Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Immunity ; 56(5): 1013-1026.e6, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36944334

RESUMEN

Sepsis is a dysregulated inflammatory consequence of systemic infection. As a result, excessive platelet activation leads to thrombosis and coagulopathy, but we currently lack sufficient understanding of these processes. Here, using the cecal ligation and puncture (CLP) model of sepsis, we observed septic thrombosis and neutrophil extracellular trap formation (NETosis) within the mouse vasculature by intravital microscopy. STING activation in platelets was a critical driver of sepsis-induced pathology. Platelet-specific STING deficiency suppressed platelet activation and granule secretion, which alleviated sepsis-induced intravascular thrombosis and NETosis in mice. Mechanistically, sepsis-derived cGAMP promoted the binding of STING to STXBP2, the assembly of SNARE complex, granule secretion, and subsequent septic thrombosis, which probably depended on the palmitoylation of STING. We generated a peptide, C-ST5, to block STING binding to STXBP2. Septic mice treated with C-ST5 showed reduced thrombosis. Overall, platelet activation via STING reveals a potential strategy for limiting life-threatening sepsis-mediated coagulopathy.


Asunto(s)
Trampas Extracelulares , Sepsis , Trombosis , Animales , Ratones , Plaquetas/metabolismo , Trampas Extracelulares/metabolismo , Ratones Endogámicos C57BL , Proteínas Munc18/metabolismo , Activación Plaquetaria , Sepsis/metabolismo , Trombosis/metabolismo
2.
Immunity ; 55(7): 1268-1283.e9, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35700739

RESUMEN

The incidence and mortality rates of many non-reproductive human cancers are generally higher in males than in females. However, the immunological mechanism underlying sexual differences in cancers remains elusive. Here, we demonstrated that sex-related differences in tumor burden depended on adaptive immunity. Male CD8+ T cells exhibited impaired effector and stem cell-like properties compared with female CD8+ T cells. Mechanistically, androgen receptor inhibited the activity and stemness of male tumor-infiltrating CD8+ T cells by regulating epigenetic and transcriptional differentiation programs. Castration combined with anti-PD-L1 treatment synergistically restricted tumor growth in male mice. In humans, fewer male CD8+ T cells maintained a stem cell-like memory state compared with female counterparts. Moreover, AR expression correlated with tumor-infiltrating CD8+ T cell exhaustion in cancer patients. Our findings reveal sex-biased CD8+ T cell stemness programs in cancer progression and in the responses to cancer immunotherapy, providing insights into the development of sex-based immunotherapeutic strategies for cancer treatment.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Animales , Femenino , Humanos , Inmunoterapia , Masculino , Ratones , Neoplasias/terapia , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Caracteres Sexuales , Microambiente Tumoral
3.
Mol Cell ; 81(5): 940-952.e5, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33434504

RESUMEN

STING-dependent cytosolic DNA sensing in dendritic cells (DCs) initiates antitumor immune responses, but how STING signaling is metabolically regulated in the tumor microenvironment remains unknown. Here, we show that oxidative stress is required for STING-induced DC antitumor function through a process that directs SUMO-specific protease 3 (SENP3) activity. DC-specific deletion of Senp3 drives tumor progression by blunting STING-dependent type-I interferon (IFN) signaling in DCs and dampening antitumor immune responses. DC-derived reactive oxygen species (ROS) trigger SENP3 accumulation and the SENP3-IFI204 interaction, thereby catalyzing IFI204 deSUMOylation and boosting STING signaling activation in mice. Consistently, SENP3 senses ROS to facilitate STING-dependent DC activity in tissue samples from colorectal cancer patients. Our results reveal that oxidative stress as a metabolic regulator promotes STING-mediated DC antitumor immune responses and highlights SENP3 as an overflow valve for STING signaling induction in the metabolically abnormal tumor microenvironment.


Asunto(s)
Neoplasias Colorrectales/genética , Cisteína Endopeptidasas/genética , Células Dendríticas/inmunología , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Aloinjertos , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Cisteína Endopeptidasas/inmunología , Células Dendríticas/patología , Femenino , Células HEK293 , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/inmunología , Estrés Oxidativo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Análisis de Supervivencia , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
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
7.
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
8.
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
9.
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
10.
J Immunol ; 190(11): 5874-81, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23630355

RESUMEN

Local failures following radiation therapy are multifactorial, and the contributions of the tumor and the host are complex. Current models of tumor equilibrium suggest that a balance exists between cell birth and cell death due to insufficient angiogenesis, immune effects, or intrinsic cellular factors. We investigated whether host immune responses contribute to radiation-induced tumor equilibrium in animal models. We report an essential role for immune cells and their cytokines in suppressing tumor cell regrowth in two experimental animal model systems. Depletion of T cells or neutralization of IFN-γ reversed radiation-induced equilibrium, leading to tumor regrowth. We also demonstrate that PD-L1 blockade augments T cell responses, leading to rejection of tumors in radiation-induced equilibrium. We identify an active interplay between tumor cells and immune cells that occurs in radiation-induced tumor equilibrium and suggest a potential role for disruption of the PD-L1/PD-1 axis in increasing local tumor control.


Asunto(s)
Citotoxicidad Inmunológica , Neoplasias/inmunología , Linfocitos T/inmunología , Animales , Antígeno B7-H1 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Inmunoterapia , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias/patología , Neoplasias/cirugía , Receptor de Muerte Celular Programada 1 , Radiocirugia , Carga Tumoral/inmunología , Carga Tumoral/efectos de la radiación
11.
Trends Mol Med ; 30(3): 202-203, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38302316

RESUMEN

Ma et al. recently reported a systematic screening of small-molecule compounds targeting the FG-CC' cleft of T cell immunoglobulin and mucin-containing molecule 3 (TIM-3). They identified a functional Tim-3 inhibitor, ML-T7, that, as a single agent or in combination with anti-PD-1, demonstrated strong antitumor activity in preclinical mouse tumor models, supporting its potential for further clinical translation.


Asunto(s)
Receptor 2 Celular del Virus de la Hepatitis A , Neoplasias , Animales , Humanos , Ratones , Inmunoterapia , Neoplasias/tratamiento farmacológico
12.
Cancer Biol Med ; 20(12)2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38318930

RESUMEN

Maintenance immunotherapy after concurrent chemoradiotherapy remains the standard therapeutic approach in patients with unresectable locally advanced non-small cell lung cancer (LA-NSCLC). The efficacy of pembrolizumab without chemotherapy in stage IV NSCLC has incited interest in similar approaches for LA-NSCLC. Several recent investigations involving the synergistic potential of immunotherapy combined with radiotherapy (iRT) have generated encouraging results. This review discusses the existing studies and prospective directions of chemotherapy-free iRT strategies in unresectable LA-NSCLC. Although the initial findings of chemotherapy-free iRT strategies have shown promising efficacy, we must consider the methodologic limitations of current studies and the myriad of challenges that accompany the implementation of chemotherapy-free iRT. These challenges include determining the optimal dose and fractionation, precise target volume delineation, and identification of additional suitable patient cohorts. Furthermore, the feasibility of chemotherapy-free iRT as a novel treatment modality for select patients with LA-NSCLC is contingent upon validation through randomized phase III trials.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Estudios Prospectivos , Quimioradioterapia/métodos
13.
Cell Rep ; 43(2): 113712, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38294903

RESUMEN

Adoptive cell therapies are emerging forms of immunotherapy that reprogram T cells for enhanced antitumor responses. Although surface programmed cell death-ligand 1 (PD-L1)/programmed cell death protein 1 (PD-1) engagement inhibits antitumor immunity, the role of cell-intrinsic PD-L1 in adoptive T cell therapy remains unknown. Here, we found that intracellular PD-L1 was enriched in tumor-infiltrating CD8+ T cells of cancer patients. PD-L1 ablation promoted antitumor immune responses and the maintenance of an effector-like state of therapeutic CD8+ T cells, while blockade of surface PD-L1 was unable to impact on their expansion and function. Moreover, cell-intrinsic PD-L1 impeded CD8+ T cell activity, which partially relied on mTORC1 signaling. Furthermore, endogenous tumor-reactive CD8+ T cells were motivated by BATF3-driven dendritic cells after adoptive transfer of PD-L1-deficient therapeutic CD8+ T cells. This role of cell-intrinsic PD-L1 in therapeutic CD8+ T cell dysfunction highlights that disrupting cell-intrinsic PD-L1 in CD8+ T cells represents a viable approach to improving T cell-based cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Antígeno B7-H1 , Inmunoterapia , Tratamiento Basado en Trasplante de Células y Tejidos , Proteínas de la Membrana , Neoplasias/terapia
14.
Adv Sci (Weinh) ; 11(32): e2400149, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38898748

RESUMEN

The activation of cyclic GMP-AMP (cGAMP) synthase (cGAS) and its adaptor, stimulator of interferon genes (STING), is known to reprogram the immunosuppressive tumor microenvironment for promoting antitumor immunity. To enhance the efficiency of cGAS-STING pathway activation, macrophage-selective uptake, and programmable cytosolic release are crucial for the delivery of STING agonists. However, existing polymer- or lipid-based delivery systems encounter difficulty in integrating multiple functions meanwhile maintaining precise control and simple procedures. Herein, inspired by cGAS being a natural DNA sensor, a modularized DNA nanodevice agonist (DNDA) is designed that enable macrophage-selective uptake and programmable activation of the cGAS-STING pathway through precise self-assembly. The resulting DNA nanodevice acts as both a nanocarrier and agonist. Upon local administration, it demonstrates the ability of macrophage-selective uptake, endosomal escape, and cytosolic release of the cGAS-recognizing DNA segment, leading to robust activation of the cGAS-STING pathway and enhanced antitumor efficacy. Moreover, DNDA elicits a synergistic therapeutic effect when combined with immune checkpoint blockade. The study broadens the application of DNA nanotechnology as an immune stimulator for cGAS-STING activation.


Asunto(s)
ADN , Inmunoterapia , Macrófagos , Proteínas de la Membrana , Nucleotidiltransferasas , Animales , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Ratones , Inmunoterapia/métodos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , ADN/inmunología , Nucleotidiltransferasas/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Humanos , Modelos Animales de Enfermedad , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico
15.
Cell Rep ; 42(9): 113108, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37708030

RESUMEN

Natural killer (NK) cells are cytotoxic innate lymphocytes that eradicate tumor cells. Inducing durable antitumor immune responses by NK cells represents a major priority of cancer immunotherapy. While cytosolic DNA sensing plays an essential role in initiating antitumor immunity, the role of NK cell-intrinsic STING signaling remains unclear. Here, we find that NK cell-intrinsic STING promotes antitumor responses and maintains a reservoir of TCF-1+ NK cells. In contrast, tumor cell-intrinsic cGAS and mtDNA are required for NK cell antitumor activity, indicating that tumor mtDNA recognition by cGAS partially triggers NK cell-intrinsic STING activation. Moreover, addition of cGAMP enables STING activation and type I interferon production in NK cells, thereby supporting the activation of NK cells in vitro. In humans, STING agonism promotes the expansion of TCF-1+ NK cells. This study provides insight into understanding how STING signaling drives NK cell antitumor immunity and the development of NK cell-based cancer immunotherapy.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Neoplasias/patología , Antineoplásicos/farmacología , Nucleotidiltransferasas/metabolismo , ADN Mitocondrial , Células Asesinas Naturales/metabolismo , Inmunidad Innata
16.
Sci Immunol ; 8(90): eadf4919, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38039379

RESUMEN

Mitochondrial antiviral signaling protein (MAVS)-mediated cytosolic RNA sensing plays a central role in tumor immunogenicity. However, the effects of host MAVS signaling on antitumor immunity remain unclear. Here, we demonstrate that the host MAVS pathway supports tumor growth and impairs antitumor immunity, whereas MAVS deficiency in dendritic cells (DCs) promotes tumor-reactive CD8+ T cell responses. Specifically, CD8+ T cell priming capacity was enhanced by MAVS ablation in a type I interferon-independent, but IL-12-dependent, manner. Mechanistically, loss of the RIG-I/MAVS cascade activated the noncanonical NF-κB pathway and in turn induced IL-12 production by DCs. MAVS-restrained IL-12 promoted cross-talk between CD8+ T cells and DCs, which was licensed by IFN-γ. Moreover, ablation of host MAVS sensitized tumors to immunotherapy and attenuated radiation resistance, thereby facilitating the maintenance of effector CD8+ T cells. These findings demonstrate that the host MAVS pathway acts as an immune regulator of DC-driven antitumor immunity and support the development of immunotherapies that antagonize MAVS signaling in DCs.


Asunto(s)
Interleucina-12 , Neoplasias , Humanos , Linfocitos T CD8-positivos , Transducción de Señal , Células Dendríticas
17.
Chem Sci ; 14(22): 5956-5964, 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37293644

RESUMEN

Controllable activation of the innate immune adapter protein - stimulator of interferon genes (STING) pathway is a critical challenge for the clinical development of STING agonists due to the potential "on-target off-tumor" toxicity caused by systematic activation of STING. Herein, we designed and synthesized a photo-caged STING agonist 2 with a tumor cell-targeting carbonic anhydrase inhibitor warhead, which could be readily uncaged by blue light to release the active STING agonist leading to remarkable activation of STING signaling. Furthermore, compound 2 was found to preferentially target tumor cells, stimulate the STING signaling in zebrafish embryo upon photo-uncaging and to induce proliferation of macrophages and upregulation of the mRNA expression of STING as well as its downstream NF-kB and cytokines, thus leading to significant suppression of tumor cell growth in a photo-dependent manner with reduced systemic toxicity. This photo-caged agonist not only provides a powerful tool to precisely trigger STING signalling, but also represents a novel controllable STING activation strategy for safer cancer immunotherapy.

18.
Cell Death Dis ; 13(7): 640, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35869062

RESUMEN

Our previous studies show that the mitotic phosphorylation of SUMO-specific protease 3 (SENP3) can inhibit its de-SUMOylation activity in G2/M phase of the cell cycle. Inhibition of SENP3 plays a critical role in the correct separation of sister chromatids in mitosis. The mutation of mitotic SENP3 phosphorylation causes chromosome instability and promotes tumorigenesis. In this study, we find that the mutation of mitotic SENP3 phosphorylation in tumor cells can suppress tumor growth in immune-competent mouse model. We further detect an increase of CD8+ T cell infiltration in the tumors, which is essential for the anti-tumor effect in immune-competent mouse model. Moreover, we find that mitotic SENP3 activation increases micronuclei formation, which can activate cGAS signaling-dependent innate immune response. We confirmed that cGAS signaling mediates the mitotic SENP3 activation-induced anti-tumor immunity. We further show that p53 responding to DNA damage activates mitotic SENP3 by inhibiting phosphorylation, and further increases cellular senescence as well as the related innate immune response in tumor cells. Furthermore, TCGA database demonstrates that the SENP3 expression positively correlates with the induction of innate immune response as well as the survival of the p53 mutant pancreatic cancer patients. Together, these data reveal that mitotic SENP3 activation in tumor cells can promote host anti-tumor immune response by coupling with cGAS signaling.


Asunto(s)
Neoplasias , Péptido Hidrolasas , Animales , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Ratones , Neoplasias/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Péptido Hidrolasas/metabolismo , Sumoilación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
19.
J Cell Biol ; 221(7)2022 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-35510944

RESUMEN

The stimulator of interferon genes (STING) plays a critical role in innate immunity. Emerging evidence suggests that STING is important for DNA or cGAMP-induced non-canonical autophagy, which is independent of a large part of canonical autophagy machineries. Here, we report that, in the absence of STING, energy stress-induced autophagy is upregulated rather than downregulated. Depletion of STING in Drosophila fat cells enhances basal- and starvation-induced autophagic flux. During acute exercise, STING knockout mice show increased autophagy flux, exercise endurance, and altered glucose metabolism. Mechanistically, these observations could be explained by the STING-STX17 interaction. STING physically interacts with STX17, a SNARE that is essential for autophagosome biogenesis and autophagosome-lysosome fusion. Energy crisis and TBK1-mediated phosphorylation both disrupt the STING-STX17 interaction, allow different pools of STX17 to translocate to phagophores and mature autophagosomes, and promote autophagic flux. Taken together, we demonstrate a heretofore unexpected function of STING in energy stress-induced autophagy through spatial regulation of autophagic SNARE STX17.


Asunto(s)
Autofagia , Metabolismo Energético , Lisosomas , Proteínas de la Membrana , Proteínas Qa-SNARE , Animales , Autofagosomas/metabolismo , Drosophila , Lisosomas/genética , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Condicionamiento Físico Animal , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo
20.
Cancer Lett ; 502: 166-179, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33450361

RESUMEN

The brain is one of the most common metastatic sites in non-small cell lung cancer (NSCLC), which is associated with an extremely poor prognosis. Despite the availability of several therapeutic options, the treatment efficacy remains unsatisfactory for NSCLC brain metastases. Anti-programmed cell death-1 (PD-1) and its ligand (PD-L1) monoclonal antibodies have reshaped therapeutic strategies in advanced NSCLC. Preliminary evidence has shown that anti-PD-(L)1 monotherapy is also effective in NSCLC patients with brain metastases. However, the traditional view asserted that these therapeutic antibodies were incapable of crossing the blood-brain barrier (BBB) with large molecular size, thus most patients with brain metastases were excluded from most studies on anti-PD-(L)1 immunotherapy. Therefore, the efficacy and its mechanisms of action of anti-PD-(L)1 immunotherapy against brain metastases in NSCLC have not been clarified. In this review, we will survey the underlying mechanisms and current clinical advances of anti-PD-(L)1 immunotherapy in the treatment of brain metastases in NSCLC. The trafficking of activated cytotoxic T cells that are mainly derived from the primary tumor and deep cervical lymph nodes is critical for the intracranial response to anti-PD-(L)1 immunotherapy, which is driven by interferon-γ (IFN-γ). Additionally, promising combined strategies with the rationale in the treatment of brain metastases will be presented to provide future directions for clinical study design. Several significant challenges in the preclinical and clinical studies of brain metastases, as well as potential solutions, will also be discussed.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Neoplasias Encefálicas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Ensayos Clínicos como Asunto , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia , Neoplasias Pulmonares/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA