Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(10): 1495-1506, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36151395

RESUMEN

The immune system can eliminate tumors, but checkpoints enable immune escape. Here, we identify immune evasion mechanisms using genome-scale in vivo CRISPR screens across cancer models treated with immune checkpoint blockade (ICB). We identify immune evasion genes and important immune inhibitory checkpoints conserved across cancers, including the non-classical major histocompatibility complex class I (MHC class I) molecule Qa-1b/HLA-E. Surprisingly, loss of tumor interferon-γ (IFNγ) signaling sensitizes many models to immunity. The immune inhibitory effects of tumor IFN sensing are mediated through two mechanisms. First, tumor upregulation of classical MHC class I inhibits natural killer cells. Second, IFN-induced expression of Qa-1b inhibits CD8+ T cells via the NKG2A/CD94 receptor, which is induced by ICB. Finally, we show that strong IFN signatures are associated with poor response to ICB in individuals with renal cell carcinoma or melanoma. This study reveals that IFN-mediated upregulation of classical and non-classical MHC class I inhibitory checkpoints can facilitate immune escape.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico , Evasión Inmune , Interferón gamma/genética , Interferón gamma/metabolismo , Subfamília C de Receptores Similares a Lectina de Células NK
2.
Immunity ; 54(3): 571-585.e6, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33497609

RESUMEN

CRISPR-Cas9 genome engineering has increased the pace of discovery for immunology and cancer biology, revealing potential therapeutic targets and providing insight into mechanisms underlying resistance to immunotherapy. However, endogenous immune recognition of Cas9 has limited the applicability of CRISPR technologies in vivo. Here, we characterized immune responses against Cas9 and other expressed CRISPR vector components that cause antigen-specific tumor rejection in several mouse cancer models. To avoid unwanted immune recognition, we designed a lentiviral vector system that allowed selective CRISPR antigen removal (SCAR) from tumor cells. The SCAR system reversed immune-mediated rejection of CRISPR-modified tumor cells in vivo and enabled high-throughput genetic screens in previously intractable models. A pooled in vivo screen using SCAR in a CRISPR-antigen-sensitive renal cell carcinoma revealed resistance pathways associated with autophagy and major histocompatibility complex class I (MHC class I) expression. Thus, SCAR presents a resource that enables CRISPR-based studies of tumor-immune interactions and prevents unwanted immune recognition of genetically engineered cells, with implications for clinical applications.


Asunto(s)
Carcinoma de Células Renales/inmunología , Pruebas Genéticas/métodos , Vectores Genéticos/genética , Inmunoterapia/métodos , Neoplasias Renales/inmunología , Células Asesinas Naturales/inmunología , Lentivirus/genética , Animales , Presentación de Antígeno , Autofagia , Carcinoma de Células Renales/terapia , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Ingeniería Genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Neoplasias Renales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Molecular Dirigida
3.
Immunity ; 45(6): 1205-1218, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28002729

RESUMEN

Inflammation triggers the differentiation of Ly6Chi monocytes into microbicidal macrophages or monocyte-derived dendritic cells (moDCs). Yet, it is unclear whether environmental inflammatory cues control the polarization of monocytes toward each of these fates or whether specialized monocyte progenitor subsets exist before inflammation. Here, we have shown that naive monocytes are phenotypically heterogeneous and contain an NR4A1- and Flt3L-independent, CCR2-dependent, Flt3+CD11c-MHCII+PU.1hi subset. This subset acted as a precursor for FcγRIII+PD-L2+CD209a+, GM-CSF-dependent moDCs but was distal from the DC lineage, as shown by fate-mapping experiments using Zbtb46. By contrast, Flt3-CD11c-MHCII-PU.1lo monocytes differentiated into FcγRIII+PD-L2-CD209a-iNOS+ macrophages upon microbial stimulation. Importantly, Sfpi1 haploinsufficiency genetically distinguished the precursor activities of monocytes toward moDCs or microbicidal macrophages. Indeed, Sfpi1+/- mice had reduced Flt3+CD11c-MHCII+ monocytes and GM-CSF-dependent FcγRIII+PD-L2+CD209a+ moDCs but generated iNOS+ macrophages more efficiently. Therefore, intercellular disparities of PU.1 expression within naive monocytes segregate progenitor activity for inflammatory iNOS+ macrophages or moDCs.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Traslado Adoptivo , Animales , Antígenos Ly/inmunología , Separación Celular , Células Dendríticas/citología , Citometría de Flujo , Macrófagos/citología , Ratones , Monocitos/citología , Óxido Nítrico Sintasa de Tipo II/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
4.
Nature ; 565(7737): 43-48, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559380

RESUMEN

Most patients with cancer either do not respond to immune checkpoint blockade or develop resistance to it, often because of acquired mutations that impair antigen presentation. Here we show that loss of function of the RNA-editing enzyme ADAR1 in tumour cells profoundly sensitizes tumours to immunotherapy and overcomes resistance to checkpoint blockade. In the absence of ADAR1, A-to-I editing of interferon-inducible RNA species is reduced, leading to double-stranded RNA ligand sensing by PKR and MDA5; this results in growth inhibition and tumour inflammation, respectively. Loss of ADAR1 overcomes resistance to PD-1 checkpoint blockade caused by inactivation of antigen presentation by tumour cells. Thus, effective anti-tumour immunity is constrained by inhibitory checkpoints such as ADAR1 that limit the sensing of innate ligands. The induction of sufficient inflammation in tumours that are sensitized to interferon can bypass the therapeutic requirement for CD8+ T cell recognition of cancer cells and may provide a general strategy to overcome immunotherapy resistance.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/genética , Animales , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia , Inflamación/genética , Inflamación/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferones/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Fenotipo , Edición de ARN , ARN Bicatenario/genética , Proteínas de Unión al ARN/genética , Receptores Acoplados a Proteínas G/metabolismo
5.
PLoS Pathog ; 11(10): e1005211, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26473595

RESUMEN

Toxoplasma gondii possesses sets of dense granule proteins (GRAs) that either assemble at, or cross the parasitophorous vacuole membrane (PVM) and exhibit motifs resembling the HT/PEXEL previously identified in a repertoire of exported Plasmodium proteins. Within Plasmodium spp., cleavage of the HT/PEXEL motif by the endoplasmic reticulum-resident protease Plasmepsin V precedes trafficking to and export across the PVM of proteins involved in pathogenicity and host cell remodelling. Here, we have functionally characterized the T. gondii aspartyl protease 5 (ASP5), a Golgi-resident protease that is phylogenetically related to Plasmepsin V. We show that deletion of ASP5 causes a significant loss in parasite fitness in vitro and an altered virulence in vivo. Furthermore, we reveal that ASP5 is necessary for the cleavage of GRA16, GRA19 and GRA20 at the PEXEL-like motif. In the absence of ASP5, the intravacuolar nanotubular network disappears and several GRAs fail to localize to the PVM, while GRA16 and GRA24, both known to be targeted to the host cell nucleus, are retained within the vacuolar space. Additionally, hypermigration of dendritic cells and bradyzoite cyst wall formation are impaired, critically impacting on parasite dissemination and persistence. Overall, the absence of ASP5 dramatically compromises the parasite's ability to modulate host signalling pathways and immune responses.


Asunto(s)
Proteasas de Ácido Aspártico/metabolismo , Aparato de Golgi/enzimología , Interacciones Huésped-Parásitos/fisiología , Toxoplasma/patogenicidad , Toxoplasmosis/enzimología , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Transporte de Proteínas , Reacción en Cadena en Tiempo Real de la Polimerasa , Toxoplasma/enzimología , Transfección
6.
J Autoimmun ; 67: 8-18, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26341385

RESUMEN

Plasmacytoid dendritic cells (pDCs) exhibit both innate and adaptive functions. In particular they are the main source of type I IFNs and directly impact T cell responses through antigen presentation. We have previously demonstrated that during experimental autoimmune encephalomyelitis (EAE) initiation, myelin-antigen presentation by pDCs is associated with suppressive Treg development and results in attenuated EAE. Here, we show that pDCs transferred during acute disease phase confer recovery from EAE. Clinical improvement is associated with migration of injected pDCs into inflamed CNS and is dependent on the subsequent and selective chemerin-mediated recruitment of endogenous pDCs to the CNS. The protective effect requires pDC pre-loading with myelin antigen, and is associated with the modulation of CNS-infiltrating pDC phenotype and inhibition of CNS encephalitogenic T cells. This study may pave the way for novel pDC-based cell therapies in autoimmune diseases, aiming at specifically modulating pathogenic cells that induce and sustain autoimmune inflammation.


Asunto(s)
Traslado Adoptivo , Quimiotaxis/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Animales , Autoantígenos/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Noqueados , Vaina de Mielina/inmunología , Receptores de Quimiocina , Receptores Acoplados a Proteínas G/metabolismo , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología
7.
J Autoimmun ; 75: 39-49, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27470005

RESUMEN

Plasmacytoid dendritic cells (pDCs) have been shown to both mediate and prevent autoimmunity, and the regulation of their immunogenic versus tolerogenic functions remains incompletely understood. Here we demonstrate that, compared to other cells, pDCs are the major expressors of Indoleamine-2,3-dioxygenase (IDO) in steady-state lymph nodes (LNs). IDO expression by LN pDCs was closely dependent on MHCII-mediated, antigen-dependent, interactions with Treg. We further established that IDO production by pDCs was necessary to confer suppressive function to Tregs. During EAE development, IDO expression by pDCs was required for the generation of Tregs capable of dampening the priming of encephalitogenic T cell and disease severity. Thus, we describe a novel crosstalk between pDCs and Tregs: Tregs shape tolerogenic functions of pDCs prior to inflammation, such that pDCs in turn, promote Treg suppressive functions during autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Células Dendríticas/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad/genética , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Citometría de Flujo , Regulación Enzimológica de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ganglios Linfáticos/enzimología , Ganglios Linfáticos/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Reguladores/metabolismo
8.
Mol Ther ; 20(9): 1664-75, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22735380

RESUMEN

Intratumoral injection of Semliki Forest virus encoding interleukin-12 (SFV-IL-12) combines acute expression of IL-12 and stressful apoptosis of infected malignant cells. Agonist antibodies directed to costimulatory receptor CD137 (4-1BB) strongly amplify pre-existing cellular immune responses toward weak tumor antigens. In this study, we provide evidence for powerful synergistic effects of a combined strategy consisting of intratumoral injection of SFV-IL-12 and systemic delivery of agonist anti-CD137 monoclonal antibodies (mAbs), which was substantiated against poorly immunogenic B16 melanomas (B16-OVA and B16.F10) and TC-1 lung carcinomas. Effector CD8(ß)(+) T cells were sufficient to mediate complete tumor eradications. Accordingly, there was an intensely synergistic in vivo enhancement of cytotoxic T lymphocytes (CTL)-mediated immunity against the tumor antigens OVA and tyrosine-related protein-2 (TRP-2). This train of phenomena led to long-lasting tumor-specific immunity against rechallenge, attained transient control of the progression of concomitant tumor lesions that were not directly treated with SFV-IL-12 and caused autoimmune vitiligo. Importantly, we found that SFV-IL-12 intratumoral injection induces bright expression of CD137 on most tumor-infiltrating CD8(+) T lymphocytes, thereby providing more abundant targets for the action of the agonist antibody. This efficacious combinatorial immunotherapy strategy offers feasibility for clinical translation since anti-CD137 mAbs are already undergoing clinical trials and development of clinical-grade SFV-IL-12 vectors is in progress.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Carcinoma/terapia , Interleucina-12/inmunología , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Virus de los Bosques Semliki/inmunología , Neoplasias Cutáneas/terapia , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Carcinoma/inmunología , Carcinoma/mortalidad , Línea Celular Tumoral , Cricetinae , Expresión Génica/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Inmunoterapia/métodos , Inyecciones Intralesiones , Inyecciones Intravenosas , Interleucina-12/administración & dosificación , Interleucina-12/genética , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Ratones , Virus de los Bosques Semliki/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Tasa de Supervivencia , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
9.
STAR Protoc ; 4(1): 102082, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36861834

RESUMEN

Recognition of Cas9 and other proteins encoded in delivery vectors has limited CRISPR technology in vivo. Here, we present a protocol for genome engineering using selective CRISPR antigen removal (SCAR) lentiviral vectors in Renca mouse model. This protocol describes how to conduct an in vivo genetic screen with a sgRNA library and SCAR vectors that can be applied to different cell lines and contexts. For complete details on the use and execution of this protocol, please refer to Dubrot et al. (2021).1.


Asunto(s)
Sistemas CRISPR-Cas , ARN Guía de Sistemas CRISPR-Cas , Ratones , Animales , Sistemas CRISPR-Cas/genética , Biblioteca de Genes , Genoma , Línea Celular
10.
Nat Commun ; 14(1): 8048, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052854

RESUMEN

CAR-T therapy is a promising, novel treatment modality for B-cell malignancies and yet many patients relapse through a variety of means, including loss of CAR-T cells and antigen escape. To investigate leukemia-intrinsic CAR-T resistance mechanisms, we performed genome-wide CRISPR-Cas9 loss-of-function screens in an immunocompetent murine model of B-cell acute lymphoblastic leukemia (B-ALL) utilizing a modular guide RNA library. We identified IFNγR/JAK/STAT signaling and components of antigen processing and presentation pathway as key mediators of resistance to CAR-T therapy in vivo; intriguingly, loss of this pathway yielded the opposite effect in vitro (sensitized leukemia to CAR-T cells). Transcriptional characterization of this model demonstrated upregulation of these pathways in tumors relapsed after CAR-T treatment, and functional studies showed a surprising role for natural killer (NK) cells in engaging this resistance program. Finally, examination of data from B-ALL patients treated with CAR-T revealed an association between poor outcomes and increased expression of JAK/STAT and MHC-I in leukemia cells. Overall, our data identify an unexpected mechanism of resistance to CAR-T therapy in which tumor cell interaction with the in vivo tumor microenvironment, including NK cells, induces expression of an adaptive, therapy-induced, T-cell resistance program in tumor cells.


Asunto(s)
Linfoma de Burkitt , Leucemia , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , ARN Guía de Sistemas CRISPR-Cas , Inmunoterapia Adoptiva , Linfocitos T , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Microambiente Tumoral
11.
Cancer Discov ; 12(3): 812-835, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34848557

RESUMEN

Isocitrate dehydrogenase 1 mutations (mIDH1) are common in cholangiocarcinoma. (R)-2-hydroxyglutarate generated by the mIDH1 enzyme inhibits multiple α-ketoglutarate-dependent enzymes, altering epigenetics and metabolism. Here, by developing mIDH1-driven genetically engineered mouse models, we show that mIDH1 supports cholangiocarcinoma tumor maintenance through an immunoevasion program centered on dual (R)-2-hydroxyglutarate-mediated mechanisms: suppression of CD8+ T-cell activity and tumor cell-autonomous inactivation of TET2 DNA demethylase. Pharmacologic mIDH1 inhibition stimulates CD8+ T-cell recruitment and interferon γ (IFNγ) expression and promotes TET2-dependent induction of IFNγ response genes in tumor cells. CD8+ T-cell depletion or tumor cell-specific ablation of TET2 or IFNγ receptor 1 causes treatment resistance. Whereas immune-checkpoint activation limits mIDH1 inhibitor efficacy, CTLA4 blockade overcomes immunosuppression, providing therapeutic synergy. The findings in this mouse model of cholangiocarcinoma demonstrate that immune function and the IFNγ-TET2 axis are essential for response to mIDH1 inhibition and suggest a novel strategy for potentiating efficacy. SIGNIFICANCE: Mutant IDH1 inhibition stimulates cytotoxic T-cell function and derepression of the DNA demethylating enzyme TET2, which is required for tumor cells to respond to IFNγ. The discovery of mechanisms of treatment efficacy and the identification of synergy by combined CTLA4 blockade provide the foundation for new therapeutic strategies. See related commentary by Zhu and Kwong, p. 604. This article is highlighted in the In This Issue feature, p. 587.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Dioxigenasas , Animales , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/metabolismo , Antígeno CTLA-4/genética , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Humanos , Interferón gamma/genética , Isocitrato Deshidrogenasa , Ratones , Mutación
12.
Int J Cancer ; 128(1): 105-18, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20309938

RESUMEN

CD137 artificial costimulation results in complete tumor rejection in several mouse models. Type I interferons (IFN) exert antitumor effects through an array of molecular functions on malignant cells, tumor stroma and immune system cells. The fact that agonist anti-CD137 mAb induce tumor regressions in mice deficient in the unique receptor for Type I IFNs (IFNAR(-/-) ) indicated potential for treatment combinations. Indeed, combination of intratumor injections of mouse IFN-α and intraperitoneal injections of anti-CD137 mAb synergized as seen on subcutaneous lesions derived from the MC38 colon carcinoma, which is resistant to each treatment if given separately. Therapeutic activity was achieved both against lesions directly injected with IFN-α and against distant concomitant tumors. Experiments in bone marrow chimeras prepared with IFNAR(-/-) and WT mice concluded that expression of the receptor for Type I interferons is mainly required on cells of the hematopoietic compartment. Synergistic effects correlated with a remarkable cellular hyperplasia of the tumor draining lymph nodes (TDLNs). Enlarged TDLNs contained more plasmacytoid and conventional dendritic cells (DC) that more readily cross-presented. Importantly, numbers of both DC subtypes inversely correlated with the tumor size. Numbers of CD8 T cells specific for a dominant tumor antigen were increased at TDLNs by each separate treatment but only with slight augments due to the combination. Combined antitumor effects of the therapeutic strategy were also seen on subcutaneous TC-1 tumors established for 24 days before treatment onset. The described strategy is realistic because (i) agents of each kind are clinically available and (ii) equivalent procedures in humans are feasible.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inmunoterapia/métodos , Neoplasias Experimentales/terapia , Receptor de Interferón alfa y beta/deficiencia , Animales , Anticuerpos Monoclonales/administración & dosificación , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Citometría de Flujo , Humanos , Inyecciones Intralesiones , Inyecciones Intraperitoneales , Interferón-alfa/administración & dosificación , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Receptor de Interferón alfa y beta/genética , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
13.
Int J Cancer ; 129(2): 374-86, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20853321

RESUMEN

Anti-CTLA-4 monoclonal antibodies (mAb) that block the interaction of CTLA-4 with CD80 and CD86 such as tremelimumab and ipilimumab are currently being tested in the clinic for cancer treatment exploiting their properties to de-repress tumor-specific cellular immunity. Addition of the fully human anti-CTLA-4 (tremelimumab) to cultures of human T cells with allogenic dendritic cells (DCs) did not increase proliferation. Magnetic bead-mediated elimination of CD4(+) CD25(+) regulatory T cells (T(reg)) before setting up those alloreactive cultures also largely failed to increase primary proliferation. In contrast, predepletion of CD4(+) CD25(+) T(reg) and culture in the presence of tremelimumab synergistically resulted in increased proliferation and DC:T-cell aggregation. These effects were much more prominent in CD4 than in CD8 T cells. The synergy mechanism can be traced to enhanced CTLA-4 expression in effector cells as a result of T(reg) elimination, thereby offering more targets to the blocking antibody. Human T cells and allogenic DCs (derived both from healthy donors and advanced cancer patients) were coinjected in the peritoneum of Rag2(-/-) IL-2Rγ(-/-) mice. In these conditions, tremelimumab injected intravenously did not significantly enhance alloreactive proliferation unless T(reg) cells had been predepleted. Synergistic effects in vivo were again largely restricted to the CD4 T-cell compartment. In addition, T(reg) depletion and CTLA-4 blockade synergistically enhanced specific cytotoxicity raised in culture against autologous EBV-transformed cell lines. Taken together, these experiments indicate that tremelimumab therapy may benefit from previous or concomitant T(reg) depletion.


Asunto(s)
Anticuerpos Bloqueadores , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Factores Inmunológicos/farmacología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales Humanizados , Antígeno CTLA-4 , Proliferación Celular/efectos de los fármacos , Trasplante de Células , Células Cultivadas , Células Dendríticas/trasplante , Humanos , Ratones , Ratones Transgénicos , Linfocitos T/inmunología , Linfocitos T/trasplante , Linfocitos T Reguladores/efectos de los fármacos
14.
Eur J Immunol ; 40(12): 3389-402, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21108462

RESUMEN

IFN-α/ß link innate and adaptive immune responses by directly acting on naïve CD8(+) T cells. This concept unveiled in mice remains unexplored in humans. To investigate that, human CD8(+) CD45RO(-) cells were stimulated with beads coated with anti-CD3 and anti-CD28 mAb, mimicking Ag (type-1) and co-stimulatory (type-2) signals, in the presence or absence of IFN-α and their transcriptional profiles were defined by cDNA-microarrays. We show that IFN-α provides a strong third signal directly to human CD8(+) T cells resulting in regulation of critical genes for their overall activation. This transcriptional effect was substantiated at the protein level and verified by functional assays. Interestingly, the biological effects derived from this stimulation vary depending on the CD8(+) T-cell population. Thus, whereas IFN-α increases the proliferative capacity of naïve CD8(+) T cells, it inhibits or does not affect the proliferation of Ag-experienced cells, such as memory and effector CTL, including CMV-specific lymphocytes. Cytolysis and IFN-γ-secretion of all these populations are enhanced by IFN-α-derived signals, which are critical in naïve CD8(+) T cells for acquisition of effector functions. Our findings in human CD8(+) T cells are informative to understand and improve IFN-α-based therapies for viral and malignant diseases.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Interferón-alfa/farmacología , Interferón gamma/biosíntesis , Fosfoproteínas/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Proteínas de la Matriz Viral/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Inmunización , Memoria Inmunológica/efectos de los fármacos , Interferón gamma/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
15.
Eur J Immunol ; 40(11): 3054-63, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21061437

RESUMEN

Migration of DC into lymphatic vessels ferries antigenic cargo and pro-inflammatory stimuli into the draining LN. Given that tissues under the influence of viral infections produce type I IFN, it is conceivable that these cytokines enhance DC migration in order to facilitate an antiviral immune response. Cultured lymphatic endothelium monolayers pretreated with TNF-α were used to model this phenomenon under inflammatory conditions. DC differentiated in the presence of either IFN-α2b or IFN-α5 showed enhanced adhesion to cultured lymphatic endothelial cells. These pro-adhesive effects were mediated by DC, not the lymphatic endothelium, and correlated with increased DC transmigration across lymphatic endothelial cell monolayers. Transmigration was guided by chemokines acting on DC, and blocking experiments with mAb indicated a role for LFA-1. Furthermore, incubation of DC with IFN-α led to the appearance of active conformation epitopes on the CD11a integrin chains expressed by DC. Differentiation of mouse DC in the presence of IFN-α also increased DC migration from inflammed footpads toward popliteal LN. Collectively, these results indicate a role for type I IFN in directing DC toward LN under inflammatory conditions.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Endotelio Linfático/inmunología , Interferón-alfa/inmunología , Animales , Antígeno CD11a/inmunología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Humanos , Interferón-alfa/farmacología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Ratones , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología
16.
Mol Ther ; 18(4): 754-65, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20087317

RESUMEN

Repeated administration of gene therapies is hampered by host immunity toward vectors and transgenes. Attempts to circumvent antivector immunity include pharmacological immunosuppression or alternating different vectors and vector serotypes with the same transgene. Our studies show that B-cell depletion with anti-CD20 monoclonal antibody and concomitant T-cell inhibition with clinically available drugs permits repeated liver gene transfer to a limited number of nonhuman primates with recombinant adenovirus. Adenoviral vector-mediated transfer of the herpes simplex virus type 1 thymidine kinase (HSV1-tk) reporter gene was visualized in vivo with a semiquantitative transgene-specific positron emission tomography (PET) technique, liver immunohistochemistry, and immunoblot for the reporter transgene in needle biopsies. Neutralizing antibody and T cell-mediated responses toward the viral capsids were sequentially monitored and found to be repressed by the drug combinations tested. Repeated liver transfer of the HSV1-tk reporter gene with the same recombinant adenoviral vector was achieved in macaques undergoing a clinically feasible immunosuppressive treatment that ablated humoral and cellular immune responses. This strategy allows measurable gene retransfer to the liver as late as 15 months following the first adenoviral exposure in a macaque, which has undergone a total of four treatments with the same adenoviral vector.


Asunto(s)
Adenoviridae/inmunología , Anticuerpos Monoclonales/administración & dosificación , Técnicas de Transferencia de Gen , Vectores Genéticos/inmunología , Inmunosupresores/administración & dosificación , Hígado/inmunología , Depleción Linfocítica , Adenoviridae/genética , Animales , Anticuerpos Monoclonales de Origen Murino , Anticuerpos Neutralizantes/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Femenino , Terapia Genética , Vectores Genéticos/genética , Macaca , Virus Reordenados , Recombinación Genética , Rituximab , Linfocitos T/inmunología , Tacrolimus/administración & dosificación
17.
J Exp Med ; 218(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33861848

RESUMEN

Lymphatic endothelial cells (LECs) present peripheral tissue antigens to induce T cell tolerance. In addition, LECs are the main source of sphingosine-1-phosphate (S1P), promoting naive T cell survival and effector T cell exit from lymph nodes (LNs). Autophagy is a physiological process essential for cellular homeostasis. We investigated whether autophagy in LECs modulates T cell activation in experimental arthritis. Whereas genetic abrogation of autophagy in LECs does not alter immune homeostasis, it induces alterations of the regulatory T cell (T reg cell) population in LNs from arthritic mice, which might be linked to MHCII-mediated antigen presentation by LECs. Furthermore, inflammation-induced autophagy in LECs promotes the degradation of Sphingosine kinase 1 (SphK1), resulting in decreased S1P production. Consequently, in arthritic mice lacking autophagy in LECs, pathogenic Th17 cell migration toward LEC-derived S1P gradients and egress from LNs are enhanced, as well as infiltration of inflamed joints, resulting in exacerbated arthritis. Our results highlight the autophagy pathway as an important regulator of LEC immunomodulatory functions in inflammatory conditions.


Asunto(s)
Autoinmunidad/inmunología , Células Endoteliales/inmunología , Macroautofagia/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Artritis/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Humanos , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Ganglios Linfáticos/inmunología , Vasos Linfáticos/inmunología , Lisofosfolípidos/inmunología , Ratones , Ratones Endogámicos C57BL , Esfingosina/análogos & derivados , Esfingosina/inmunología
18.
Cancer Immunol Res ; 9(7): 748-764, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33952631

RESUMEN

Several solid malignancies trigger lymphangiogenesis, facilitating metastasis. Tumor-associated lymphatic vessels significantly contribute to the generation of an immunosuppressive tumor microenvironment (TME). Here, we have investigated the ability of tumoral lymphatic endothelial cells (LEC) to function as MHC class II-restricted antigen-presenting cells in the regulation of antitumor immunity. Using murine models of lymphangiogenic tumors engrafted under the skin, we have shown that tumoral LECs upregulate MHC class II and the MHC class II antigen-processing machinery, and that they promote regulatory T-cell (Treg) expansion ex vivo. In mice with LEC-restricted lack of MHC class II expression, tumor growth was severely impaired, whereas tumor-infiltrating effector T cells were increased. Reduction of tumor growth and reinvigoration of tumor-specific T-cell responses both resulted from alterations of the tumor-infiltrating Treg transcriptome and phenotype. Treg-suppressive functions were profoundly altered in tumors lacking MHC class II in LECs. No difference in effector T-cell responses or Treg phenotype and functions was observed in tumor-draining lymph nodes, indicating that MHC class II-restricted antigen presentation by LECs was required locally in the TME to confer potent suppressive functions to Tregs. Altogether, our study suggests that MHC class II-restricted antigen-presenting tumoral LECs function as a local brake, dampening T cell-mediated antitumor immunity and promoting intratumoral Treg-suppressive functions.


Asunto(s)
Células Endoteliales/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Escape del Tumor , Animales , Presentación de Antígeno , Comunicación Celular/inmunología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Femenino , Humanos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Vasos Linfáticos/citología , Vasos Linfáticos/inmunología , Ratones , Cultivo Primario de Células , Células Tumorales Cultivadas , Microambiente Tumoral/inmunología
19.
J Hepatol ; 52(3): 417-24, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19815305

RESUMEN

BACKGROUND & AIMS: Acute intermittent porphyria (AIP) is characterized by hepatic porphobilinogen deaminase (PBGD) deficiency resulting in a marked overproduction of presumably toxic porphyrin precursors. Our study aimed to assess the protective effects of bone marrow transplantation or PBGD gene transfer into the liver against phenotypic manifestations of acute porphyria attack induced in an AIP murine model. METHODS: Lethally irradiated AIP mice were intravenously injected with 5x10(6) nucleated bone marrow cells from wild type or AIP donor mice. To achieve liver gene transfer, AIP mice received via hydrodynamic injection plasmids expressing human PBGD or luciferase, driven by a liver-specific promoter. RESULTS: Erythrocyte PBGD activity increased 2.4-fold in AIP mice receiving bone marrow cells from normal animals. Nevertheless, phenobarbital administration in these mice reproduced key features of acute attacks, such as massively increased urinary porphyrin precursor excretion and decreased motor coordination. Hepatic PBGD activity increased 2.2-fold after hydrodynamic injection of therapeutic plasmid. Mice injected with the luciferase control plasmid showed a high excretion of porphyrin precursors after phenobarbital administration whereas just a small increase was observed in AIP mice injected with the PBGD plasmid. Furthermore, motor disturbance was almost completely abolished in AIP mice treated with the therapeutic plasmid. CONCLUSIONS: PBGD deficiency in erythroid tissue is not associated with phenotypic manifestations of acute porphyria. In contrast, PBGD over-expression in hepatocytes, albeit in a low proportion, reduced precursor accumulation, which is the hallmark of acute porphyric attacks. Liver-directed gene therapy might offer an alternative to liver transplantation applicable in patients with severe and recurrent manifestations.


Asunto(s)
Eritrocitos/metabolismo , Hepatocitos/metabolismo , Hidroximetilbilano Sintasa/metabolismo , Porfiria Intermitente Aguda/metabolismo , Porfirinas/metabolismo , Ácido Aminolevulínico/metabolismo , Animales , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Eritrocitos/patología , Femenino , Técnicas de Transferencia de Gen , Hepatocitos/patología , Humanos , Hidroximetilbilano Sintasa/genética , Ratones , Ratones Endogámicos C57BL , Plásmidos , Porfobilinógeno/metabolismo , Porfiria Intermitente Aguda/patología , Porfiria Intermitente Aguda/terapia
20.
Eur J Immunol ; 39(9): 2424-36, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19662633

RESUMEN

Anti-CD137 mAb are capable of inducing tumor rejection in several syngeneic murine tumor models and are undergoing clinical trials for cancer. The anti-tumor effect involves co-stimulation of tumor-specific CD8(+) T cells. Whether antigen cross-presenting DC are required for the efficacy of anti-CD137 mAb treatment has never been examined. Here we show that the administration of anti-CD137 mAb eradicates EG7-OVA tumors by a strictly CD8beta(+) T-cell-dependent mechanism that correlates with increased CTL activity. Ex vivo analyses to determine the identity of the draining lymph node cell type responsible for tumor antigen cross-presentation revealed that CD11c(+) cells, most likely DC, are the main players in this tumor model. A minute number of tumor cells, revealed by the presence of OVA cDNA, reach tumor-draining lymph nodes. Direct antigen presentation by tumor cells themselves also participates in anti-OVA CTL induction. Using CD11c diphtheria toxin receptor-green fluorescent protein-->C57BL/6 BM chimeric mice, which allow for sustained ablation of DC with diphtheria toxin, we confirmed the involvement of DC in tumor antigen cross-presentation in CTL induction against OVA(257-264) epitope and in the antitumor efficacy induced by anti-CD137 mAb.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Células Dendríticas/inmunología , Neoplasias/terapia , Linfocitos T Citotóxicos/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Animales , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Reactividad Cruzada/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/inmunología , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA