Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Mater ; 35(36): e2303080, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37249019

RESUMEN

To demonstrate potent efficacy, a cancer vaccine needs to activate both innate and adaptive immune cells. Personalized cancer vaccine strategies often require the identification of patient-specific neoantigens; however, the clonal and mutational heterogeneity of cancer cells presents inherent challenges. Here, extracellular nanovesicles derived from alpha-galactosylceramide-conjugated autologous acute myeloid leukemia (AML) cells (ECNV-αGC) are presented as a personalized therapeutic vaccine that activates both innate and adaptive immune responses, bypassing the need to identify patient-specific neoantigens. ECNV-αGC vaccination directly engages with and activates both invariant natural killer T (iNKT) cells and leukemia-specific CD8+ T cells in mice with AML, thereby promoting long-term anti-leukemic immune memory. ECNV-αGC sufficiently serves as an antigen-presenting platform that can directly activate antigen-specific CD8+ T cells even in the absence of dendritic cells, thereby demonstrating a multifaceted cellular mechanism of immune activation. Moreover, ECNV-αGC vaccination results in a significantly lower AML burden and higher percentage of leukemia-free survivors among cytarabine-treated hosts with AML. Human AML-derived ECNV-αGCs activate iNKT cells in both healthy individuals and patients with AML regardless of responsiveness to conventional therapies. Together, autologous AML-derived ECNV-αGCs may be a promising personalized therapeutic vaccine that efficiently establishes AML-specific long-term immunity without requiring the identification of neoantigens.


Asunto(s)
Vacunas contra el Cáncer , Leucemia Mieloide Aguda , Células T Asesinas Naturales , Humanos , Animales , Ratones , Linfocitos T CD8-positivos , Activación de Linfocitos , Leucemia Mieloide Aguda/terapia
2.
J Immunother Cancer ; 11(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36693679

RESUMEN

BACKGROUND: Immunological contexture differs across malignancies, and understanding it in the tumor microenvironment (TME) is essential for development of new anticancer agents in order to achieve synergistic effects with anti-programmed cell death protein-1 (PD-1) therapy. TYRO3, AXL, and MERTK receptors are bi-expressed in both cancer and immune cells, and thus emerge as promising targets for therapeutic intervention. Whereas AXL and MERTK have been extensively studied, the role of TYRO3, in the TME, is still undetermined. METHODS: Here, we screened the TYRO3-focused chemical library consisting of 208 compounds and presented a potent and highly selective TYRO3 inhibitor, KRCT87. We explored the role of TYRO3 using mouse engrafting MC38 or 4T1 tumors. We validated the results using flow cytometry, RNA sequencing analysis, gene knockdown or overexpression, ex vivo immune cells isolation from mouse models, immunoblotting and quantitative PCR. Flow cytometry was used for the quantification of cell populations and immunophenotyping of macrophages and T cells. Co-cultures of macrophages and T cells were performed to verify the role of CCN1 in the tumors. RESULTS: TYRO3 blockade boosts antitumor immune responses in both the tumor-draining lymph nodes and tumors in MC38-syngeneic mice models. Moreover, the combination of KRCT87 and anti-PD-1 therapy exerts significant synergistic antitumor effects in anti-PD-1-non-responsive 4T1-syngeneic model. Mechanistically, we demonstrated that inhibition of TYRO3-driven CCN1 secretion fosters macrophages into M1-skewing phenotypes, thereby triggering antitumor T-cell responses. CCN1 overexpression in MC38 tumors diminishes responsiveness to anti-PD-1 therapy. CONCLUSIONS: The activated TYRO3-CCN1 axis in cancer could dampen anti-PD-1 therapy responses. These findings highlight the potential of TYRO3 blockade to improve the clinical outcomes of anti-PD-1 therapy.


Asunto(s)
Microambiente Tumoral , Ratones , Animales , Tirosina Quinasa c-Mer , Línea Celular Tumoral , Modelos Animales de Enfermedad
3.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34663730

RESUMEN

GPCR-Gα protein-mediated signal transduction contributes to spatiotemporal interactions between immune cells to fine-tune and facilitate the process of inflammation and host protection. Beyond this, however, how Gα proteins contribute to the helper T cell subset differentiation and adaptive response have been underappreciated. Here, we found that Gα13 signaling in T cells plays a crucial role in inducing follicular helper T (Tfh) cell differentiation in vivo. T cell-specific Gα13-deficient mice have diminished Tfh cell responses in a cell-intrinsic manner in response to immunization, lymphocytic choriomeningitis virus infection, and allergen challenges. Moreover, Gα13-deficient Tfh cells express reduced levels of Bcl-6 and CXCR5 and are functionally impaired in their ability to adhere to and stimulate B cells. Mechanistically, Gα13-deficient Tfh cells harbor defective Rho-ROCK2 activation, and Rho agonist treatment recuperates Tfh cell differentiation and expression of Bcl-6 and CXCR5 in Tfh cells of T cell-specific Gα13-deficient mice. Conversely, ROCK inhibitor treatment hampers Tfh cell differentiation in wild-type mice. These findings unveil a crucial regulatory role of Gα13-Rho-ROCK axis in optimal Tfh cell differentiation and function, which might be a promising target for pharmacologic intervention in vaccine development as well as antibody-mediated immune disorders.


Asunto(s)
Diferenciación Celular , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Transducción de Señal , Células T Auxiliares Foliculares/citología , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Receptores CXCR5/metabolismo , Timo/citología , Timo/crecimiento & desarrollo , Timo/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
4.
J Immunother Cancer ; 9(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34083422

RESUMEN

BACKGROUND: Multiple types of immune cells producing IL-17 are found in the tumor microenvironment. However, their roles in tumor progression and exhaustion of CD8+ tumor-infiltrating lymphocytes (TILs) remain unclear. METHODS: To determine the role of type 17 immunity in tumor, we investigated the growth of B16F10 melanoma and the exhaustion of CD8+ TILs in Il17a-/- mice, Il17aCreR26DTA mice, RORγt inhibitor-treated mice, or their respective control mice. Adoptive transfer of tumor-specific IL-17-producing T cells was performed in B16F10-bearing congenic mice. Anti-CD4 or anti-Ly6G antibodies were used to deplete CD4+ T cells or CD11b+Gr-1hi myeloid cells in vivo, respectively. Correlation between type 17 immunity and T cell exhaustion in human cancer was evaluated by interrogating TCGA dataset. RESULTS: Depletion of CD4+ T cells promotes the exhaustion of CD8+ T cells with a concomitant increase in IL-17-producing CD8+ T (Tc17) cells in the tumor. Unlike IFN-γ-producing CD8+ T (Tc1) cells, tumor-infiltrating Tc17 cells exhibit CD103+KLRG1-IL-7Rαhi tissue resident memory-like phenotypes and are poorly cytolytic. Adoptive transfer of IL-17-producing tumor-specific T cells increases, while depletion of IL-17-producing cells decreases, the frequency of PD-1hiTim3+TOX+ terminally exhausted CD8+ T cells in the tumor. Blockade of IL-17 or RORγt pathway inhibits exhaustion of CD8+ T cells and also delays tumor growth in vivo. Consistent with these results, human TCGA analyses reveal a strong positive correlation between type 17 and CD8+ T cell exhaustion signature gene sets in multiple cancers. CONCLUSION: IL-17-producing cells promote terminal exhaustion of CD8+ T cells and tumor progression in vivo, which can be reversed by blockade of IL-17 or RORγt pathway. These findings unveil a novel role for IL-17-producing cells as tumor-promoting cells facilitating CD8+ T cell exhaustion, and propose type 17 immunity as a promising target for cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Eliminación de Gen , Interleucina-17/genética , Melanoma Experimental/terapia , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Femenino , Humanos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Microambiente Tumoral
5.
Exp Mol Med ; 52(6): 896-910, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32576930

RESUMEN

As the key governors of diverse physiological processes, G protein-coupled receptors (GPCRs) have drawn attention as primary targets for several diseases, including diabetes and cardiovascular disease. Heterotrimeric G proteins converge signals from ~800 members of the GPCR family. Among the members of the G protein α family, the Gα12 family members comprising Gα12 and Gα13 have been referred to as gep oncogenes. Gα12/13 levels are altered in metabolic organs, including the liver and muscles, in metabolic diseases. The roles of Gα12/13 in metabolic diseases have been investigated. In this review, we highlight findings demonstrating Gα12/13 amplifying or dampening regulators of phenotype changes. We discuss the molecular basis of G protein biology in the context of posttranslational modifications to heterotrimeric G proteins and the cell signaling axis. We also highlight findings providing insights into the organ-specific, metabolic and pathological roles of G proteins in changes associated with specific cells, energy homeostasis, glucose metabolism, liver fibrosis and the immune and cardiovascular systems. This review summarizes the currently available knowledge on the importance of Gα12/13 in the physiology and pathogenesis of metabolic diseases, which is presented according to the basic understanding of their metabolic actions and underlying cellular and molecular bases.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Enfermedades Metabólicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Humanos , Enfermedades Metabólicas/genética , Receptores Acoplados a Proteínas G/genética
6.
Biomolecules ; 10(6)2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32471185

RESUMEN

Transforming growth factor beta 1 (TGF-ß1) is an immunosuppresive cytokine that plays an essential role in immune homeostasis. It is well known that regulatory T (Treg) cells express TGF-ß1; however, the role of autocrine TGF-ß1 in the development, function, and stability of Treg cells remains poorly understood. We found that Treg cell-derived TGF-ß1 was not required for the development of thymic Treg cells in mice, but played a role in the expression of latency-associated peptide and optimal suppression of naïve T cell proliferation in vitro. Moreover, the frequency of Treg cells was significantly reduced in the mesenteric lymph nodes of the Treg cell-specific TGF-ß1-deficient mice, which was associated with increased frequency of IFN-γ-producers among Treg cells. TGF-ß1-deficient Treg cells were more prone to express IFN-γ than TGF-ß1-sufficient Treg cells in a dendritic cell-mediated stimulation in vitro as well as in an adoptive transfer study in vivo. Mechanistically, TGF-ß1-deficient Treg cells expressed higher levels of Il12rb2 and were more sensitive to IL-12-induced conversion into IFN-γ-producing Treg cells or IFN-γ-producing exTreg cells than TGF-ß1-sufficient Treg cells. Our findings demonstrate that autocrine TGF-ß1 plays a critical role in the optimal suppressive activity and stability of Treg cells by downregulating IL-12R on Treg cells.


Asunto(s)
Regulación hacia Abajo , Factores de Transcripción Forkhead/metabolismo , Receptores de Interleucina-12/metabolismo , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Comunicación Autocrina , Factores de Transcripción Forkhead/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Receptores de Interleucina-12/genética
7.
Immune Netw ; 20(1): e6, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32158594

RESUMEN

IL-17 is produced by RAR-related orphan receptor gamma t (RORγt)-expressing cells including Th17 cells, subsets of γδT cells and innate lymphoid cells (ILCs). The biological significance of IL-17-producing cells is well-studied in contexts of inflammation, autoimmunity and host defense against infection. While most of available studies in tumor immunity mainly focused on the role of T-bet-expressing cells, including cytotoxic CD8+ T cells and NK cells, and their exhaustion status, the role of IL-17-producing cells remains poorly understood. While IL-17-producing T-cells were shown to be anti-tumorigenic in adoptive T-cell therapy settings, mice deficient in type 17 genes suggest a protumorigenic potential of IL-17-producing cells. This review discusses the features of IL-17-producing cells, of both lymphocytic and myeloid origins, as well as their suggested pro- and/or anti-tumorigenic functions in an organ-dependent context. Potential therapeutic approaches targeting these cells in the tumor microenvironment will also be discussed.

8.
Exp Mol Med ; 50(8): 1-13, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30135516

RESUMEN

Recent advances in the understating of tumor immunology suggest that cancer immunotherapy is an effective treatment against various types of cancer. In particular, the remarkable successes of immune checkpoint-blocking antibodies in clinical settings have encouraged researchers to focus on developing other various immunologic strategies to combat cancer. However, such immunotherapies still face difficulties in controlling malignancy in many patients due to the heterogeneity of both tumors and individual patients. Here, we discuss how tumor-intrinsic cues, tumor environmental metabolites, and host-derived immune cells might impact the efficacy and resistance often seen during immune checkpoint blockade treatment. Furthermore, we introduce biomarkers identified from human and mouse models that predict clinical benefits for immune checkpoint blockers in cancer.


Asunto(s)
Resistencia a Antineoplásicos , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Animales , Microbioma Gastrointestinal , Humanos , Pronóstico , Microambiente Tumoral
9.
J Phys Chem A ; 118(16): 2942-51, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24689807

RESUMEN

Methanol in aprotic solvents can serve as a case study for self-association via hydrogen-bonding, which is an important process in many biological and environmental systems. Incoherent broadband cavity-enhanced absorption spectroscopy (IBBCEAS), which provides enhanced sensitivity relative to conventional single-pass absorption techniques, has been used to characterize the third "free" O-H stretching overtone of methanol in four aprotic solvents (CCl4, CHCl3, CH2Cl2, and C6H6), including the transition wavenumber, bandwidth, and molar absorptivity. The absorption band characteristics indicate an increasing degree of nonspecific methanol-solvent interaction with increasing solvent dielectric constant, except in the case of benzene, which shows evidence of a specific, H-π interaction. Density functional theory with the polarizable continuum model was used to complement the results by assessing the accuracy of computational methods for calculating anharmonic O-H stretching frequencies. Finally, the self-association of methanol in these solvents at 298 K was also investigated using the concentration dependence of the overtone absorption intensity. The propensity for methanol's self-association in the solvents studied increases in the order: CH2Cl2 ∼ CHCl3 < C6H6 < CCl4.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...