos T CD8-positivos , Antígenos Bacterianos , Salmonella , Inmunidad , Vacunas Bacterianas , Infecciones por Chlamydia/prevención & control , Anticuerpos Antibacterianos , Proteínas de la Membrana Bacteriana Externa
8.
J Immunol ; 187(5): 2302-9, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21804012

RESUMEN

Exposure of cells to inflammatory cytokines induces the expression of three proteasome immunosubunits, two of which are encoded in the MHC class II region. The induced subunits replace their constitutive homologs in newly formed "so-called" immunoproteasomes. Immunosubunit incorporation enhances the proteasome's proteolytic activity and modifies the proteasome's cleavage-site preferences, which improves the generation of many MHC class I-presented peptides and shapes the fine specificity of pathogen-specific CD8 T cell responses. In this article, we report on a second effect of immunoproteasome formation on CD8 T cell responses. We show that mice deficient for the immunosubunits ß5i/low molecular mass polypeptide (LMP7) and ß2i/multicatalytic endopeptidase complex-like-1 develop early-stage multiorgan autoimmunity following irradiation and bone marrow transplantation. Disease symptoms are caused by CD8 T cells and are transferable into immunosubunit-deficient, RAG1-deficient mice. Moreover, using the human Type 1 Diabetes Genetics Consortium MHC dataset, we identified two single nucleotide polymorphisms within the ß5i/LMP7-encoding gene sequences, which were in strong linkage disequilibrium, as independent genetic risk factors for type 1 diabetes development in humans. Strikingly, these single nucleotide polymorphisms significantly enhanced the risk conferred by HLA haplotypes that were previously shown to predispose for type 1 diabetes. These data suggested that inflammation-induced immunosubunit expression in peripheral tissues constitutes a mechanism that prevents the development of CD8 T cell-mediated autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Linfocitos T CD8-positivos/inmunología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Predisposición Genética a la Enfermedad , Humanos , Desequilibrio de Ligamiento , Ratones , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Factores de Riesgo
9.
Cancer Res Commun ; 3(1): 109-118, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36968226

RESUMEN

In the last decades, antibody-based tumor therapy has fundamentally improved the efficacy of treatment for patients with cancer. Currently, almost all tumor antigen-targeting antibodies approved for clinical application are of IgG1 Fc isotype. Similarly, the mouse homolog mIgG2a is the most commonly used in tumor mouse models. However, in mice, the efficacy of antibody-based tumor therapy is largely restricted to a prophylactic application. Direct isotype comparison studies in mice in a therapeutic setting are scarce. In this study, we assessed the efficacy of mouse tumor-targeting antibodies of different isotypes in a therapeutic setting using a highly systematic approach. To this end, we engineered and expressed antibodies of the same specificity but different isotypes, targeting the artificial tumor antigen CD90.1/Thy1.1 expressed by B16 melanoma cells. Our experiments revealed that in a therapeutic setting mIgG2a was superior to both mIgE and mIgG1 in controlling tumor growth. Furthermore, the observed mIgG2a antitumor effect was entirely Fc mediated as the protection was lost when an Fc-silenced mIgG2a isotype (LALA-PG mutations) was used. These data confirm mIgG2a superiority in a therapeutic tumor model. Significance: Direct comparisons of different antibody isotypes of the same specificity in cancer settings are still scarce. Here, it is shown that mIgG2a has a greater effect compared with mIgG1 and mIgE in controlling tumor growth in a therapeutic setting.


Asunto(s)
Inmunoglobulina G , Neoplasias , Animales , Ratones , Receptores Fc , Neoplasias/terapia , Antígenos de Neoplasias
10.
PLoS One ; 18(11): e0292757, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37939066

RESUMEN

Macrophages can reversibly polarize into multiple functional subsets depending on their micro-environment. Identification and understanding the functionality of these subsets is relevant for the study of immune­related diseases. However, knowledge about canine macrophage polarization is still in its infancy. In this study, we polarized canine monocytes using GM-CSF/IFN- γ and LPS towards M1 macrophages or M-CSF and IL-4 towards M2 macrophages and compared them to undifferentiated monocytes (M0). Polarized M1 and M2 macrophages were thoroughly characterized for morphology, surface marker features, gene profiles and functional properties. Our results showed that canine M1-polarized macrophages obtained a characteristic large, roundish, or amoeboid shape, while M2-polarized macrophages were smaller and adopted an elongated spindle-like morphology. Phenotypically, all macrophage subsets expressed the pan-macrophage markers CD14 and CD11b. M1-polarized macrophages expressed increased levels of CD40, CD80 CD86 and MHC II, while a significant increase in the expression levels of CD206, CD209, and CD163 was observed in M2-polarized macrophages. RNAseq of the three macrophage subsets showed distinct gene expression profiles, which are closely associated with immune responsiveness, cell differentiation and phagocytosis. However, the complexity of the gene expression patterns makes it difficult to assign clear new polarization markers. Functionally, undifferentiated -monocytes, and M1- and M2- like subsets of canine macrophages can all phagocytose latex beads. M2-polarized macrophages exhibited the strongest phagocytic capacity compared to undifferentiated monocytes- and M1-polarized cells. Taken together, this study showed that canine M1 and M2-like macrophages have distinct features largely in parallel to those of well-studied species, such as human, mouse and pig. These findings enable future use of monocyte derived polarized macrophages particularly in studies of immune related diseases in dogs.


Asunto(s)
Macrófagos , Monocitos , Animales , Perros , Diferenciación Celular , Macrófagos/metabolismo , Monocitos/metabolismo , Fagocitosis
11.
Eur J Immunol ; 41(4): 926-35, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21360704

RESUMEN

Proteasomes play a fundamental role in the processing of intracellular antigens into peptides that bind to MHC class I molecules for the presentation of CD8(+) T cells. Three IFN-γ-inducible catalytic proteasome (immuno)subunits as well as the IFN-γ-inducible proteasome activator PA28 dramatically accelerate the generation of a subset of MHC class I-presented antigenic peptides. To determine whether these IFN-γ-inducible proteasome components play a compounded role in antigen processing, we generated mice lacking both PA28 and immunosubunits ß5i/LMP7 and ß2i/MECL-1. Analyses of MHC class I cell-surface levels ex vivo demonstrated that PA28 deficiency reduced the production of MHC class I-binding peptides both in cells with and without immunosubunits, in the latter cells further decreasing an already diminished production of MHC ligands in the absence of immunoproteasomes. In contrast, the immunosubunits but not PA28 appeared to be of critical importance for the induction of CD8(+) T-cell responses to multiple dominant Influenza and Listeria-derived epitopes. Taken together, our data demonstrate that PA28 and the proteasome immunosubunits use fundamentally different mechanisms to enhance the supply of MHC class I-binding peptides; however, only the immunosubunit-imposed effects on proteolytic epitope processing appear to have substantial influence on the specificity of pathogen-specific CD8(+) T-cell responses.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Péptidos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Péptidos/química , Complejo de la Endopetidasa Proteasomal/deficiencia
12.
Biomolecules ; 12(10)2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36291540

RESUMEN

T cell engager (TCE) antibodies have emerged as promising cancer therapeutics that link cytotoxic T-cells to tumor cells by simultaneously binding to CD3E on T-cells and to a tumor-associated antigen (TAA) expressed by tumor cells. We previously reported a novel bispecific format, the IgG-like Fab x sdAb-Fc (also known as half-IG_VH-h-CH2-CH3), combining a conventional antigen-binding fragment (Fab) with a single domain antibody (sdAb). Here, we evaluated this Fab x sdAb-Fc format as a T-cell redirecting bispecific antibody (TbsAbs) by targeting mEGFR on tumor cells and mCD3E on T cells. We focused our attention specifically on the hinge design of the sdAb arm of the bispecific antibody. Our data show that a TbsAb with a shorter hinge of 23 amino acids (TbsAb.short) showed a significantly better T cell redirected tumor cell elimination than the TbsAb with a longer, classical antibody hinge of 39 amino acids (TbsAb.long). Moreover, the TbsAb.short form mediated better T cell-tumor cell aggregation and increased CD69 and CD25 expression levels on T cells more than the TbsAb.long form. Taken together, our results indicate that already minor changes in the hinge design of TbsAbs can have significant impact on the anti-tumor activity of TbsAbs and may provide a new means to improve their potency.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Anticuerpos de Dominio Único , Humanos , Anticuerpos Biespecíficos/química , Neoplasias/terapia , Inmunoglobulina G , Aminoácidos , Muerte Celular
13.
Cells ; 10(12)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34943869

RESUMEN

Proteasomes are responsible for intracellular proteolysis and play an important role in cellular protein homeostasis. Cells of the immune system assemble a specialized form of proteasomes, known as immunoproteasomes, in which the constitutive catalytic sites are replaced for cytokine-inducible homologues. While immunoproteasomes may fulfill all standard proteasome' functions, they seem specially adapted for a role in MHC class I antigen processing and CD8+ T-cell activation. In this way, they may contribute to CD8+ T-cell-mediated control of intracellular infections, but also to the immunopathogenesis of autoimmune diseases. Starting at the discovery of its catalytic subunits in the genome, here, we review the observations shaping our current understanding of immunoproteasome function, and the consequential novel opportunities for immune intervention.


Asunto(s)
Alergia e Inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Animales , Presentación de Antígeno/inmunología , Humanos , Inmunidad , Inflamación/inmunología , Inflamación/patología , Linfocitos T/inmunología
14.
J Immunol Methods ; 499: 113173, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34699840

RESUMEN

Tumor necrosis factor receptor 2 (TNFR2) has gained much research interest in recent years because of its potential pivotal role in autoimmune disease and cancer. However, its function in regulating different immune cells is not well understood. There is a need for well-characterized reagents to selectively modulate TNFR2 function, thereby enabling definition of TNFR2-dependent biology in human and mouse surrogate models. Here, we describe the generation, production, purification, and characterization of a panel of novel antibodies targeting mouse TNFR2. The antibodies display functional differences in binding affinity and potency to block TNFα. Furthermore, epitope binding showed that the anti-mTNFR2 antibodies target different domains on the TNFR2 protein, associated with varying capacity to enhance CD8+ T-cell activation and costimulation. Moreover, the anti-TNFR2 antibodies demonstrate binding to isolated splenic mouse Tregs ex vivo and activated CD8+ cells, reinforcing their potential use to establish TNFR2-dependent immune modulation in translational models of autoimmunity and cancer.


Asunto(s)
Anticuerpos/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Animales , Células CHO , Cricetulus , Femenino , Ratones , Ratas , Ratas Sprague-Dawley
15.
J Extracell Vesicles ; 10(5): e12071, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33732416

RESUMEN

Maternal milk is nature's first functional food. It plays a crucial role in the development of the infant's gastrointestinal (GI) tract and the immune system. Extracellular vesicles (EVs) are a heterogeneous population of lipid bilayer enclosed vesicles released by cells for intercellular communication and are a component of milk. Recently, we discovered that human milk EVs contain a unique proteome compared to other milk components. Here, we show that physiological concentrations of milk EVs support epithelial barrier function by increasing cell migration via the p38 MAPK pathway. Additionally, milk EVs inhibit agonist-induced activation of endosomal Toll like receptors TLR3 and TLR9. Furthermore, milk EVs directly inhibit activation of CD4+ T cells by temporarily suppressing T cell activation without inducing tolerance. We show that milk EV proteins target key hotspots of signalling networks that can modulate cellular processes in various cell types of the GI tract.


Asunto(s)
Vesículas Extracelulares/metabolismo , Sistema de Señalización de MAP Quinasas , Leche Humana/citología , Mucosa Bucal/fisiología , Adulto , Línea Celular , Vesículas Extracelulares/inmunología , Femenino , Humanos , Mucosa Bucal/inmunología , Linfocitos T/inmunología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 9/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Cancer Res Clin Oncol ; 146(12): 3111-3122, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32989604

RESUMEN

PURPOSE: Bispecific antibodies (BsAbs) have emerged as a leading drug class for cancer therapy and are becoming increasingly of interest for therapeutic applications. As of April 2020, over 123 BsAbs are under clinical evaluation for use in oncology (including the two marketed BsAbs Blinatumomab and Catumaxomab). The majority (82 of 123) of BsAbs under clinical evaluation can be categorized as bispecific immune cell engager whereas a second less well-discussed subclass of BsAbs targets two tumor-associated antigens (TAAs). In this review, we summarize the clinical development of dual TAAs targeting BsAbs and provide an overview of critical considerations when designing dual TAA targeting BsAbs. METHODS: Herein the relevant literature and clinical trials published in English until April 1st 2020 were searched using PubMed and ClinicalTrials.gov database. BsAbs were considered to be active in clinic if their clinical trials were not terminated, withdrawn or completed before 2018 without reporting results. Data missed by searching ClinicalTrials.gov was manually curated. RESULTS: Dual TAAs targeting BsAbs offer several advantages including increased tumor selectivity, potential to concurrently modulate two functional pathways in the tumor cell and may yield improved payload delivery. CONCLUSIONS: Dual TAAs targeting BsAbs represent a valuable class of biologics and early stage clinical studies have demonstrated promising anti-tumor efficacy in both hematologic malignancies and solid tumors.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antígenos de Neoplasias/inmunología , Neoplasias/terapia , Anticuerpos Biespecíficos/inmunología , Antígenos de Neoplasias/efectos de los fármacos , Humanos , Neoplasias/inmunología , Neoplasias/patología
17.
J Immunol Methods ; 483: 112811, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32569598

RESUMEN

Due to the technical innovations in generating bispecific antibodies (BsAbs) in recent years, BsAbs have become important reagents for diagnostic and therapeutic applications. However, the difficulty of producing a heterodimer consisting of two different arms with high yield and purity constituted a major limitation for their application in academic and clinical settings. Here, we describe a novel Fc-containing BsAb format (Fab × sdAb-Fc) composed of a conventional antigen-binding fragment (Fab), and a single domain antibody (sdAb), which avoids heavy-light chain mis-pairing during antibody assembly. In this study, the Fab x sdAb-Fc BsAbs were efficiently produced by three widely used heavy-heavy chain heterodimerization methods: Knobs-into-holes (KIH), Charge-pairs (CP) and controlled Fab-arm exchange (cFAE), respectively. The novel Fab x sdAb-Fc format provided a rapid and efficient strategy to generate BsAb with high purity and a unique possibility to further purify desired BsAbs from undesired antibodies based on molecular weight (MW). Compared to conventional BsAb formats, the advantages of Fab x sdAb-Fc format may thus provide a straightforward opportunity to apply bispecific antibody principles to research and development of novel targets and pathways in diseases such as cancer and autoimmunity.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Receptores ErbB/inmunología , Glutamato Carboxipeptidasa II/inmunología , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Glicoproteínas de Membrana/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Anticuerpos Biespecíficos/biosíntesis , Anticuerpos Biespecíficos/genética , Especificidad de Anticuerpos , Células CHO , Cricetulus , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glutamato Carboxipeptidasa II/genética , Glutamato Carboxipeptidasa II/metabolismo , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/biosíntesis , Fragmentos Fc de Inmunoglobulinas/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Peso Molecular , Mutación , Prueba de Estudio Conceptual , Multimerización de Proteína , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/genética
18.
Cell Stress Chaperones ; 25(2): 235-243, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31940135

RESUMEN

Dysregulation of retinal pigment epithelium (RPE) cells is the main cause of a variety of ocular diseases. Potentially heat shock proteins, by preventing molecular and cellular damage and modulating inflammatory disease, may exert a protective role in eye disease. In particular, the inducible form of heat shock protein 70 (Hsp70) is widely upregulated in inflamed tissues, and in vivo upregulation of Hsp70 expression by HSP co-inducing compounds has been shown to be a potential therapeutic strategy for inflammatory diseases. In order to gain further understanding of the potential protective effects of Hsp70 in RPE cells, we developed a method for isolation and culture of canine RPE cells. Identity of RPE cells was confirmed by detection of its specific marker, RPE65, in qPCR, flow cytometry, and immunocytochemistry analysis. The ability of RPE cells to express Hsp70 upon experimental induction of cell stress, by arsenite, was analyzed by flow cytometry. Finally, in search of a potential Hsp70 co-inducer, we investigated whether the compound leucinostatin could enhance Hsp70 expression in stressed RPE cells. Canine RPE cells were isolated and cultured successfully. Purity of cells that strongly expressed RPE65 was over 90%. Arsenite-induced stress led to a time- and dose-dependent increase in Hsp70 expression in canine RPE cells in vitro. In addition, leucinostatin, which enhanced heat shock factor-1-induced transcription from the heat shock promoter in DNAJB1-luc-O23 reporter cell line, also enhanced Hsp70 expression in arsenite-stressed RPE cells, in a dose-dependent fashion. These findings demonstrate that leucinostatin can boost Hsp70 expression in canine RPE cells, most likely by activating heat shock factor-1, suggesting that leucinostatin might be applied as a new co-inducer for Hsp70 expression.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Células Epiteliales/citología , Proteínas HSP70 de Choque Térmico/metabolismo , Epitelio Pigmentado de la Retina/citología , Estrés Fisiológico/efectos de los fármacos , Animales , Células Cultivadas , Perros
19.
Infect Immun ; 76(3): 1207-13, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18160473

RESUMEN

Homeostatic regulatory mechanisms maintain the constant ratios between different lymphocyte subsets in the secondary lymphoid organs. How this dynamic equilibrium is achieved, in particular following the clonal expansion and subsequent contraction of different cells after infection, remains poorly understood. Expression of the proteasome immunosubunits has been shown to influence not only major histocompatibility complex class I (MHC-I) antigen processing and thereby T-cell responses, but also the CD4/CD8 T-cell ratios in lymphoid organs. We examined the relationships between these different immunosubunit-mediated effects in mice of various proteasome subunit compositions during infection with Listeria monocytogenes. Mice that lacked the immunosubunit multicatalytic endopeptidase complex-like 1 (MECL-1) maintained enhanced CD4/CD8 T-cell ratios during infection, while MHC-I surface levels resembled those in wild-type (wt) mice. LMP7 gene-deficient mice, on the other hand, showed reduced MHC-I expression, while their splenic CD4/CD8 ratios were similar to those in wt mice. Remarkably, analysis of bone marrow-chimeric immunosubunit gene-deficient mice, reconstituted with a mixture of wt and LMP7- plus MECL-1-deficient bone marrow, revealed that the LMP7- plus MECL-1-deficient T-cell population maintained a higher CD4/CD8 T-cell ratio than the wt T-cell population before, during, and after infection and T-cell memory formation. Since in these mice the immunosubunit-positive and immunosubunit-negative T-cell populations were selected in the same thymus and expanded in the same lymphoid environments, our findings indicate that MECL-1 influences the homeostatic equilibrium between T-cell subsets, not through indirect extracellular signals, such as MHC-I expression or the cytokine milieu, but through direct effects on T-cell-intrinsic processes.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Listeria monocytogenes/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Relación CD4-CD8 , Cisteína Endopeptidasas/deficiencia , Femenino , Antígenos de Histocompatibilidad Clase I/análisis , Interferón gamma/análisis , Depleción Linfocítica , Ratones , Ratones Noqueados , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Bazo/citología , Bazo/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/química
20.
Infect Immun ; 76(10): 4609-14, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18678661

RESUMEN

Cytotoxicity is a key effector function of CD8 T cells. However, what proportion of antigen-specific CD8 T cells in vivo exert cytotoxic activity during a functional CD8 T-cell response to infection still remains unknown. We used the Lysispot assay to directly enumerate cytotoxic CD8 T cells from the spleen ex vivo during the immune response to infection with the intracellular bacterium Listeria monocytogenes. We demonstrate that not all antigen-responsive gamma interferon (IFN-gamma)-secreting T cells display cytotoxic activity. Most CD8 T cells detected at early time points of the response were cytotoxic. This percentage continuously declined during both the expansion and contraction phases to about 50% at the peak and to <10% of IFN-gamma-producing cells in the memory phase. As described for clonal expansion, this elaboration of a program of differentiation after an initial stimulus was not affected by antigen or CD4 help but, like proliferation, could be influenced by later reinfection. These data indicate that cytotoxic effector function during the response to infection is regulated independently from IFN-gamma secretion or expansion or contraction of the overall CD8 T-cell response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Listeria monocytogenes/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Pruebas Inmunológicas de Citotoxicidad , Citotoxicidad Inmunológica , Femenino , Interferón gamma/metabolismo , Recuento de Linfocitos , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA