Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 351
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(6): 1037-1039, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32142652

RESUMEN

Tumor-infiltrating B cells are heterogeneous, and their roles in tumor immunity are controversial. In this issue of Cell, Lu and colleagues demonstrate that chemotherapy-induced complement signals promote the generation of ICOSL+B cells, which enhance tumor-specific T cell responses.


Asunto(s)
Antineoplásicos , Neoplasias , Linfocitos B , Humanos , Linfocitos T
2.
Nat Immunol ; 21(11): 1470, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32939095

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nat Immunol ; 21(5): 546-554, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32231300

RESUMEN

High-dose radiation activates caspases in tumor cells to produce abundant DNA fragments for DNA sensing in antigen-presenting cells, but the intrinsic DNA sensing in tumor cells after radiation is rather limited. Here we demonstrate that irradiated tumor cells hijack caspase 9 signaling to suppress intrinsic DNA sensing. Instead of apoptotic genomic DNA, tumor-derived mitochondrial DNA triggers intrinsic DNA sensing. Specifically, loss of mitochondrial DNA sensing in Casp9-/- tumors abolishes the enhanced therapeutic effect of radiation. We demonstrated that combining emricasan, a pan-caspase inhibitor, with radiation generates synergistic therapeutic effects. Moreover, loss of CASP9 signaling in tumor cells led to adaptive resistance by upregulating programmed death-ligand 1 (PD-L1) and resulted in tumor relapse. Additional anti-PD-L1 blockade can further overcome this acquired immune resistance. Therefore, combining radiation with a caspase inhibitor and anti-PD-L1 can effectively control tumors by sequentially blocking both intrinsic and extrinsic inhibitory signaling.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Caspasa 9/metabolismo , Inhibidores de Caspasas/uso terapéutico , Quimioradioterapia/métodos , Neoplasias Colorrectales/terapia , Ácidos Pentanoicos/uso terapéutico , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Caspasa 9/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Transducción de Señal , Regulación hacia Arriba
4.
Immunity ; 49(3): 490-503.e4, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30170810

RESUMEN

The NF-κB pathway plays a crucial role in supporting tumor initiation, progression, and radioresistance of tumor cells. However, the role of the NF-κB pathway in radiation-induced anti-tumor host immunity remains unclear. Here we demonstrated that inhibiting the canonical NF-κB pathway dampened the therapeutic effect of ionizing radiation (IR), whereas non-canonical NF-κB deficiency promoted IR-induced anti-tumor immunity. Mechanistic studies revealed that non-canonical NF-κB signaling in dendritic cells (DCs) was activated by the STING sensor-dependent DNA-sensing pathway. By suppressing recruitment of the transcription factor RelA onto the Ifnb promoter, activation of the non-canonical NF-κB pathway resulted in decreased type I IFN expression. Administration of a specific inhibitor of the non-canonical NF-κB pathway enhanced the anti-tumor effect of IR in murine models. These findings reveal the potentially interactive roles for canonical and non-canonical NF-κB pathways in IR-induced STING-IFN production and provide an alternative strategy to improve cancer radiotherapy.


Asunto(s)
Neoplasias del Colon/radioterapia , Células Dendríticas/inmunología , Melanoma/radioterapia , FN-kappa B/metabolismo , Neoplasias Experimentales/radioterapia , Radioterapia/métodos , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Neoplasias del Colon/inmunología , ADN/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular , Melanoma/inmunología , Melanoma Experimental , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Tolerancia a Radiación , Radiación Ionizante , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Immunity ; 47(2): 363-373.e5, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28801234

RESUMEN

Inhibition of cytosolic DNA sensing represents a strategy that tumor cells use for immune evasion, but the underlying mechanisms are unclear. Here we have shown that CD47-signal regulatory protein α (SIRPα) axis dictates the fate of ingested DNA in DCs for immune evasion. Although macrophages were more potent in uptaking tumor DNA, increase of DNA sensing by blocking the interaction of SIRPα with CD47 preferentially occurred in dendritic cells (DCs) but not in macrophages. Mechanistically, CD47 blockade enabled the activation of NADPH oxidase NOX2 in DCs, which in turn inhibited phagosomal acidification and reduced the degradation of tumor mitochondrial DNA (mtDNA) in DCs. mtDNA was recognized by cyclic-GMP-AMP synthase (cGAS) in the DC cytosol, contributing to type I interferon (IFN) production and antitumor adaptive immunity. Thus, our findings have demonstrated how tumor cells inhibit innate sensing in DCs and suggested that the CD47-SIRPα axis is critical for DC-driven antitumor immunity.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Neoplasias del Colon/inmunología , ADN Mitocondrial/inmunología , Células Dendríticas/inmunología , Proteínas de la Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Anticuerpos Bloqueadores/uso terapéutico , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/terapia , Reactividad Cruzada , Modelos Animales de Enfermedad , Humanos , Interferón Tipo I/metabolismo , Macrófagos/inmunología , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Escape del Tumor
6.
Proc Natl Acad Sci U S A ; 120(16): e2205085120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37036990

RESUMEN

PD-L1 has two receptors: PD-1 and CD80. Previous reports assumed that PD-L1 and CD80 interacted in trans, but recent reports showed that only cis PD-L1/CD80 interactions existed, and prevention of cis PD-L1/CD80 interactions on antigen-presenting cells (APCs) reduced antitumor immunity via augmenting PD-L1/PD-1 and CD80/CTLA4 interactions between T and APCs. Here, using tumor-bearing mice capable of cis and trans or trans only PD-L1/CD80 interactions, we show that trans PD-L1/CD80 interactions do exist between tumor and T cells, and the effects of trans PD-L1/CD80 interactions require tumor cell expression of MHC-I and T cell expression of CD28. The blockade of PD-L1/CD80 interactions in mice with both cis and trans interactions or with only trans interactions augments antitumor immunity by expanding IFN-γ-producing CD8+ T cells and IFN-γ-dependent NOS2-expressing tumor-associated macrophages. Our studies indicate that although cis and trans PD-L1/CD80 interactions may have opposite effects on antitumor immunity, the net effect of blocking PD-L1/CD80 interactions in vivo augments CD8+ T cell-mediated antitumor immunity.


Asunto(s)
Antígeno B7-H1 , Linfocitos T CD8-positivos , Ratones , Animales , Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Activación de Linfocitos , Antígeno B7-1 , Moléculas de Adhesión Celular
8.
Nat Immunol ; 14(10): 1014-22, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24048123

RESUMEN

Most tumor cells express antigens that can mediate recognition by host CD8(+) T cells. Cancers that are detected clinically must have evaded antitumor immune responses to grow progressively. Recent work has suggested two broad categories of tumor escape based on cellular and molecular characteristics of the tumor microenvironment. One major subset shows a T cell-inflamed phenotype consisting of infiltrating T cells, a broad chemokine profile and a type I interferon signature indicative of innate immune activation. These tumors appear to resist immune attack through the dominant inhibitory effects of immune system-suppressive pathways. The other major phenotype lacks this T cell-inflamed phenotype and appears to resist immune attack through immune system exclusion or ignorance. These two major phenotypes of tumor microenvironment may require distinct immunotherapeutic interventions for maximal therapeutic effect.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Activación de Linfocitos , Neoplasias/terapia , Células del Estroma/inmunología , Células del Estroma/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
9.
Proc Natl Acad Sci U S A ; 119(11): e2107339119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35254903

RESUMEN

SignificanceOutside the neurogenic niches, the adult brain lacks multipotent progenitor cells. In this study, we performed a series of in vivo screens and reveal that a single factor can induce resident brain astrocytes to become induced neural progenitor cells (iNPCs), which then generate neurons, astrocytes, and oligodendrocytes. Such a conclusion is supported by single-cell RNA sequencing and multiple lineage-tracing experiments. Our discovery of iNPCs is fundamentally important for regenerative medicine since neural injuries or degeneration often lead to loss/dysfunction of all three neural lineages. Our findings also provide insights into cell plasticity in the adult mammalian brain, which has largely lost the regenerative capacity.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Diferenciación Celular , Linaje de la Célula , Reprogramación Celular , Cuerpo Estriado/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Reprogramación Celular/genética , Cuerpo Estriado/metabolismo , Técnica del Anticuerpo Fluorescente , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Genes Reporteros , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , RNA-Seq , Receptores Notch/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Nat Immunol ; 13(10): 981-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22842344

RESUMEN

The mechanisms that regulate the T(H)9 subset of helper T cells and diseases mediated by T(H)9 cells remain poorly defined. Here we found that the costimulatory receptor OX40 was a powerful inducer of T(H)9 cells in vitro and T(H)9 cell-dependent airway inflammation in vivo. In polarizing conditions based on transforming growth factor-ß (TGF-ß), ligation of OX40 inhibited the production of induced regulatory T cells and the T(H)17 subset of helper T cells and diverted CD4(+)Foxp3(-) T cells to a T(H)9 phenotype. Mechanistically, OX40 activated the ubiquitin ligase TRAF6, which triggered induction of the kinase NIK in CD4(+) T cells and the noncanonical transcription factor NF-κB pathway; this subsequently led to the generation of T(H)9 cells. Thus, our study identifies a previously unknown mechanism for the induction of T(H)9 cells and may have important clinical implications in allergic inflammation.


Asunto(s)
Ligando OX40/metabolismo , Receptores OX40/metabolismo , Sistema Respiratorio/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antígenos CD4/biosíntesis , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-9/biosíntesis , Interleucina-9/metabolismo , Ratones , FN-kappa B/metabolismo , Ligando OX40/inmunología , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Receptores OX40/inmunología , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Factor 6 Asociado a Receptor de TNF/biosíntesis , Factor 6 Asociado a Receptor de TNF/metabolismo , Transactivadores/inmunología , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Quinasa de Factor Nuclear kappa B
11.
Nat Immunol ; 13(10): 947-53, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22922363

RESUMEN

Microbiota are essential for weight gain in mouse models of diet-induced obesity (DIO), but the pathways that cause the microbiota to induce weight gain are unknown. We report that mice deficient in lymphotoxin, a key molecule in gut immunity, were resistant to DIO. Ltbr(-/-) mice had different microbial community composition compared to their heterozygous littermates, including an overgrowth of segmented filamentous bacteria (SFB). Furthermore, cecal transplantation conferred leanness to germ-free recipients. Housing Ltbr(-/-) mice with their obese siblings rescued weight gain in Ltbr(-/-) mice, demonstrating the communicability of the obese phenotype. Ltbr(-/-) mice lacked interleukin 23 (IL-23) and IL-22, which can regulate SFB. Mice deficient in these pathways also resisted DIO, demonstrating that intact mucosal immunity guides diet-induced changes to the microbiota to enable obesity.


Asunto(s)
Inmunidad Mucosa , Receptor beta de Linfotoxina/fisiología , Linfotoxina-alfa/fisiología , Obesidad , Animales , Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Ciego/microbiología , Ciego/trasplante , Dieta , Metabolismo Energético , Vida Libre de Gérmenes , Interleucina-23/deficiencia , Interleucina-23/fisiología , Interleucinas/deficiencia , Interleucinas/fisiología , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/deficiencia , Linfotoxina-alfa/genética , Metagenoma , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/inmunología , Obesidad/metabolismo , Aumento de Peso/inmunología , Interleucina-22
12.
Immunity ; 42(4): 731-43, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902484

RESUMEN

Microbiota-mediated effects on the host immune response facilitate colonization resistance against pathogens. However, it is unclear whether and how the host immune response can regulate the microbiota to mediate colonization resistance. ID2, an essential transcriptional regulator for the development of innate lymphoid cell (ILC) progenitors, remains highly expressed in differentiated ILCs with unknown function. Using conditionally deficient mice in which ID2 is deleted from differentiated ILC3s, we observed that these mutant mice exhibited greatly impaired gut colonization resistance against Citrobacter rodentium. Utilizing gnotobiotic hosts, we showed that the ID2-dependent early colonization resistance was mediated by interleukin-22 (IL-22) regulation of the microbiota. In addition to regulating development, ID2 maintained homeostasis of ILC3s and controlled IL-22 production through an aryl hydrocarbon receptor (AhR) and IL-23 receptor pathway. Thus, ILC3s can mediate immune surveillance, which constantly maintains a proper microbiota, to facilitate early colonization resistance through an ID2-dependent regulation of IL-22.


Asunto(s)
Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/patología , Proteína 2 Inhibidora de la Diferenciación/inmunología , Interleucinas/inmunología , Linfocitos/patología , Receptores de Hidrocarburo de Aril/inmunología , Animales , Diferenciación Celular , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/microbiología , Regulación de la Expresión Génica , Vida Libre de Gérmenes/inmunología , Homeostasis/inmunología , Inmunidad Innata , Proteína 2 Inhibidora de la Diferenciación/deficiencia , Proteína 2 Inhibidora de la Diferenciación/genética , Interleucinas/genética , Linfocitos/inmunología , Linfocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/inmunología , Receptores de Hidrocarburo de Aril/genética , Receptores de Interleucina/genética , Receptores de Interleucina/inmunología , Transducción de Señal , Interleucina-22
13.
Immunity ; 42(4): 719-30, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902483

RESUMEN

Within secondary lymphoid tissues, stromal reticular cells support lymphocyte function, and targeting reticular cells is a potential strategy for controlling pathogenic lymphocytes in disease. However, the mechanisms that regulate reticular cell function are not well understood. Here we found that during an immune response in lymph nodes, dendritic cells (DCs) maintain reticular cell survival in multiple compartments. DC-derived lymphotoxin beta receptor (LTßR) ligands were critical mediators, and LTßR signaling on reticular cells mediated cell survival by modulating podoplanin (PDPN). PDPN modulated integrin-mediated cell adhesion, which maintained cell survival. This DC-stromal axis maintained lymphocyte survival and the ongoing immune response. Our findings provide insight into the functions of DCs, LTßR, and PDPN and delineate a DC-stromal axis that can potentially be targeted in autoimmune or lymphoproliferative diseases.


Asunto(s)
Células Dendríticas/citología , Ganglios Linfáticos/citología , Receptor beta de Linfotoxina/inmunología , Glicoproteínas de Membrana/inmunología , Células del Estroma/citología , Animales , Adhesión Celular , Supervivencia Celular/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica , Inmunofenotipificación , Ganglios Linfáticos/inmunología , Depleción Linfocítica , Receptor beta de Linfotoxina/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Células del Estroma/inmunología
14.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 174-181, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38650145

RESUMEN

Ovarian cancer is a prevalent malignancy in the female reproductive system, representing a significantly fatal and incurable tumor. Chelerythrine (CHE), a natural benzopyridine alkaloid, has demonstrated a broad spectrum of anticancer activities. Nevertheless, the ovarian cancer inhibitory impact of CHE remains unclear. In this study, we investigated the cytotoxic mechanism and potential targets of CHE on in vitro cultures of A2780 and SKOV3 cells derived from ovarian cancer. Additionally, in vivo experiments were conducted to confirm the suppressive impact of CHE on tumor growth in nude mice. The findings revealed that CHE impeded the growth of A2780 and SKOV3 cells in a concentration-time-dependent manner and significantly suppressed the development of tumors in nude mice. CHE elevated the level of oxidative stress in tumor cells, prompted cell cycle halt in the S phase, and increased their mitochondrial membrane potential. Western blotting results demonstrated that CHE could modulate the expression of proteins associated with apoptotic and ferroptosis processes in A2780 and SKOV3 cells. Nrf2 was verified to be an upstream key target mediating the inhibitory impact of CHE on ovarian cancer cells. In summary, CHE exerts its anti-cancer effects on ovarian cancer by modulating Nrf2, inhibiting cellular proliferation, and promoting apoptosis and ferroptosis.


Asunto(s)
Apoptosis , Benzofenantridinas , Proliferación Celular , Ferroptosis , Ratones Desnudos , Factor 2 Relacionado con NF-E2 , Neoplasias Ováricas , Femenino , Benzofenantridinas/farmacología , Humanos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Línea Celular Tumoral , Ferroptosis/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
15.
Nature ; 562(7728): 605-609, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30333625

RESUMEN

Immune checkpoint blockade therapy has been successful in treating some types of cancer but has not shown clinical benefits for treating leukaemia1. This result suggests that leukaemia uses unique mechanisms to evade this therapy. Certain immune inhibitory receptors that are expressed by normal immune cells are also present on leukaemia cells. Whether these receptors can initiate immune-related primary signalling in tumour cells remains unknown. Here we use mouse models and human cells to show that LILRB4, an immunoreceptor tyrosine-based inhibition motif-containing receptor and a marker of monocytic leukaemia, supports tumour cell infiltration into tissues and suppresses T cell activity via a signalling pathway that involves APOE, LILRB4, SHP-2, uPAR and ARG1 in acute myeloid leukaemia (AML) cells. Deletion of LILRB4 or the use of antibodies to block LILRB4 signalling impeded AML development. Thus, LILRB4 orchestrates tumour invasion pathways in monocytic leukaemia cells by creating an immunosuppressive microenvironment. LILRB4 represents a compelling target for the treatment of monocytic AML.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Escape del Tumor/inmunología , Animales , Apolipoproteínas E/metabolismo , Arginasa/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Tolerancia Inmunológica/inmunología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Masculino , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores Inmunológicos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Escape del Tumor/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Gut ; 72(8): 1544-1554, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36316098

RESUMEN

OBJECTIVE: The purpose of this study is to develop an anti-PDL1-based interferon (IFN) fusion protein to overcome the chronic hepatitis B virus (HBV)-induced immune tolerance, and combine this immunotherapy with a HBV vaccine to achieve the functional cure of chronic hepatitis B (CHB) infection. DESIGN: We designed an anti-PDL1-IFNα heterodimeric fusion protein, in which one arm was derived from anti-PDL1 antibody and the other arm was IFNα, to allow targeted delivery of IFNα into the liver by anti-PDL1 antibody. The effect of the anti-PDL1-IFNα heterodimer on overcoming hepatitis B surface antigen (HBsAg) vaccine resistance was evaluated in chronic HBV carrier mice. RESULTS: The anti-PDL1-IFNα heterodimer preferentially targeted the liver and resulted in viral suppression, the PD1/PDL1 immune checkpoint blockade and dendritic cell activation/antigen presentation to activate HBsAg-specific T cells, thus breaking immune tolerance in chronic HBV carrier mice. When an HBsAg vaccine was administered soon after anti-PDL1-IFNα heterodimer treatment, we observed strong anti-HBsAg antibody and HBsAg-specific T cell responses for efficient HBsAg clearance in chronic HBV carrier mice that received the combination treatment but not in those that received either single treatment. CONCLUSIONS: Targeting the liver with an engineered anti-PDL1-IFNα heterodimer can break HBV-induced immune tolerance to an HBsAg vaccine, offering a promising translatable therapeutic strategy for the functional cure of CHB.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B Crónica , Ratones , Animales , Virus de la Hepatitis B/genética , Hepatitis B Crónica/tratamiento farmacológico , Antígenos de Superficie de la Hepatitis B , Vacunas contra Hepatitis B , Interferón-alfa/uso terapéutico , Tolerancia Inmunológica
17.
Immunity ; 40(1): 25-39, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24412612

RESUMEN

Inhibitors of the transcription factor STAT3 target STAT3-dependent tumorigenesis but patients often develop diarrhea from unknown mechanisms. Here we showed that STAT3 deficiency increased morbidity and mortality after Citrobacter rodentium infection with decreased secretion of cytokines including IL-17 and IL-22 associated with the transcription factor RORγt. Administration of the cytokine IL-22 was sufficient to rescue STAT3-deficient mice from lethal infection. Although STAT3 was required for IL-22 production in both innate and adaptive arms, by using conditional gene-deficient mice, we observed that STAT3 expression in RORγt(+) innate lymphoid cells (ILC3s), but not T cells, was essential for the protection. However, STAT3 was required for RORγt expression in T helper cells, but not in ILC3s. Activated STAT3 could directly bind to the Il22 locus. Thus, cancer therapies that utilize STAT3 inhibitors increase the risk for pathogen-mediated diarrhea through direct suppression of IL-22 from gut ILCs.


Asunto(s)
Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Interleucinas/metabolismo , Intestinos/inmunología , Linfocitos/inmunología , Factor de Transcripción STAT3/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Células Cultivadas , Diarrea/metabolismo , Diarrea/prevención & control , Humanos , Inmunidad Innata , Indoles/administración & dosificación , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucinas/administración & dosificación , Interleucinas/genética , Intestinos/efectos de los fármacos , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Membrana Mucosa , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Pirroles/administración & dosificación , Factor de Transcripción STAT3/antagonistas & inhibidores , Sunitinib , Interleucina-22
18.
Immunity ; 41(5): 843-52, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25517616

RESUMEN

Ionizing radiation-mediated tumor regression depends on type I interferon (IFN) and the adaptive immune response, but several pathways control I IFN induction. Here, we demonstrate that adaptor protein STING, but not MyD88, is required for type I IFN-dependent antitumor effects of radiation. In dendritic cells (DCs), STING was required for IFN-? induction in response to irradiated-tumor cells. The cytosolic DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS) mediated sensing of irradiated-tumor cells in DCs. Moreover, STING was essential for radiation-induced adaptive immune responses, which relied on type I IFN signaling on DCs. Exogenous IFN-? treatment rescued the cross-priming by cGAS or STING-deficient DCs. Accordingly, activation of STING by a second messenger cGAMP administration enhanced antitumor immunity induced by radiation. Thus radiation-mediated antitumor immunity in immunogenic tumors requires a functional cytosolic DNA-sensing pathway and suggests that cGAMP treatment might provide a new strategy to improve radiotherapy.


Asunto(s)
ADN/inmunología , Proteínas de la Membrana/genética , Neoplasias/radioterapia , Nucleotidiltransferasas/inmunología , Inmunidad Adaptativa , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Antineoplásicos/farmacología , Células Cultivadas , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Interferón beta/biosíntesis , Interferón beta/inmunología , Interferón beta/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Neoplasias/inmunología , Nucleótidos Cíclicos/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Radiación Ionizante , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Xantonas/farmacología
19.
Gynecol Oncol ; 178: 80-88, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37820398

RESUMEN

OBJECTIVE: Inhibition of the MAPK pathway by MEK inhibitors (MEKi) is currently a therapeutic standard in several cancer types, including ovarian low-grade serous carcinoma (LGSC). A common MAPK pathway alteration in tubo-ovarian high-grade serous carcinoma (HGSC) is the genomic inactivation of neurofibromin 1 (NF1). The primary objectives of our study were to survey the prevalence of NF1 inactivation in the principal ovarian carcinoma histotype as well as to evaluate its associations with clinico-pathological parameters and key biomarkers including BRCA1/2 status in HGSC. METHODS: A recently commercialized NF1 antibody (clone NFC) was orthogonally validated on an automated immunohistochemistry (IHC) platform and IHC was performed on tissue microarrays containing 2140 ovarian carcinoma cases. Expression was interpreted as loss/inactivated (complete or subclonal) versus normal/retained. RESULTS: Loss of NF1 expression was detected in 250/1429 (17.4%) HGSC including 11% with subclonal loss. Survival of NF1-inactivated HGSC patients was intermediate between favorable BRCA1/2 mutated HGSC and unfavorable CCNE1 high-level amplified HGSC. NF1 inactivation was mutually exclusive with CCNE1 high-level amplifications, co-occurred with RB1 loss and occurred at similar frequencies in BRCA1/2 mutated versus wild-type HGSC. NF1 loss was found in 21/286 (7.3%) endometrioid carcinomas with a favorable prognostic association (p = 0.048), and in 4/64 (5.9%) LGSC, mutually exclusive with other driver events. CONCLUSIONS: NF1 inactivation occurs in a significant subset of BRCA1/2 wild-type HGSC and a subset of LGSC. While the functional effects of NF1 inactivation need to be further characterized, this signifies a potential therapeutic opportunity to explore targeting NF1 inactivation in these tumors.


Asunto(s)
Carcinoma Endometrioide , Cistadenocarcinoma Seroso , Neoplasias Ováricas , Femenino , Humanos , Proteína BRCA1 , Neurofibromina 1/genética , Inmunohistoquímica , Proteína BRCA2 , Neoplasias Ováricas/patología , Carcinoma Endometrioide/patología , Cistadenocarcinoma Seroso/patología , Carcinoma Epitelial de Ovario
20.
Gynecol Oncol ; 176: 162-172, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37556934

RESUMEN

OBJECTIVE: Dedifferentiated endometrial cancer (DDEC) is an uncommon and clinically highly aggressive subtype of endometrial cancer characterized by genomic inactivation of SWItch/Sucrose Non-Fermentable (SWI/SNF) complex protein. It responds poorly to conventional systemic treatment and its rapidly progressive clinical course limits the therapeutic windows to trial additional lines of therapies. This underscores a pressing need for biologically accurate preclinical tumor models to accelerate therapeutic development. METHODS: DDEC tumor from surgical samples were implanted into immunocompromised mice for patient-derived xenograft (PDX) and cell line development. The histologic, immunophenotypic, genetic and epigenetic features of the patient tumors and the established PDX models were characterized. The SMARCA4-deficienct DDEC model was evaluated for its sensitivity toward a KDM6A/B inhibitor (GSK-J4) that was previously reported to be effective therapy for other SMARCA4-deficient cancer types. RESULTS: All three DDEC models exhibited rapid growth in vitro and in vivo, with two PDX models showing spontaneous development of metastases in vivo. The PDX tumors maintained the same undifferentiated histology and immunophenotype, and exhibited identical genomic and methylation profiles as seen in the respective parental tumors, including a mismatch repair (MMR)-deficient DDEC with genomic inactivation of SMARCA4, and two MMR-deficient DDECs with genomic inactivation of both ARID1A and ARID1B. Although the SMARCA4-deficient cell line showed low micromolecular sensitivity to GSK-J4, no significant tumor growth inhibition was observed in the corresponding PDX model. CONCLUSIONS: These established patient tumor-derived models accurately depict DDEC and represent valuable preclinical tools to gain therapeutic insights into this aggressive tumor type.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Colorrectales , Neoplasias Endometriales , Femenino , Humanos , Animales , Ratones , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Diferenciación Celular , Biomarcadores de Tumor/genética , ADN Helicasas , Proteínas Nucleares/genética , Factores de Transcripción/genética , Proteínas de Unión al ADN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA