Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Immunity ; 41(5): 789-801, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25464855

RESUMEN

Microbial colonization of the gut induces the development of gut-associated lymphoid tissue (GALT). The molecular mechanisms that regulate GALT function and result in gut-commensal homeostasis are poorly defined. T follicular helper (Tfh) cells in Peyer's patches (PPs) promote high-affinity IgA responses. Here we found that the ATP-gated ionotropic P2X7 receptor controls Tfh cell numbers in PPs. Lack of P2X7 in Tfh cells enhanced germinal center reactions and high-affinity IgA secretion and binding to commensals. The ensuing depletion of mucosal bacteria resulted in reduced systemic translocation of microbial components, lowering B1 cell stimulation and serum IgM concentrations. Mice lacking P2X7 had increased susceptibility to polymicrobial sepsis, which was rescued by Tfh cell depletion or administration of purified IgM. Thus, regulation of Tfh cells by P2X7 activity is important for mucosal colonization, which in turn results in IgM serum concentrations necessary to protect the host from bacteremia.


Asunto(s)
Mucosa Intestinal/inmunología , Ganglios Linfáticos Agregados/inmunología , Receptores Purinérgicos P2X7/inmunología , Simbiosis/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Adenosina Trifosfato/metabolismo , Animales , Linfocitos B/inmunología , Bacteriemia/inmunología , Predisposición Genética a la Enfermedad , Centro Germinal/inmunología , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina M/sangre , Mucosa Intestinal/microbiología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/inmunología , Ganglios Linfáticos Agregados/citología , Receptores Purinérgicos P2X7/genética , Sepsis/inmunología , Sepsis/microbiología
2.
J Immunol ; 207(2): 493-504, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34215653

RESUMEN

The availability of Ags on the surface of tumor cells is crucial for the efficacy of cancer immunotherapeutic approaches using large molecules, such as T cell bispecific Abs (TCBs). Tumor Ags are processed through intracellular proteasomal protein degradation and are displayed as peptides on MHC class I (MHC I). Ag recognition through TCRs on the surface of CD8+ T cells can elicit a tumor-selective immune response. In this article, we show that proteolysis-targeting chimeras (PROTACs) that target bromo- and extraterminal domain proteins increase the abundance of the corresponding target-derived peptide Ags on MHC I in both liquid and solid tumor-derived human cell lines. This increase depends on the engagement of the E3 ligase to bromo- and extraterminal domain protein. Similarly, targeting of a doxycycline-inducible Wilms tumor 1 (WT1)-FKBP12F36V fusion protein, by a mutant-selective FKBP12F36V degrader, increases the presentation of WT1 Ags in human breast cancer cells. T cell-mediated response directed against cancer cells was tested on treatment with a TCR-like TCB, which was able to bridge human T cells to a WT1 peptide displayed on MHC I. FKBP12F36V degrader treatment increased the expression of early and late activation markers (CD69, CD25) in T cells; the secretion of granzyme ß, IFN-γ, and TNF-α; and cancer cell killing in a tumor-T cell coculture model. This study supports harnessing targeted protein degradation in tumor cells, for modulation of T cell effector function, by investigating for the first time, to our knowledge, the potential of combining a degrader and a TCB in a cancer immunotherapy setting.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Quimera/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/inmunología , Línea Celular Tumoral , Epítopos de Linfocito T/inmunología , Humanos , Proteolisis , Receptores de Antígenos de Linfocitos T/inmunología
3.
Front Immunol ; 14: 1297175, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022587

RESUMEN

Following the success of cancer immunotherapy using large molecules against immune checkpoint inhibitors, the concept of using small molecules to interfere with intracellular negative regulators of anti-tumor immune responses has emerged in recent years. The main targets for small molecule drugs currently include enzymes of negative feedback loops in signaling pathways of immune cells and proteins that promote immunosuppressive signals within the tumor microenvironment. In the adaptive immune system, negative regulators of T cell receptor signaling (MAP4K1, DGKα/ζ, CBL-B, PTPN2, PTPN22, SHP1), co-receptor signaling (CBL-B) and cytokine signaling (PTPN2) have been preclinically validated as promising targets and initial clinical trials with small molecule inhibitors are underway. To enhance innate anti-tumor immune responses, inhibitory immunomodulation of cGAS/STING has been in the focus, and inhibitors of ENPP1 and TREX1 have reached the clinic. In addition, immunosuppressive signals via adenosine can be counteracted by CD39 and CD73 inhibition, while suppression via intratumoral immunosuppressive prostaglandin E can be targeted by EP2/EP4 antagonists. Here, we present the status of the most promising small molecule drug candidates for cancer immunotherapy, all residing relatively early in development, and the potential of relevant biomarkers.


Asunto(s)
Neoplasias , Proteína Tirosina Fosfatasa no Receptora Tipo 2 , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Inmunomodulación , Biomarcadores , Microambiente Tumoral , Proteína Tirosina Fosfatasa no Receptora Tipo 22
4.
Cancer Res ; 82(14): 2552-2564, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35584009

RESUMEN

The therapeutic benefit of approved BRAF and MEK inhibitors (BRAFi/MEKi) in patients with brain metastatic BRAF V600E/K-mutated melanoma is limited and transient. Resistance largely occurs through the restoration of MAPK signaling via paradoxical BRAF activation, highlighting the need for more effective therapeutic options. Aiming to address this clinical challenge, we characterized the activity of a potent, brain-penetrant paradox breaker BRAFi (compound 1a, C1a) as first-line therapy and following progression upon treatment with approved BRAFi and BRAFi/MEKi therapies. C1a activity was evaluated in vitro and in vivo in melanoma cell lines and patient-derived models of BRAF V600E-mutant melanoma brain metastases following relapse after treatment with BRAFi/MEKi. C1a showed superior efficacy compared with approved BRAFi in both subcutaneous and brain metastatic models. Importantly, C1a manifested potent and prolonged antitumor activity even in models that progressed on BRAFi/MEKi treatment. Analysis of mechanisms of resistance to C1a revealed MAPK reactivation under drug treatment as the predominant resistance-driving event in both subcutaneous and intracranial tumors. Specifically, BRAF kinase domain duplication was identified as a frequently occurring driver of resistance to C1a. Combination therapies of C1a and anti-PD-1 antibody proved to significantly reduce disease recurrence. Collectively, these preclinical studies validate the outstanding antitumor activity of C1a in brain metastasis, support clinical investigation of this agent in patients pretreated with BRAFi/MEKi, unveil genetic drivers of tumor escape from C1a, and identify a combinatorial treatment that achieves long-lasting responses. SIGNIFICANCE: A brain-penetrant BRAF inhibitor demonstrates potent activity in brain metastatic melanoma, even upon relapse following standard BRAF inhibitor therapy, supporting further investigation into its clinical utility.


Asunto(s)
Neoplasias Encefálicas , Melanoma , Encéfalo/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos , Mutación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf
5.
Cancer Res ; 80(18): 3906-3919, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32699136

RESUMEN

Extracellular ATP (eATP) is a signaling molecule that variably affects all cells of the immune system either directly or after hydrolysis to adenosine. Although eATP is virtually absent in the interstitium of normal tissues, it can be present in the hundreds of micromolar range in tumors, a concentration compatible with activation of the ATP-gated ionotropic P2X7 receptor. Here, we show that P2X7 activity in tumor-infiltrating lymphocytes (TIL) induces cellular senescence and limits tumor suppression. P2X7 stimulation affected cell cycling of effector T cells and resulted in generation of mitochondrial reactive oxygen species and p38 MAPK-dependent upregulation of cyclin-dependent kinase inhibitor 1A (Cdkn1a, encoding for p21Waf1/Cip1). Lack of P2X7 promoted a transcriptional signature that correlated with enhanced cytotoxic T-cell response in human solid tumors. In mice, transfer of tumor-specific T cells with deletion of P2rx7 significantly reduced tumor growth and extended survival. Collectively, these findings uncover a purinergic checkpoint that can be targeted to improve the efficacy of cancer immunotherapy strategies. SIGNIFICANCE: These findings suggest that the purinergic checkpoint P2X7 may be targeted to enhance T-cell-mediated cancer immunotherapy and improve T effector cell accumulation in the tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/18/3906/F1.large.jpg.


Asunto(s)
Inhibición de Migración Celular , Senescencia Celular/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/inmunología , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Ciclo Celular , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citometría de Flujo/métodos , Perfilación de la Expresión Génica , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Trasplante de Neoplasias , Antagonistas del Receptor Purinérgico P2X , Especies Reactivas de Oxígeno/metabolismo , Receptores Purinérgicos P2X7/deficiencia , Linfocitos T Citotóxicos/inmunología , Transcripción Genética , Microambiente Tumoral/inmunología , Regulación hacia Arriba
6.
J Exp Med ; 216(2): 317-336, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30655308

RESUMEN

Altered control of T follicular helper (Tfh) cells can lead to generation of autoantibodies and autoimmune manifestations. Signaling pathways that selectively limit pathogenic responses without affecting the protective function of Tfh cells are unknown. Here we show that the ATP-gated ionotropic P2X7 receptor restricts the expansion of aberrant Tfh cells and the generation of self-reactive antibodies in experimental murine lupus, but its activity is dispensable for the expansion of antigen-specific Tfh cells during vaccination. P2X7 stimulation promotes caspase-mediated pyroptosis of Tfh cells and controls the development of pathogenic ICOS+ IFN-γ-secreting cells. Circulating Tfh cells from patients with systemic lupus erythematosus (SLE) but not primary antiphospholipid syndrome (PAPS), a nonlupus systemic autoimmune disease, were hyporesponsive to P2X7 stimulation and resistant to P2X7-mediated inhibition of cytokine-driven expansion. These data point to the P2X7 receptor as a checkpoint regulator of Tfh cells; thus, restoring P2X7 activity in SLE patients could selectively limit the progressive amplification of pathogenic autoantibodies, which deteriorate patients' conditions.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Receptores Purinérgicos P2X7/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Modelos Animales de Enfermedad , Interferón gamma/genética , Interferón gamma/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Noqueados , Piroptosis/genética , Piroptosis/inmunología , Receptores Purinérgicos P2X7/genética , Linfocitos T Colaboradores-Inductores/patología
7.
Nat Commun ; 8(1): 1917, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29203869

RESUMEN

The melastatin-like transient-receptor-potential-7 protein (TRPM7), harbouring a cation channel and a serine/threonine kinase, has been implicated in thymopoiesis and cytokine expression. Here we show, by analysing TRPM7 kinase-dead mutant (Trpm7 R/R ) mice, that the enzymatic activity of the receptor is not essential for thymopoiesis, but is required for CD103 transcription and gut-homing of intra-epithelial lymphocytes. Defective T cell gut colonization reduces MHCII expression in intestinal epithelial cells. Mechanistically, TRPM7 kinase activity controls TGF-ß-induced CD103 expression and pro-inflammatory T helper 17, but not regulatory T, cell differentiation by modulating SMAD2. Notably, we find that the TRPM7 kinase activity promotes gut colonization by alloreactive T cells in acute graft-versus-host disease. Thus, our results unravel a function of TRPM7 kinase in T cell activity and suggest a therapeutic potential of kinase inhibitors in averting acute graft-versus-host disease.


Asunto(s)
Enfermedad Injerto contra Huésped/genética , Intestinos/inmunología , Linfopoyesis/genética , Linfocitos T Reguladores/citología , Canales Catiónicos TRPM/genética , Células Th17/citología , Animales , Antígenos CD/inmunología , Diferenciación Celular/genética , Genes MHC Clase II/genética , Genes MHC Clase II/inmunología , Enfermedad Injerto contra Huésped/inmunología , Cadenas alfa de Integrinas/inmunología , Ratones , Mutación , Proteína Smad2/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Canales Catiónicos TRPM/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta/inmunología
8.
Oncotarget ; 7(5): 5143-56, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26784253

RESUMEN

Astrocytes play a crucial role in neuroinflammation as part of the glia limitans, which regulates infiltration of the brain parenchyma by leukocytes. The signaling pathways and molecular events, which result from the interaction of activated T cells with astrocytes are poorly defined. Here we show that astrocytes promote the expression and enzymatic activity of CD39 and CD73 ectonucleotidases in recently activated CD4 cells by a contact dependent mechanism that is independent of T cell receptor interaction with class II major histocompatibility complex (MHC). Transforming growth factor-ß (TGF-ß) is robustly upregulated and sufficient to promote ectonucleotidases expression. T cell adhesion to astrocyte results in differentiation to an immunosuppressive phenotype defined by expression of the transcription factor Rorγt, which characterizes the CD4 T helper 17 subset. CD39 activity in T cells in turn inhibits spontaneous calcium oscillations in astrocytes that correlated with enhanced and reduced transcription of CCL2 chemokine and Sonic hedgehog (Shh), respectively. We hypothesize this TCR-independent interaction promote an immunosuppressive program in T cells to control possible brain injury by deregulated T cell activation during neuroinflammation. On the other hand, the increased secretion of CCL2 with concomitant reduction of Shh might promote leukocytes extravasation into the brain parenchyma.


Asunto(s)
5'-Nucleotidasa/metabolismo , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Astrocitos , Diferenciación Celular , Humanos , Transducción de Señal , Regulación hacia Arriba
9.
J Leukoc Biol ; 99(6): 1077-87, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26931577

RESUMEN

Infiltration of the central nervous system is a severe trait of T cell acute lymphoblastic leukemia. Inhibition of CXC chemokine receptor 4 significantly ameliorates T cell acute lymphoblastic leukemia in murine models of the disease; however, signaling by CXC chemokine receptor 4 is important in limiting the divagation of peripheral blood mononuclear cells out of the perivascular space into the central nervous system parenchyma. Therefore, Inhibition of CXC chemokine receptor 4 potentially may untangle T cell acute lymphoblastic leukemia cells from retention outside the brain. Here, we show that leukemic lymphoblasts massively infiltrate cranial bone marrow, with diffusion to the meninges without invasion of the brain parenchyma, in mice that underwent xenotransplantation with human T cell acute lymphoblastic leukemia cells or that developed leukemia from transformed hematopoietic progenitors. We tested the hypothesis that T cell acute lymphoblastic leukemia neuropathology results from meningeal infiltration through CXC chemokine receptor 4-mediated bone marrow colonization. Inhibition of leukemia engraftment in the bone marrow by pharmacologic CXC chemokine receptor 4 antagonism significantly ameliorated neuropathologic aspects of the disease. Genetic deletion of CXCR4 in murine hematopoietic progenitors abrogated leukemogenesis induced by constitutively active Notch1, whereas lack of CCR6 and CCR7, which have been shown to be involved in T cell and leukemia extravasation into the central nervous system, respectively, did not influence T cell acute lymphoblastic leukemia development. We hypothesize that lymphoblastic meningeal infiltration as a result of bone marrow colonization is responsible for the degenerative alterations of the neuroparenchyma as well as the alteration of cerebrospinal fluid drainage in T cell acute lymphoblastic leukemia xenografts. Therefore, CXC chemokine receptor 4 may constitute a pharmacologic target for T cell acute lymphoblastic leukemia neuropathology.


Asunto(s)
Médula Ósea/patología , Sistema Nervioso Central/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Receptores CXCR4/metabolismo , Adolescente , Animales , Bencilaminas , Médula Ósea/efectos de los fármacos , Línea Celular Transformada , Línea Celular Tumoral , Sistema Nervioso Central/efectos de los fármacos , Niño , Preescolar , Ciclamas , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/farmacología , Humanos , Hígado/citología , Hígado/embriología , Masculino , Meninges/efectos de los fármacos , Meninges/patología , Ratones , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores de Quimiocina/metabolismo , Receptores Notch/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA