Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunity ; 57(2): 223-244, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38354702

RESUMEN

Immune responses must be tightly regulated to ensure both optimal protective immunity and tolerance. Costimulatory pathways within the B7:CD28 family provide essential signals for optimal T cell activation and clonal expansion. They provide crucial inhibitory signals that maintain immune homeostasis, control resolution of inflammation, regulate host defense, and promote tolerance to prevent autoimmunity. Tumors and chronic pathogens can exploit these pathways to evade eradication by the immune system. Advances in understanding B7:CD28 pathways have ushered in a new era of immunotherapy with effective drugs to treat cancer, autoimmune diseases, infectious diseases, and transplant rejection. Here, we discuss current understanding of the mechanisms underlying the coinhibitory functions of CTLA-4, PD-1, PD-L1:B7-1 and PD-L2:RGMb interactions and less studied B7 family members, including HHLA2, VISTA, BTNL2, and BTN3A1, as well as their overlapping and unique roles in regulating immune responses, and the therapeutic potential of these insights.


Asunto(s)
Enfermedades Autoinmunes , Antígenos CD28 , Humanos , Antígenos CD28/metabolismo , Amigos , Linfocitos T , Antígeno CTLA-4/metabolismo , Inmunoterapia , Antígeno B7-1/metabolismo , Inmunoglobulinas/metabolismo , Butirofilinas/metabolismo , Antígenos CD/metabolismo
2.
Cancer Res ; 84(7): 965-976, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38266066

RESUMEN

Immune checkpoint inhibitors show remarkable responses in a wide range of cancers, yet patients develop adaptive resistance. This necessitates the identification of alternate therapies that synergize with immunotherapies. Epigenetic modifiers are potent mediators of tumor-intrinsic mechanisms and have been shown to regulate immune response genes, making them prime targets for therapeutic combinations with immune checkpoint inhibitors. Some success has been observed in early clinical studies that combined immunotherapy with agents targeting DNA methylation and histone modification; however, less is known about chromatin remodeler-targeted therapies. Here, we provide a discussion on the regulation of tumor immunogenicity by the chromatin remodeling SWI/SNF complex through multiple mechanisms associated with immunotherapy response that broadly include IFN signaling, DNA damage, mismatch repair, regulation of oncogenic programs, and polycomb-repressive complex antagonism. Context-dependent targeting of SWI/SNF subunits can elicit opportunities for synthetic lethality and reduce T-cell exhaustion. In summary, alongside the significance of SWI/SNF subunits in predicting immunotherapy outcomes, their ability to modulate the tumor immune landscape offers opportunities for therapeutic intervention.


Asunto(s)
Cromatina , Neoplasias , Humanos , Factores de Transcripción/metabolismo , Proteínas Cromosómicas no Histona/genética , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/genética , Neoplasias/terapia , Ensamble y Desensamble de Cromatina , Microambiente Tumoral
3.
Front Immunol ; 14: 1237715, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37771579

RESUMEN

CX3CL1 secreted in the tumor microenvironment serves as a chemoattractant playing a critical role in metastasis of CX3CR1 expressing cancer cells. CX3CR1 can be expressed in both cancer and immune-inhibitory myeloid cells to facilitate their migration. We generated a novel monoclonal antibody against mouse CX3CR1 that binds to CX3CR1 and blocks the CX3CL1-CX3CR1 interaction. We next explored the immune evasion strategies implemented by the CX3CL1-CX3CR1 axis and find that it initiates a resistance program in cancer cells that results in 1) facilitation of tumor cell migration, 2) secretion of soluble mediators to generate a pro-metastatic niche, 3) secretion of soluble mediators to attract myeloid populations, and 4) generation of tumor-inflammasome. The CX3CR1 monoclonal antibody reduces migration of tumor cells and decreases secretion of immune suppressive soluble mediators by tumor cells. In combination with anti-PD-1 immunotherapy, this CX3CR1 monoclonal antibody enhances survival in an immunocompetent mouse colon carcinoma model through a decrease in tumor-promoting myeloid populations. Thus, this axis is involved in the mechanisms of resistance to anti-PD-1 immunotherapy and the combination therapy can overcome a portion of the resistance mechanisms to anti-PD-1.


Asunto(s)
Anticuerpos Monoclonales , Escape del Tumor , Ratones , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Quimiocina CX3CL1/metabolismo
4.
Semin Immunol ; 52: 101480, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-34006473

RESUMEN

The PD-1 pathway is a cornerstone in immune regulation. While the PD-1 pathway has received considerable attention for its role in contributing to the maintenance of T cell exhaustion in chronic infection and cancer, the PD-1 pathway plays diverse roles in regulating host immunity beyond T cell exhaustion. Here, we discuss emerging concepts in the PD-1 pathway, including (1) the impact of PD-1 inhibitors on diverse T cell differentiation states including effector and memory T cell development during acute infection, as well as T cell exhaustion during chronic infection and cancer, (2) the role of PD-1 in regulating Treg cells, NK cells, and ILCs, and (3) the functions of PD-L1/B7-1 and PD-L2/RGMb/neogenin interactions. We then discuss the emerging use of neoadjuvant PD-1 blockade in the treatment of early-stage cancers and how the timing of PD-1 blockade may improve clinical outcomes. The diverse binding partners of PD-1 and its associated ligands, broad expression patterns of the receptors and ligands, differential impact of PD-1 modulation on cells depending on location and state of differentiation, and timing of PD-1 blockade add additional layers of complexity to the PD-1 pathway, and are important considerations for improving the efficacy and safety of PD-1 pathway therapeutics.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Humanos , Células Asesinas Naturales , Activación de Linfocitos , Neoplasias/terapia
5.
Commun Biol ; 3(1): 128, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184441

RESUMEN

Programmed cell death-1 (PD-1) inhibits T cell responses. This function relies on interaction with SHP-2. PD-1 has one immunoreceptor tyrosine-based inhibitory motif (ITIM) at Y223 and one immunoreceptor tyrosine-based switch motif (ITSM) at Y248. Only ITSM-Y248 is indispensable for PD-1-mediated inhibitory function but how SHP-2 enzymatic activation is mechanistically regulated by one PD-1 phosphotyrosine remains a puzzle. We found that after PD-1 phosphorylation, SHP-2 can bridge phosphorylated ITSM-Y248 residues on two PD-1 molecules via its amino terminal (N)-SH2 and carboxyterminal (C)-SH2 domains forming a PD-1: PD-1 dimer in live cells. The biophysical ability of SHP-2 to interact with two ITSM-pY248 residues was documented by isothermal titration calorimetry. SHP-2 interaction with two ITSM-pY248 phosphopeptides induced robust enzymatic activation. Our results unravel a mechanism of PD-1: SHP-2 interaction that depends only on ITSM-Y248 and explain how a single docking site within the PD-1 cytoplasmic tail can activate SHP-2 and PD-1-mediated inhibitory function.


Asunto(s)
Receptor de Muerte Celular Programada 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Linfocitos T/enzimología , Animales , Células COS , Chlorocebus aethiops , Activación Enzimática , Células HEK293 , Humanos , Motivo de Activación del Inmunorreceptor Basado en Tirosina , Células Jurkat , Ratones Noqueados , Fosforilación , Receptor de Muerte Celular Programada 1/química , Receptor de Muerte Celular Programada 1/genética , Unión Proteica , Multimerización de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 11/química , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Dominios Homologos src
6.
Cancer Immunol Immunother ; 68(3): 421-432, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30564891

RESUMEN

Targeting immune checkpoint pathways, such as programmed death ligand-1 (PD-L1, also known as CD274 or B7-H1) or its receptor programmed cell death-1 (PD-1) has shown improved survival for patients with numerous types of cancers, not limited to lung cancer, melanoma, renal cell carcinoma, and Hodgkin lymphoma. PD-L1 is a co-inhibitory molecule whose expression on the surface of tumor cells is associated with worse prognosis in many tumors. Here we describe a splice variant (secPD-L1) that does not splice into the transmembrane domain, but instead produces a secreted form of PD-L1 that has a unique 18 amino acid tail containing a cysteine that allows it to homodimerize and more effectively inhibit lymphocyte function than monomeric soluble PD-L1. We show that recombinant secPD-L1 can dimerize and inhibit T-cell proliferation and IFN-gamma production in vitro. The secPD-L1 variant is expressed by malignant cells in vitro that also express high levels of full-length PD-L1. Transcriptomic analysis of gene expression across The Cancer Genome Atlas found the strongest association of secPD-L1 with full-length PD-L1, but also with subsets of immunologic genes, such as in myeloid-derived suppressor cells. Moreover, the splice variant is also expressed in normal tissues and within normal peripheral blood cells it is preferentially expressed in activated myeloid cells. This is the first report of a form of secreted PD-L1 that homodimerizes and is functionally active. SecPD-L1 may function as a paracrine negative immune regulator within the tumor, since secPD-L1 does not require a cell-to-cell interaction to mediate its inhibitory effect.


Asunto(s)
Antígeno B7-H1/genética , Inmunosupresores/farmacología , Multimerización de Proteína , Empalme del ARN , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/química , Antígeno B7-H1/farmacología , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Células Supresoras de Origen Mieloide/fisiología , Placenta/metabolismo , Embarazo , Microambiente Tumoral
7.
Cancer Immunol Res ; 6(8): 921-929, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29871885

RESUMEN

Programmed death ligand 1 (PD-L1)-mediated immunosuppression regulates peripheral tolerance and is often co-opted by tumors to evade immune attack. PD-L1 binds to PD-1 but also binds to B7-1 (CD80) to regulate T-cell function. The binding interaction of PD-L1 with B7-1 and its functional role need further investigation to understand differences between PD-1 and PD-L1 tumor immunotherapy. We examined the molecular orientation of PD-L1 binding to B7-1 using cell-to-cell binding assays, ELISA, and flow cytometry. As expected, PD-L1-transfected cells bound to PD-1-transfected cells, and B7-1 cells bound to CD28 or CTLA-4-transfected cells; however, PD-L1 cells did not bind to B7-1 cells. By ELISA and flow cytometry with purified proteins, we found PD-L1 and B7-1 had a strong binding interaction only when PD-L1 was flexible. Soluble PD-1 and B7-1 competed for binding to PD-L1. Binding of native PD-L1 and B7-1 in cis on the same cell surface was demonstrated with NanoBiT proximity assays. Thus, PD-L1-B7-1 interaction can occur in cis on the same cell but not in trans between two cells, which suggests a model in which PD-L1 can bend via its 11-amino acid, flexible stalk to bind to B7-1 in cis, in a manner that can competitively block the binding of PD-L1 to PD-1 or of B7-1 to CD28. This binding orientation emphasizes the functional importance of coexpression of PD-L1 and B7-1 on the same cell. We found such coexpression on tumor-infiltrating myeloid cells. Our findings may help better utilize these pathways in cancer immunotherapy. Cancer Immunol Res; 6(8); 921-9. ©2018 AACR.


Asunto(s)
Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Animales , Sitios de Unión , Unión Competitiva/inmunología , Antígenos CD28/metabolismo , Antígeno CTLA-4/metabolismo , Neoplasias del Colon/inmunología , Ratones , Modelos Inmunológicos , Células Mieloides/inmunología , Receptor de Muerte Celular Programada 1/metabolismo
8.
Aquat Toxicol ; 138-139: 1-11, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23680676

RESUMEN

In the current study, two aspects concerning (i) the cytotoxicity potential of TiO2 nanoparticles (NPs) toward freshwater algal isolate Scenedesmus obliquus and (ii) the potential detoxification of NPs by the microalgae were assessed under light (UV-illumination) and dark conditions at low exposure levels (≤1 µg/mL), using sterile freshwater as the test medium. The statistically significant reduction in cell viability, increase in reactive oxygen species production and membrane permeability (light vs. dark) suggested photo-induced toxicity of TiO2 NPs. The electron micrographs demonstrated adsorption of the NPs onto the cell surface and substantiated their internalization/uptake. The fluorescence micrographs and the confocal laser scanning (CLSM) images suggested the absence of a definite/intact nucleus in the light treated cells pointing toward the probable genotoxic effects of NPs. In a separate three cycle experiment, a continuous decrease in the cytotoxicity was observed, whereas, at the end of each cycle only fresh algae were added to the supernatant containing NPs from the previous cycle. The decreasing concentrations of the NPs in the subsequent cycles owing to agglomeration-sedimentation processes exacerbated by the algal interactions played a crucial role in the detoxification. In addition, the exo-polymeric substances produced by the cells could have rendered the available NPs less reactive, thereby, enhancing the detoxification effects.


Asunto(s)
Contaminantes Ambientales/toxicidad , Nanopartículas del Metal/toxicidad , Scenedesmus/efectos de los fármacos , Titanio/toxicidad , Adsorción , Biodegradación Ambiental , Recuento de Células , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Agua Dulce , Nanopartículas del Metal/ultraestructura , Microscopía Electrónica de Rastreo , Tamaño de la Partícula , Especies Reactivas de Oxígeno/metabolismo , Scenedesmus/ultraestructura , Sales de Tetrazolio , Tiazoles , Pruebas de Toxicidad , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...