Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biomacromolecules ; 25(3): 1749-1758, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38236997

RESUMEN

The antitumor immunity can be enhanced through the synchronized codelivery of antigens and immunostimulatory adjuvants to antigen-presenting cells, particularly dendritic cells (DCs), using nanovaccines (NVs). To study the influence of intracellular vaccine cargo release kinetics on the T cell activating capacities of DCs, we compared stimuli-responsive to nonresponsive polymersome NVs. To do so, we employed "AND gate" multiresponsive (MR) amphiphilic block copolymers that decompose only in response to the combination of chemical cues present in the environment of the intracellular compartments in antigen cross-presenting DCs: low pH and high reactive oxygen species (ROS) levels. After being unmasked by ROS, pH-responsive side chains are exposed and can undergo a charge shift within a relevant pH window of the intracellular compartments in antigen cross-presenting DCs. NVs containing the model antigen Ovalbumin (OVA) and the iNKT cell activating adjuvant α-Galactosylceramide (α-Galcer) were fabricated using microfluidics self-assembly. The MR NVs outperformed the nonresponsive NV in vitro, inducing enhanced classical- and cross-presentation of the OVA by DCs, effectively activating CD8+, CD4+ T cells, and iNKT cells. Interestingly, in vivo, the nonresponsive NVs outperformed the responsive vaccines. These differences in polymersome vaccine performance are likely linked to the kinetics of cargo release, highlighting the crucial chemical requirements for successful cancer nanovaccines.


Asunto(s)
Nanovacunas , Vacunas , Animales , Ratones , Especies Reactivas de Oxígeno , Linfocitos T CD8-positivos , Células Dendríticas , Antígenos/química , Adyuvantes Inmunológicos/farmacología , Vacunas/química , Ovalbúmina , Concentración de Iones de Hidrógeno , Ratones Endogámicos C57BL
2.
ACS Nano ; 17(13): 12101-12117, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37338806

RESUMEN

Adoptive T cell therapy has successfully been implemented for the treatment of cancer. Nevertheless, ex vivo expansion of T cells by artificial antigen-presenting cells (aAPCs) remains cumbersome and can compromise T cell functionality, thereby limiting their therapeutic potential. We propose a radically different approach aimed at direct expansion of T cells in vivo, thereby omitting the need for large-scale ex vivo T cell production. We engineered nanosized immunofilaments (IFs), with a soluble semiflexible polyisocyanopeptide backbone that presents peptide-loaded major histocompatibility complexes and costimulatory molecules multivalently. IFs readily activated and expanded antigen-specific T cells like natural APCs, as evidenced by transcriptomic analyses of T cells. Upon intravenous injection, IFs reach the spleen and lymph nodes and induce antigen-specific T cell responses in vivo. Moreover, IFs display strong antitumor efficacy resulting in inhibition of the formation of melanoma metastases and reduction of primary tumor growth in synergy with immune checkpoint blockade. In conclusion, nanosized IFs represent a powerful modular platform for direct activation and expansion of antigen-specific T cells in vivo, which can greatly contribute to cancer immunotherapy.


Asunto(s)
Melanoma , Linfocitos T , Humanos , Células Presentadoras de Antígenos , Melanoma/terapia , Inmunoterapia , Inmunoterapia Adoptiva
3.
J Immunother Cancer ; 10(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35428705

RESUMEN

BACKGROUND: Type 1 conventional dendritic cells (cDC1s) are characterized by their ability to induce potent CD8+ T cell responses. In efforts to generate novel vaccination strategies, notably against cancer, human cDC1s emerge as an ideal target to deliver antigens. cDC1s uniquely express XCR1, a seven transmembrane G protein-coupled receptor. Due to its restricted expression and endocytic nature, XCR1 represents an attractive receptor to mediate antigen-delivery to human cDC1s. METHODS: To explore tumor antigen delivery to human cDC1s, we used an engineered version of XCR1-binding lymphotactin (XCL1), XCL1(CC3). Site-specific sortase-mediated transpeptidation was performed to conjugate XCL1(CC3) to an analog of the HLA-A*02:01 epitope of the cancer testis antigen New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1). While poor epitope solubility prevented isolation of stable XCL1-antigen conjugates, incorporation of a single polyethylene glycol (PEG) chain upstream of the epitope-containing peptide enabled generation of soluble XCL1(CC3)-antigen fusion constructs. Binding and chemotactic characteristics of the XCL1-antigen conjugate, as well as its ability to induce antigen-specific CD8+ T cell activation by cDC1s, was assessed. RESULTS: PEGylated XCL1(CC3)-antigen conjugates retained binding to XCR1, and induced cDC1 chemoattraction in vitro. The model epitope was efficiently cross-presented by human cDC1s to activate NY-ESO-1-specific CD8+ T cells. Importantly, vaccine activity was increased by targeting XCR1 at the surface of cDC1s. CONCLUSION: Our results present a novel strategy for the generation of targeted vaccines fused to insoluble antigens. Moreover, our data emphasize the potential of targeting XCR1 at the surface of primary human cDC1s to induce potent CD8+ T cell responses.


Asunto(s)
Antígenos de Neoplasias , Vacunas contra el Cáncer , Células Dendríticas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Linfocinas , Proteínas de la Membrana , Sialoglicoproteínas , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Reactividad Cruzada , Células Dendríticas/inmunología , Epítopos/inmunología , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/terapia , Carcinoma de Células Escamosas de Esófago/inmunología , Carcinoma de Células Escamosas de Esófago/terapia , Humanos , Linfocinas/administración & dosificación , Linfocinas/inmunología , Masculino , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/inmunología , Sialoglicoproteínas/administración & dosificación , Sialoglicoproteínas/inmunología
4.
Adv Healthc Mater ; 10(13): e2100304, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34050625

RESUMEN

Anticancer drug delivery strategies are designed to take advantage of the differential chemical environment in solid tumors independently, or to high levels of reactive oxygen species (ROS) or to low pH, compared to healthy tissue. Here, the design and thorough characterization of two functionalizable "AND gate" multiresponsive (MR) block amphiphilic copolymers are reported, aimed to take full advantage of the coexistence of two chemical cues-ROS and low pH-present in the tumor microenvironment. The hydrophobic blocks contain masked pH-responsive side chains, which are exposed exclusively in response to ROS. Hence, the hydrophobic polymer side chains will undergo a charge shift in a very relevant pH window present in the extracellular milieu in most solid tumors (pH 5.6-7.2) after demasking by ROS. Doxorubicin (DOX)-loaded nanosized "AND gate" MR polymersomes (MRPs) are fabricated via microfluidic self-assembly. Chemical characterization reveals ROS-dependent pH sensitivity and accelerated DOX release under influence of both ROS and low pH. Treatment of tumor-bearing mice with DOX-loaded nonresponsive and "AND gate" MRPs dramatically decreases cardiac toxicity. The most optimal "AND gate" MRPs outperform free DOX in terms of tumor growth inhibition and survival, shedding light on chemical requirements for successful cancer nanomedicine.


Asunto(s)
Nanomedicina , Nanopartículas , Animales , Doxorrubicina/farmacología , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Concentración de Iones de Hidrógeno , Ratones , Micelas , Oxígeno , Especies Reactivas de Oxígeno
5.
Front Immunol ; 12: 641703, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717196

RESUMEN

Tumor-specific neoantigens can be highly immunogenic, but their identification for each patient and the production of personalized cancer vaccines can be time-consuming and prohibitively expensive. In contrast, tumor-associated antigens are widely expressed and suitable as an off the shelf immunotherapy. Here, we developed a PLGA-based nanoparticle vaccine that contains both the immunogenic cancer germline antigen NY-ESO-1 and an α-GalCer analog IMM60, as a novel iNKT cell agonist and dendritic cell transactivator. Three peptide sequences (85-111, 117-143, and 157-165) derived from immunodominant regions of NY-ESO-1 were selected. These peptides have a wide HLA coverage and were efficiently processed and presented by dendritic cells via various HLA subtypes. Co-delivery of IMM60 enhanced CD4 and CD8 T cell responses and antibody levels against NY-ESO-1 in vivo. Moreover, the nanoparticles have negligible systemic toxicity in high doses, and they could be produced according to GMP guidelines. Together, we demonstrated the feasibility of producing a PLGA-based nanovaccine containing immunogenic peptides and an iNKT cell agonist, that is activating DCs to induce antigen-specific T cell responses.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Portadores de Fármacos/farmacología , Nanopartículas/uso terapéutico , Proteínas de Neoplasias/farmacología , Fragmentos de Péptidos/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Portadores de Fármacos/química , Humanos , Nanopartículas/química , Proteínas de Neoplasias/química , Fragmentos de Péptidos/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
6.
Oncoimmunology ; 9(1): 1738813, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33457086

RESUMEN

Nanovaccines, co-delivering antigen and invariant natural killer T (iNKT) cell agonists, proved to be very effective in inducing anti-tumor T cell responses due to their exceptional helper function. However, it is known that iNKT cells are not equally present in all lymphoid organs and nanoparticles do not get evenly distributed to all immune compartments. In this study, we evaluated the effect of the vaccination route on iNKT cell help to T and B cell responses for the first time in an antigen and agonist co-delivery setting. Intravenous administration of PLGA nanoparticles was mainly targeting liver and spleen where iNKT1 cells are abundant and induced the highest serum IFN-y levels, T cell cytotoxicity, and Th-1 type antibody responses. In comparison, after subcutaneous or intranodal injections, nanoparticles mostly drained or remained in regional lymph nodes where iNKT17 cells were abundant. After subcutaneous and intranodal injections, antigen-specific IgG2 c production was hampered and IFN-y production, as well as cytotoxic T cell responses, depended on sporadic systemic drainage. Therapeutic anti-tumor experiments also demonstrated a clear advantage of intravenous injection over intranodal or subcutaneous vaccinations. Moreover, tumor control could be further improved by PD-1 immune checkpoint blockade after intravenous vaccination, but not by intranodal vaccination. Anti PD-1 antibody combination mainly exerts its effect by prolonging the cytotoxicity of T cells. Nanovaccines also demonstrated synergism with anti-4-1BB agonistic antibody treatment in controlling tumor growth. We conclude that nanovaccines containing iNKT cell agonists shall be preferentially administered intravenously, to optimally reach cellular partners for inducing effective anti-tumor immune responses.


Asunto(s)
Células T Asesinas Naturales , Neoplasias , Anticuerpos , Linfocitos B , Humanos
7.
iScience ; 22: 240-255, 2019 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-31786520

RESUMEN

Endogenous extracellular Galectins constitute a novel mechanism of membrane protein organization at the cell surface. Although Galectins are also highly expressed intracellularly, their cytosolic functions are poorly understood. Here, we investigated the role of Galectin-9 in dendritic cell (DC) surface organization and function. By combining functional, super-resolution and atomic force microscopy experiments to analyze membrane stiffness, we identified intracellular Galectin-9 to be indispensable for plasma membrane integrity and structure in DCs. Galectin-9 knockdown studies revealed intracellular Galectin-9 to directly control cortical membrane structure by modulating Rac1 activity, providing the underlying mechanism of Galectin-9-dependent actin cytoskeleton organization. Consequent to its role in maintaining plasma membrane structure, phagocytosis studies revealed that Galectin-9 was essential for C-type-lectin receptor-mediated pathogen uptake by DCs. This was confirmed by the impaired phagocytic capacity of Galectin-9-null murine DCs. Together, this study demonstrates a novel role for intracellular Galectin-9 in modulating DC function, which may be evolutionarily conserved.

8.
Carbohydr Res ; 486: 107840, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31689579

RESUMEN

Herein, an efficient synthesis of BODIPY-α-Galactosylceramide 3, which can be used to study the cellular uptake of the potent immunostimulatory parent compound α-Galactosylceramide, is reported. Key in our synthetic strategy is the six-step synthesis of the core BODIPY scaffold (64% yield overall) and its quantitative conversion to an N-hydroxysuccinimidyl ester to facilitate conjugation and purification of the target glycolipid. For the preparation of the core of the glycolipid, the solubility of the lipid acceptor proved to be critical. The ability of BODIPY-αGalCer 3 to activate invariant natural killer cells was then demonstrated in vitro.


Asunto(s)
Compuestos de Boro/química , Compuestos de Boro/síntesis química , Galactosilceramidas/química , Galactosilceramidas/metabolismo , Sondas Moleculares/química , Sondas Moleculares/síntesis química , Transporte Biológico , Línea Celular , Técnicas de Química Sintética
9.
Pharmaceutics ; 11(11)2019 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-31717354

RESUMEN

Polymeric particles made up of biodegradable and biocompatible polymers such as poly(lactic-co-glycolic acid) (PLGA) are promising tools for several biomedical applications including drug delivery. Particular emphasis is placed on the size and surface functionality of these systems as they are regarded as the main protagonists in dictating the particle behavior in vitro and in vivo. Current methods of manufacturing polymeric drug carriers offer a wide range of achievable particle sizes, however, they are unlikely to accurately control the size while maintaining the same production method and particle uniformity, as well as final production yield. Microfluidics technology has emerged as an efficient tool to manufacture particles in a highly controllable manner. Here, we report on tuning the size of PLGA particles at diameters ranging from sub-micron to microns using a single microfluidics device, and demonstrate how particle size influences the release characteristics, cellular uptake and in vivo clearance of these particles. Highly controlled production of PLGA particles with ~100 nm, ~200 nm, and >1000 nm diameter is achieved through modification of flow and formulation parameters. Efficiency of particle uptake by dendritic cells and myeloid-derived suppressor cells isolated from mice is strongly correlated with particle size and is most efficient for ~100 nm particles. Particles systemically administered to mice mainly accumulate in liver and ~100 nm particles are cleared slower. Our study shows the direct relation between particle size varied through microfluidics and the pharmacokinetics behavior of particles, which provides a further step towards the establishment of a customizable production process to generate tailor-made nanomedicines.

10.
Sci Adv ; 5(8): eaaw1822, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31489367

RESUMEN

Hybridoma technology is instrumental for the development of novel antibody therapeutics and diagnostics. Recent preclinical and clinical studies highlight the importance of antibody isotype for therapeutic efficacy. However, since the sequence encoding the constant domains is fixed, tuning antibody function in hybridomas has been restricted. Here, we demonstrate a versatile CRISPR/HDR platform to rapidly engineer the constant immunoglobulin domains to obtain recombinant hybridomas, which secrete antibodies in the preferred format, species, and isotype. Using this platform, we obtained recombinant hybridomas secreting Fab' fragments, isotype-switched chimeric antibodies, and Fc-silent mutants. These antibody products are stable, retain their antigen specificity, and display their intrinsic Fc-effector functions in vitro and in vivo. Furthermore, we can site-specifically attach cargo to these antibody products via chemoenzymatic modification. We believe that this versatile platform facilitates antibody engineering for the entire scientific community, empowering preclinical antibody research.


Asunto(s)
Anticuerpos Monoclonales/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Hibridomas/fisiología , Animales , Especificidad de Anticuerpos/genética , Línea Celular Tumoral , Genómica/métodos , Fragmentos Fab de Inmunoglobulinas/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/genética
11.
Adv Funct Mater ; 29(19)2019 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32132881

RESUMEN

Ultrasound is the most commonly used clinical imaging modality. However, in applications requiring cell-labeling, the large size and short active lifetime of ultrasound contrast agents limit their longitudinal use. Here, 100 nm radius, clinically applicable, polymeric nanoparticles containing a liquid perfluorocarbon, which enhance ultrasound contrast during repeated ultrasound imaging over the course of at least 48 h, are described. The perfluorocarbon enables monitoring the nanoparticles with quantitative 19F magnetic resonance imaging, making these particles effective multimodal imaging agents. Unlike typical core-shell perfluorocarbon-based ultrasound contrast agents, these nanoparticles have an atypical fractal internal structure. The nonvaporizing highly hydrophobic perfluorocarbon forms multiple cores within the polymeric matrix and is, surprisingly, hydrated with water, as determined from small-angle neutron scattering and nuclear magnetic resonance spectroscopy. Finally, the nanoparticles are used to image therapeutic dendritic cells with ultrasound in vivo, as well as with 19F MRI and fluorescence imaging, demonstrating their potential for long-term in vivo multimodal imaging.

12.
Front Immunol ; 9: 2798, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30546367

RESUMEN

Biomaterial-based scaffolds are promising tools for controlled immunomodulation. They can be applied as three dimensional (3D) culture systems in vitro, whereas in vivo they may be used to dictate cellular localization and exert spatiotemporal control over cues presented to the immune system. As such, scaffolds can be exploited to enhance the efficacy of cancer immunotherapies such as adoptive T cell transfer, in which localization and persistence of tumor-specific T cells dictates treatment outcome. Biomimetic polyisocyanopeptide (PIC) hydrogels are polymeric scaffolds with beneficial characteristics as they display reversible thermally-induced gelation at temperatures above 16°C, which allows for their minimally invasive delivery via injection. Moreover, incorporation of azide-terminated monomers introduces functional handles that can be exploited to include immune cell-modulating cues. Here, we explore the potential of synthetic PIC hydrogels to promote the in vitro expansion and in vivo local delivery of pre-activated T cells. We found that PIC hydrogels support the survival and vigorous expansion of pre-stimulated T cells in vitro even at high cell densities, highlighting their potential as 3D culture systems for efficient expansion of T cells for their adoptive transfer. In particular, the reversible thermo-sensitive behavior of the PIC scaffolds favors straightforward recovery of cells. PIC hydrogels that were injected subcutaneously gelated instantly in vivo, after which a confined 3D structure was formed that remained localized for at least 4 weeks. Importantly, we noticed no signs of inflammation, indicating that PIC hydrogels are non-immunogenic. Cells co-delivered with PIC polymers were encapsulated within the scaffold in vivo. Cells egressed gradually from the PIC gel and migrated into distant organs. This confirms that PIC hydrogels can be used to locally deliver cells within a supportive environment. These results demonstrate that PIC hydrogels are highly promising for both the in vitro expansion and in vivo delivery of pre-activated T cells. Covalent attachment of biomolecules onto azide-functionalized PIC polymers provides the opportunity to steer the phenotype, survival or functional response of the adoptively transferred cells. As such, PIC hydrogels can be used as valuable tools to improve current adoptive T cell therapy strategies.


Asunto(s)
Traslado Adoptivo/métodos , Materiales Biomiméticos , Proliferación Celular/efectos de los fármacos , Hidrogeles , Linfocitos T , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Linfocitos T/inmunología , Linfocitos T/trasplante
13.
Small ; 14(15): e1703539, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29493121

RESUMEN

The activation of tumor-specific effector immune cells is key for successful immunotherapy and vaccination is a powerful strategy to induce such adaptive immune responses. However, the generation of effective anticancer vaccines is challenging. To overcome these challenges, a novel straight-forward strategy of adjuvant-induced tumor antigen assembly to generate nanovaccines with superior antigen/adjuvant loading efficiency is developed. To protect nanovaccines in circulation and to introduce additional functionalities, a biocompatible polyphenol coating is installed. The resulting functionalizable nanovaccines are equipped with a pH (low) insertion peptide (pHLIP) to facilitate endolysosomal escape and to promote cytoplasmic localization, with the aim to enhance cross-presentation of the antigen by dendritic cells to effectively activate CD8+ T cell. The results demonstrate that pHLIP-functionalized model nanovaccine can induce endolysosomal escape and enhance CD8+ T cell activation both in vitro and in vivo. Furthermore, based on the adjuvant-induced antigen assembly, nanovaccines of the clinically relevant tumor-associated antigen NY-ESO-1 are generated and show excellent capacity to elicit NY-ESO-1-specific CD8+ T cell activation, demonstrating a high potential of this functionalizable nanovaccine formulation strategy for clinical applications.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/inmunología , Adyuvantes Inmunológicos , Línea Celular , Humanos , Cinética , Activación de Linfocitos/fisiología , Polifenoles/química
14.
Oncoimmunology ; 5(1): e1068493, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26942088

RESUMEN

Antitumor immunity can be enhanced by the coordinated release and delivery of antigens and immune-stimulating agents to antigen-presenting cells via biodegradable vaccine carriers. So far, encapsulation of TLR ligands and tumor-associated antigens augmented cytotoxic T cell (CTLs) responses. Here, we compared the efficacy of the invariant NKT (iNKT) cell agonist α-galactosylceramide (α-GalCer) and TLR ligands (R848 and poly I:C) as an adjuvant for the full length ovalbumin (OVA) in PLGA nanoparticles. We observed that OVA+α-GalCer nanoparticles (NP) are superior over OVA+TLR-L NP in generating and stimulating antigen-specific cytotoxic T lymphocytes without the need for CD4+ T cell help. Not only a 4-fold higher induction of antigen-specific T cells was observed, but also a more profound IFN-γ secretion was obtained by the addition α-GalCer. Surprisingly, we observed that mixtures of OVA containing NP with α-GalCer were ineffective, demonstrating that co-encapsulation of both α-GalCer and antigen within the same nanoparticle is essential for the observed T cell responses. Moreover, a single immunization with OVA+α-GalCer NP provided substantial protection from tumor formation and even delayed the growth of already established tumors, which coincided with a prominent and enhanced antigen-specific CD8+ T cell infiltration. The provided evidence on the advantage of antigen and α-GalCer coencapsulation should be considered in the design of future nanoparticle vaccines for therapeutic purposes.

15.
Eur J Immunol ; 45(10): 2798-809, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26255864

RESUMEN

Novel approaches of dendritic cell (DC) based cancer immunotherapy aim at harnessing the unique attributes of different DC subsets. Classical monocyte-derived DC vaccines are currently being replaced by either applying primary DCs or specifically targeting antigens and adjuvants to these subsets in vivo. Appropriate DC activation in both strategies is essential for optimal effect. For this purpose TLR agonists are favorable adjuvant choices, with TLR7 triggering being essential for inducing strong Th1 responses. However, mouse CD8α(+) DCs, considered to be the major cross-presenting subset, lack TLR7 expression. Interestingly, this DC subset can respond to TLR7 ligand upon concurrent TLR3 triggering. Nevertheless, the mechanism underlying this synergy remains obscure. We now show that TLR3 ligation results in the production of IFN-α, which rapidly induces the expression of TLR7, resulting in synergistic activation. Moreover, we demonstrate that this mechanism conversely holds for plasmacytoid DCs that respond to TLR3 ligation when TLR7 pathway is mobilized. We further demonstrate that this mechanism of sharpening DC senses is also conserved in human BDCA1(+) DCs and plasmacytoid DCs. These findings have important implications for future clinical trials as it suggests that combinations of TLR ligands should be applied irrespective of initial TLR expression profiles on natural DC subsets for optimal stimulation.


Asunto(s)
Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Glicoproteínas de Membrana/agonistas , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 7/agonistas , Animales , Células Dendríticas/citología , Humanos , Glicoproteínas de Membrana/inmunología , Ratones , Células TH1/citología , Células TH1/inmunología , Receptor Toll-Like 7/inmunología
16.
Cell Immunol ; 295(1): 29-35, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25732602

RESUMEN

Limited knowledge is available on myeloid derived suppressor cells (MDSCs) of rat origin. We examined the myeloid cells from peripheral blood, bone marrow and spleens of healthy and mammary tumor bearing rats employing a novel immunophenotyping strategy with CD172a, HIS48, and Rp-1 antibodies. We addressed rat granulocytes by Rp-1 positivity and used HIS48 in discrimination of two mononuclear cell subsets. An expansion of granulocyte numbers was detected in peripheral blood and spleens of mammary tumor-bearing animals. The purified granulocytes were able to impair antigen-specific helper T-cell proliferation, and therefore nominated as granulocytic MDSCs of this rat tumor model. HIS48(+) mononuclear cell numbers were also increased in the blood and spleens of mammary tumor bearing rats with a lower MHC class II positivity. Despite the lack of an antigen specific suppression of CD4(+) T cells, HIS48(+) monocytes resemble monocytic MDSCs with their inflammatory phenotype. Together, these results provide evidence for the existence and phenotypic characterization of a granulocytic MDSC subset in a rat model of mammary carcinoma.


Asunto(s)
Granulocitos/inmunología , Neoplasias Mamarias Animales/inmunología , Monocitos/inmunología , Células Mieloides/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Granulocitos/metabolismo , Inmunofenotipificación/métodos , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Monocitos/metabolismo , Células Mieloides/metabolismo , Ratas Sprague-Dawley , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología
18.
Eur J Immunol ; 43(3): 747-57, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23175469

RESUMEN

Expression of the B7 family molecules in acute myeloid leukemia (AML) has been demonstrated by independent clinical studies. Intriguingly, the expression of the most potent costimulatory molecules B7-2 (CD86) and B7-H2 (ICOS Ligand) on AML cells has been associated with poor prognosis and disease severity. Here, this phenomenon was modeled in vitro with the myeloid leukemia cell line HL-60, which is capable of differentiating through the FAB M2/M3 and M4/M5 immunophenotypes. These derivatives of HL-60 harbored a B7-2(+) subpopulation and recapitulated the distribution of B7 ligands previously reported in primary AML cases. B7-2(+) AML cells significantly contributed to T-cell responses. This costimulatory activity enabled helper (Th)-cell activation, proliferation, and production of Th1-associated cytokines. Conversely, even a short-term incubation with stimulated T cells resulted in upregulation of inhibitory B7-H1 (PD-L1) and B7-DC (PD-L2), and downregulation of stimulatory B7-H2 molecules on leukemia cells. Purified from iHL-60-T-cell co-cultures, these myeloid leukemia cells severely suppressed Th-cell responses specifically through the PD-1 pathway. In conclusion, Th-cell responses can be directly supported by B7-2(+) leukemia subpopulations. However, this interaction can facilitate the acquisition of a suppressive character that may contribute to immune evasion in myeloid leukemia.


Asunto(s)
Antígeno B7-2/metabolismo , Tolerancia Inmunológica , Leucemia Mieloide/inmunología , Leucemia Mieloide/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Antígeno B7-2/genética , Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Expresión Génica , Células HL-60 , Humanos , Leucemia Mieloide/genética , Ligandos , Activación de Linfocitos/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Proteína Quinasa C/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA