Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Comput Biol ; 20(4): e1011945, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38578805

RESUMEN

Early identification of safe and efficacious disease targets is crucial to alleviating the tremendous cost of drug discovery projects. However, existing experimental methods for identifying new targets are generally labor-intensive and failure-prone. On the other hand, computational approaches, especially machine learning-based frameworks, have shown remarkable application potential in drug discovery. In this work, we propose Progeni, a novel machine learning-based framework for target identification. In addition to fully exploiting the known heterogeneous biological networks from various sources, Progeni integrates literature evidence about the relations between biological entities to construct a probabilistic knowledge graph. Graph neural networks are then employed in Progeni to learn the feature embeddings of biological entities to facilitate the identification of biologically relevant target candidates. A comprehensive evaluation of Progeni demonstrated its superior predictive power over the baseline methods on the target identification task. In addition, our extensive tests showed that Progeni exhibited high robustness to the negative effect of exposure bias, a common phenomenon in recommendation systems, and effectively identified new targets that can be strongly supported by the literature. Moreover, our wet lab experiments successfully validated the biological significance of the top target candidates predicted by Progeni for melanoma and colorectal cancer. All these results suggested that Progeni can identify biologically effective targets and thus provide a powerful and useful tool for advancing the drug discovery process.


Asunto(s)
Biología Computacional , Descubrimiento de Drogas , Aprendizaje Automático , Redes Neurales de la Computación , Humanos , Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Algoritmos , Melanoma , Probabilidad , Neoplasias Colorrectales
2.
J Immunother Cancer ; 12(3)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38458637

RESUMEN

BACKGROUND: Dendritic cell (DC)-mediated antigen presentation is essential for the priming and activation of tumor-specific T cells. However, few drugs that specifically manipulate DC functions are available. The identification of drugs targeting DC holds great promise for cancer immunotherapy. METHODS: We observed that type 1 conventional DCs (cDC1s) initiated a distinct transcriptional program during antigen presentation. We used a network-based approach to screen for cDC1-targeting therapeutics. The antitumor potency and underlying mechanisms of the candidate drug were investigated in vitro and in vivo. RESULTS: Sitagliptin, an oral gliptin widely used for type 2 diabetes, was identified as a drug that targets DCs. In mouse models, sitagliptin inhibited tumor growth by enhancing cDC1-mediated antigen presentation, leading to better T-cell activation. Mechanistically, inhibition of dipeptidyl peptidase 4 (DPP4) by sitagliptin prevented the truncation and degradation of chemokines/cytokines that are important for DC activation. Sitagliptin enhanced cancer immunotherapy by facilitating the priming of antigen-specific T cells by DCs. In humans, the use of sitagliptin correlated with a lower risk of tumor recurrence in patients with colorectal cancer undergoing curative surgery. CONCLUSIONS: Our findings indicate that sitagliptin-mediated DPP4 inhibition promotes antitumor immune response by augmenting cDC1 functions. These data suggest that sitagliptin can be repurposed as an antitumor drug targeting DC, which provides a potential strategy for cancer immunotherapy.


Asunto(s)
Antineoplásicos , Diabetes Mellitus Tipo 2 , Neoplasias , Ratones , Animales , Humanos , Dipeptidil Peptidasa 4/metabolismo , Células Dendríticas , Fosfato de Sitagliptina/farmacología , Fosfato de Sitagliptina/uso terapéutico , Fosfato de Sitagliptina/metabolismo , Presentación de Antígeno , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
4.
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
6.
Cell Res ; 31(9): 1011-1023, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34267349

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global crisis, urgently necessitating the development of safe, efficacious, convenient-to-store, and low-cost vaccine options. A major challenge is that the receptor-binding domain (RBD)-only vaccine fails to trigger long-lasting protective immunity if used alone for vaccination. To enhance antigen processing and cross-presentation in draining lymph nodes (DLNs), we developed an interferon (IFN)-armed RBD dimerized by an immunoglobulin fragment (I-R-F). I-R-F efficiently directs immunity against RBD to DLNs. A low dose of I-R-F induces not only high titers of long-lasting neutralizing antibodies (NAbs) but also more comprehensive T cell responses than RBD. Notably, I-R-F provides comprehensive protection in the form of a one-dose vaccine without an adjuvant. Our study shows that the pan-epitope modified human I-R-F (I-P-R-F) vaccine provides rapid and complete protection throughout the upper and lower respiratory tracts against a high-dose SARS-CoV-2 challenge in rhesus macaques. Based on these promising results, we have initiated a randomized, placebo-controlled, phase I/II trial of the human I-P-R-F vaccine (V-01) in 180 healthy adults, and the vaccine appears safe and elicits strong antiviral immune responses. Due to its potency and safety, this engineered vaccine may become a next-generation vaccine candidate in the global effort to overcome COVID-19.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Inmunogenicidad Vacunal/inmunología , Unión Proteica/inmunología , Dominios Proteicos/inmunología , SARS-CoV-2/inmunología , Adolescente , Adulto , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Antivirales/inmunología , Línea Celular , Chlorocebus aethiops , Método Doble Ciego , Femenino , Células HEK293 , Humanos , Interferones/inmunología , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Persona de Mediana Edad , Vacunación/métodos , Células Vero , Adulto Joven
7.
Nat Commun ; 11(1): 4835, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32973173

RESUMEN

Immune checkpoint blockade therapies have shown clinical promise in a variety of cancers, but how tumor-infiltrating T cells are activated remains unclear. In this study, we explore the functions of PD-L1 on dendritic cells (DCs), which highly express PD-L1. We observe that PD-L1 on DC plays a critical role in limiting T cell responses. Type 1 conventional DCs are essential for PD-L1 blockade and they upregulate PD-L1 upon antigen uptake. Upregulation of PD-L1 on DC is mediated by type II interferon. While DCs are the major antigen presenting cells for cross-presenting tumor antigens to T cells, subsequent PD-L1 upregulation protects them from killing by cytotoxic T lymphocytes, yet dampens the antitumor responses. Blocking PD-L1 in established tumors promotes re-activation of tumor-infiltrating T cells for tumor control. Our study identifies a critical and dynamic role of PD-L1 on DC, which needs to be harnessed for better invigoration of antitumor immune responses.


Asunto(s)
Antígeno B7-H1/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Activación de Linfocitos , Animales , Antígenos de Neoplasias/metabolismo , Antígeno B7-H1/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Interferón gamma/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología , Regulación hacia Arriba
8.
Nat Commun ; 11(1): 1578, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32221289

RESUMEN

PARP1 and PARP2 dual inhibitors, such as olaparib, have been recently FDA approved for the treatment of advanced breast and ovarian cancers. However, their effects on bone mass and bone metastasis are unknown. Here we show that olaparib increases breast cancer bone metastasis through PARP2, but not PARP1, specifically in the myeloid lineage, but not in the cancer cells. Olaparib treatment or PARP1/2 deletion promotes osteoclast differentiation and bone loss. Intriguingly, myeloid deletion of PARP2, but not PARP1, increases the population of immature myeloid cells in bone marrow, and impairs the expression of chemokines such as CCL3 through enhancing the transcriptional repression by ß-catenin. Compromised CCL3 production in turn creates an immune-suppressive milieu by altering T cell subpopulations. Our findings warrant careful examination of current PARP inhibitors on bone metastasis and bone loss, and suggest cotreatment with CCL3, ß-catenin inhibitors, anti-RANKL or bisphosphonates as potential combination therapy for PARP inhibitors.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Resorción Ósea/patología , Neoplasias de la Mama/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Quimiocina CCL3/deficiencia , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Femenino , Eliminación de Gen , Humanos , Ratones Noqueados , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoclastos/patología , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/deficiencia , Regiones Promotoras Genéticas/genética , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Tibia/diagnóstico por imagen , Tibia/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , beta Catenina/metabolismo
9.
Nat Commun ; 9(1): 4586, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389912

RESUMEN

Many patients remain unresponsive to intensive PD-1/PD-L1 blockade therapy despite the presence of tumor-infiltrating lymphocytes. We propose that impaired innate sensing might limit the complete activation of tumor-specific T cells after PD-1/PD-L1 blockade. Local delivery of type I interferons (IFNs) restores antigen presentation, but upregulates PD-L1, dampening subsequent T-cell activation. Therefore, we armed anti-PD-L1 antibody with IFNα (IFNα-anti-PD-L1) to create feedforward responses. Here, we find that a synergistic effect is achieved to overcome both type I IFN and checkpoint blockade therapy resistance with the least side effects in advanced tumors. Intriguingly, PD-L1 expressed in either tumor cells or tumor-associated host cells is sufficient for fusion protein targeting. IFNα-anti-PD-L1 activates IFNAR signaling in host cells, but not in tumor cells to initiate T-cell reactivation. Our data suggest that a next-generation PD-L1 antibody armed with IFNα improves tumor targeting and antigen presentation, while countering innate or T-cell-driven PD-L1 upregulation within tumor.


Asunto(s)
Anticuerpos/farmacología , Antineoplásicos/farmacología , Antígeno B7-H1/metabolismo , Inmunoterapia , Interferón-alfa/farmacología , Neoplasias/terapia , Animales , Femenino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/patología , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal
10.
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
11.
Methods Mol Biol ; 1845: 275-286, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30141019

RESUMEN

Tumor microenvironments (TME) are usually immunosuppressive and prevent lymphocyte priming. Recent clinical trials have shown that cancer immunotherapy such as immune checkpoint inhibitors can induce unprecedented durable responses in patients with a variety of cancers. Tertiary lymphoid structures (TLS) can form inside or adjacent to tumor tissues due to persistent inflammation. The formation of TLS facilitates lymphocyte trafficking and infiltration into tumor tissues. It can also support effective antigen presentation and lymphocyte activation. Thus, TLS have become an intriguing target to manipulate antitumor immunity. Several therapeutics targeting TLS have been developed and shown promising antitumor effects in various mouse models. In this chapter, we describe the general approach to establish transplantable mouse tumor models for the study of immunotherapy. We introduce the strategies for therapy through systemic or local treatment targeting TLS. We also present approaches to evaluate the antitumor immune responses provoked by the therapies.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/patología , Estructuras Linfoides Terciarias/inmunología , Estructuras Linfoides Terciarias/patología , Microambiente Tumoral/inmunología , Animales , Biomarcadores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental , Ratones , Neoplasias/metabolismo , Neoplasias/terapia , Estructuras Linfoides Terciarias/metabolismo
12.
Cell Metab ; 27(5): 945-946, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29628418

RESUMEN

Accumulating data suggest an important role of tumor metabolism during cancer development, metastasis, and therapeutic resistance. In Cell Metabolism, Cascone et al. (2018) show that increased tumor glycolysis suppresses anti-tumor immunity by impairing T cell killing and trafficking to the tumor microenvironment.


Asunto(s)
Glucólisis , Neoplasias , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Evasión Inmune , Microambiente Tumoral
13.
J Clin Invest ; 128(2): 580-588, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29337303

RESUMEN

Programmed death-ligand 1 (PD-L1) expression on tumor cells is essential for T cell impairment, and PD-L1 blockade therapy has shown unprecedented durable responses in several clinical studies. Although higher expression of PD-L1 on tumor cells is associated with a better immune response after Ab blockade, some PD-L1-negative patients also respond to this therapy. In the current study, we explored whether PD-L1 on tumor or host cells was essential for anti-PD-L1-mediated therapy in 2 different murine tumor models. Using real-time imaging in whole tumor tissues, we found that anti-PD-L1 Ab accumulates in tumor tissues, regardless of the status of PD-L1 expression on tumor cells. We further observed that, while PD-L1 on tumor cells was largely dispensable for the response to checkpoint blockade, PD-L1 in host myeloid cells was essential for this response. Additionally, PD-L1 signaling in defined antigen-presenting cells (APCs) negatively regulated and inhibited T cell activation. PD-L1 blockade inside tumors was not sufficient to mediate regression, as limiting T cell trafficking reduced the efficacy of the blockade. Together, these findings demonstrate that PD-L1 expressed in APCs, rather than on tumor cells, plays an essential role in checkpoint blockade therapy, providing an insight into the mechanisms of this therapy.


Asunto(s)
Antígeno B7-H1/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/inmunología , Células Presentadoras de Antígenos/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular , Línea Celular Tumoral , Citometría de Flujo , Inmunidad Celular , Inmunoterapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Microambiente Tumoral/inmunología
14.
Cell Mol Immunol ; 15(7): 697-709, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28579615

RESUMEN

Splenomegaly is a well-known phenomenon typically associated with inflammation. However, the underlying cause of this phenotype has not been well characterized. Furthermore, the splenomegaly phenotype seen in lymphotoxin (LT) signaling-deficient mice is characterized by increased numbers of splenocytes and splenic neutrophils. Splenomegaly, as well as the related phenotype of increased lymphocyte counts in non-lymphoid tissues, is thought to result from the absence of secondary lymphoid tissues in LT-deficient mice. We now present evidence that mice deficient in LTα1ß2 or LTßR develop splenomegaly and increased numbers of lymphocytes in non-lymphoid tissues in a microbiota-dependent manner. Antibiotic administration to LTα1ß2- or LTßR-deficient mice reduces splenomegaly. Furthermore, re-derived germ-free Ltbr-/- mice do not exhibit splenomegaly or increased inflammation in non-lymphoid tissues compared to specific pathogen-free Ltbr-/- mice. By using various LTß- and LTßR-conditional knockout mice, we demonstrate that retinoic acid-related orphan receptor γT-positive type 3 innate lymphoid cells provide the required active LT signaling to prevent the development of splenomegaly. Thus, this study demonstrates the importance of LT-mediated immune responses for the prevention of splenomegaly and systemic inflammation induced by microbiota.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Heterotrímero de Linfotoxina alfa1 y beta2/inmunología , Receptor beta de Linfotoxina/inmunología , Microbiota/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Animales , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Linfocitos/patología , Heterotrímero de Linfotoxina alfa1 y beta2/genética , Receptor beta de Linfotoxina/genética , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/genética
15.
Nat Biomed Eng ; 2(8): 600-610, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-31015630

RESUMEN

Checkpoint blockade immunotherapy relies on energized cytotoxic T cells attacking tumour tissue systemically. However, for many cancers, the reliance on T cell infiltration leads to low response rates. Conversely, radiotherapy has served as a powerful therapy for local tumours over the past 100 years, yet is rarely sufficient to cause systemic tumour rejection. Here, we describe a treatment strategy that combines nanoscale metal-organic framework (nMOF)-enabled radiotherapy-radiodynamic therapy with checkpoint blockade immunotherapy for both local and systemic tumour elimination. In mouse models of breast and colorectal cancer, intratumorally injected nMOFs treated with low doses of X-ray irradiation led to the eradication of local tumours and, when loaded with an inhibitor of the immune checkpoint molecule indoleamine 2,3-dioxygenase, the irradiated nMOFs led to consistent abscopal responses that rejected distal tumours. By combining the advantages of local radiotherapy and systemic tumour rejection via synergistic X-ray-induced in situ vaccination and indoleamine 2,3-dioxygenase inhibition, nMOFs may overcome some of the limitations of checkpoint blockade in cancer treatment.


Asunto(s)
Inmunoterapia/métodos , Estructuras Metalorgánicas/química , Nanoestructuras/química , Terapia por Rayos X/métodos , Animales , Antineoplásicos , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Terapia Combinada , Humanos , Estructuras Metalorgánicas/farmacología , Ratones , Nanomedicina
16.
Cell Mol Immunol ; 14(10): 809-818, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28413217

RESUMEN

Tertiary lymphoid structures (TLS) often develop at sites of persistent inflammation, including cancers and autoimmune diseases. In most cases, the presence of TLS correlates with active immune responses. Because of their proximity to pathological loci, TLS are an intriguing target for the manipulation of immune responses. For several years, it has become clear that lymphotoxin (LT) signalling plays critical roles in lymphoid tissue organogenesis and maintenance. In the current review, we will discuss the role of LT signalling in the development of TLS. With a focus on cancers and autoimmune diseases, we will highlight the correlations between TLS and disease progression. We will also discuss the current efforts and potential directions for manipulating TLS for immunotherapies.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Inmunoterapia/métodos , Tejido Linfoide/fisiología , Linfotoxina-alfa/metabolismo , Neoplasias/inmunología , Estructuras Linfoides Terciarias/inmunología , Animales , Carcinogénesis , Progresión de la Enfermedad , Humanos , Organogénesis , Transducción de Señal
17.
Curr Opin Immunol ; 45: 16-20, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28088707

RESUMEN

The identification of critical DNA sensors and their pathways has led to revealing the central role of DNA sensing in immune system. It has been initially demonstrated that DNA sensing and immune responses have high impacts on the development and prevention of infection and inflammatory. In addition to toll-like receptor pathways, there is now also emerging evidence that cytosolic enzyme cyclic GMP-AMP synthase (cGAS) is essential for the recognition of not only pathogen-derived DNA but also tumor DNA for innate sensing. The strategies through activating DNA sensing pathways toward enhancing antitumor immunity have shown promise and are further tested in clinical studies. Here, we highlight recent progresses in understanding mechanisms activated by DNA sensing mediated immune responses in cancer therapy.


Asunto(s)
ADN de Neoplasias/inmunología , Inmunidad Innata , Neoplasias/inmunología , Nucleotidiltransferasas/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Animales , Humanos , Neoplasias/patología , Neoplasias/terapia
18.
J Immunol ; 198(1): 55-60, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27913631

RESUMEN

The immunological components that control resolution of Salmonella infection and successful vaccination are poorly defined. In a model of chronic gastrointestinal infection, we observed that the lymphotoxin (LT) pathway is essential for the clearance and resolution of primary infection of attenuated Salmonella enterica Typhimurium strain SL3261 ΔaroA Using gnotobiotic mice, we show that LTß receptor (LTßR) signaling and the microbiota are required to promote clearance of attenuated S. enterica Typhimurium from the gut lumen. We also found that LTßR signaling was required for successful immunization and subsequent protection upon challenge with a virulent strain of S enterica Typhimurium. LTßR signaling promoted the development of specific IgG recognizing S enterica Typhimurium during infection, as well as Ag-driven IFN-γ responses. B cell- and type 3 innate lymphoid cell-derived LT signaling, but not T cell-derived LT, contributes to anti-S enterica Typhimurium protective responses. Collectively, our results suggest that LT signaling is essential for multiple steps of anti-S enterica Typhimurium immune responses.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Linfotoxina-alfa/inmunología , Salmonelosis Animal/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Vida Libre de Gérmenes , Ratones , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología
20.
Cancer Cell ; 29(3): 285-296, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26977880

RESUMEN

Immune checkpoint blockade therapies fail to induce responses in the majority of cancer patients, so how to increase the objective response rate becomes an urgent challenge. Here, we demonstrate that sufficient T cell infiltration in tumor tissues is a prerequisite for response to PD-L1 blockade. Targeting tumors with tumor necrosis factor superfamily member LIGHT activates lymphotoxin ß-receptor signaling, leading to the production of chemokines that recruit massive numbers of T cells. Furthermore, targeting non-T cell-inflamed tumor tissues by antibody-guided LIGHT creates a T cell-inflamed microenvironment and overcomes tumor resistance to checkpoint blockade. Our data indicate that targeting LIGHT might be a potent strategy to increase the responses to checkpoint blockades and other immunotherapies in non-T cell-inflamed tumors.


Asunto(s)
Antineoplásicos/inmunología , Antígeno B7-H1/inmunología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Secuencia de Aminoácidos , Animales , Resistencia a Antineoplásicos/inmunología , Humanos , Inmunoterapia/métodos , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...