Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Viral Immunol ; 37(1): 24-35, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38301135

RESUMEN

Interleukin-36 (IL-36) signaling plays an important role in promoting CD8+ T cell-mediated antitumor immune responses. The role of IL-36 signaling in CD8+ T cells that are involved in host immune responses during human immunodeficiency virus-1 (HIV-1) infection has not been characterized. Sixty-one patients living with chronic HIV-1 infection and 23 controls were enrolled in this study. The levels of IL-36 cytokine family members were measured by enzyme-linked immunosorbent assay. Purified CD8+ T cells were stimulated with recombinant IL-36gamma (1 or 10 ng/mL). The expression of inhibitory receptors, the secretion of cytotoxic molecules and interferon-gamma, and the mRNA levels of apoptosis-related ligands were assessed to evaluate the effect of IL-36gamma on CD8+ T cell function in vitro. There were no significant differences in IL-36alpha, IL-36beta, or IL-36 receptor antagonist levels between patients living with chronic HIV-1 infection and controls. Plasma IL-36gamma levels were reduced in patients living with chronic HIV-1 infection. Perforin, granzyme B, and granulysin secretion, as well as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) mRNA expression, but not programmed death-1 (PD-1) or cytotoxic T lymphocyte-associated protein-4 (CTLA-4) expression was downregulated in CD8+ T cells from patients living with chronic HIV-1 infection. The addition of both 1 and 10 ng/mL IL-36gamma enhanced perforin, granzyme B, granulysin, and interferon-gamma secretion by CD8+ T cells without affecting PD-1/CTLA-4 or TRAIL/FasL mRNA expression in CD8+ T cells from patients living with chronic HIV-1 infection. The addition of 1 ng/mL IL-36gamma also promoted perforin and granzyme B secretion by HIV-1-specific CD8+ T cells from patients living with chronic HIV-1 infection. The reduced IL-36gamma levels in patients living with chronic HIV-1 infection might be insufficient for the activation of CD8+ T cells, leading to CD8+ T cell exhaustion.


Asunto(s)
Linfocitos T CD8-positivos , Infecciones por VIH , Humanos , Antígeno CTLA-4 , Granzimas/farmacología , VIH , Interferón gamma , Interleucinas/farmacología , Perforina/farmacología , Receptor de Muerte Celular Programada 1 , ARN Mensajero
2.
Hepatol Int ; 18(2): 610-622, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37864726

RESUMEN

BACKGROUND & AIMS: Combination immunotherapy refers to the use of immune checkpoint inhibitors (ICI) and molecular-targeted agents (MTA), which have recently been approved for the treatment of advanced hepatocellular carcinoma (HCC). Owing to its relatively low antitumor effect (up to 30%), sequential therapy following ICIs treatment is required in patients with HCC. This study aimed to determine the impact of MTAs on the tumor immune microenvironment (TIME). METHODS: We established immune syngeneic orthotopic HCC mouse models using Hep-55.1C and Hep-53.4, and treated them with MTAs (lenvatinib, sorafenib, regorafenib, cabozantinib, and DC101 as anti-vascular endothelial growth factor receptor-2 antibodies, and AZD4547 as a fibroblast growth factor receptor (FGFR)-1/2/3/4 inhibitor) for 2 weeks. Subsequently, alterations in the TIME caused by MTAs were evaluated using immunohistochemistry (antibodies for CD3, CD8, Foxp3, Granzyme B, Arginase-1, NK1.1, F4/80, CD11c, PD-1, and PD-L1). We conducted RNA-seq analysis using lenvatinib- and AZD4547-treated tumors. To confirm the clinical relevance of these findings, we analyzed the transcriptome data of human HCC cells (MHCC-97H) treated with various concentrations of lenvatinib for 24 h using RNA-seq data from the Gene Expression Omnibus database. RESULTS: The number of Foxp3- and F4/80-positive cells in the TIME was decreased in many MTAs. Cabozantinib increased the numbers in NK1.1-, Granzyme B, and CD11c-positive cells. Lenvatinib and AZD4547 increased the number of CD8, Granzyme B, and PD-L1-positive cells. Gene ontology enrichment analysis revealed that lipid metabolism-related genes were downregulated by lenvatinib and AZD4547. In total, 161 genes downregulated by FGFR inhibition in rodent models overlapped with those downregulated by lenvatinib in human HCC cells. CONCLUSIONS: In this study, we showed that cabozantinib activated the innate immune system, and lenvatinib and AZD4547, which commonly inhibit FGFR signaling, altered TIME to a hot immune state by downregulating lipid metabolism-related genes. These findings support the therapeutic use of combination immunotherapies.


Asunto(s)
Anilidas , Antineoplásicos , Benzamidas , Carcinoma Hepatocelular , Neoplasias Hepáticas , Compuestos de Fenilurea , Piperazinas , Pirazoles , Piridinas , Quinolinas , Animales , Ratones , Humanos , Carcinoma Hepatocelular/patología , Antígeno B7-H1 , Granzimas/farmacología , Granzimas/uso terapéutico , Neoplasias Hepáticas/patología , Factores de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inmunosupresores/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Factores de Transcripción Forkhead/farmacología , Factores de Transcripción Forkhead/uso terapéutico , Microambiente Tumoral
3.
J Nanobiotechnology ; 21(1): 440, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993847

RESUMEN

BACKGROUND: Hypoxia, a common characteristic of the tumour microenvironment, is involved in tumour progression and immune evasion. Targeting the hypoxic microenvironment has been implicated as a promising antitumour therapeutic strategy. TH-302 can be selectively activated under hypoxic conditions. However, the effectiveness of TH-302 in gastric cancer combined immunotherapy remains unclear. METHODS: We designed mPEG-PLGA-encapsulated TH-302 (TH-302 NPs) to target the hypoxic area of tumour tissues. A particle size analyzer was used to measure the average size and zeta potential of TH-302 NPs. The morphology was observed by transmission electron microscopy and scanning electron microscopy. The hypoxic area of tumour tissues was examined by immunofluorescence assays using pimonidazole. Flow cytometry analysis was performed to measure the levels of TNF-α, IFN-γ, and granzyme B. The synergistic antitumour activity of the combination of TH-302 NPs with anti-PD-1 (α-PD-1) therapy was assessed in vitro and in vivo. Haematoxylin and eosin staining of major organs and biochemical indicator detection were performed to investigate the biological safety of TH-302 NPs in vivo. RESULTS: TH-302 NPs inhibited the proliferation and promoted the apoptosis of gastric cancer cells under hypoxic conditions. In vitro and in vivo experiments confirmed that TH-302 NPs could effectively alleviate tumour hypoxia. TH-302 NPs exhibited high bioavailability, effective tumour-targeting ability and satisfactory biosafety. Moreover, the combination of TH-302 NPs with α-PD-1 significantly improved immunotherapeutic efficacy in vivo. Mechanistically, TH-302 NPs reduced the expression of HIF-1α and PD-L1, facilitated the infiltration of CD8+ T cells and increased the levels of TNF-α, IFN-γ, and granzyme B in tumours, thereby enhancing the efficacy of α-PD-1 therapy. CONCLUSION: TH-302 NPs alleviated the hypoxic tumour microenvironment and enhanced the efficacy of PD-1 blockade. Our results provide evidence that TH-302 NPs can be used as a safe and effective nanodrug for combined immunotherapy in gastric cancer treatment.


Asunto(s)
Nanopartículas , Neoplasias Gástricas , Humanos , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1 , Granzimas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Microambiente Tumoral , Factor de Necrosis Tumoral alfa , Hipoxia/tratamiento farmacológico , Nanopartículas/uso terapéutico
4.
J Mol Med (Berl) ; 101(11): 1437-1448, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37725101

RESUMEN

Macrophages belong to the innate immune system, and we have recently shown that in vitro differentiated human regulatory macrophages (Mreg) release large extracellular vesicles (L-EVMreg) with an average size of 7.5 µm which regulate wound healing and angiogenesis in vitro. The aim of this study was to investigate whether L-EVMreg also affect the CD3/CD28-mediated activation of T-cells. Mreg were differentiated using blood monocytes and L-EVMreg were isolated from culture supernatants by differential centrifugation. Activation of human T-cells was induced by CD3/CD28-coated beads in the absence or presence of Mreg or different concentrations of L-EVMreg. Inhibition of T-cell activation was quantified by flow cytometry and antibodies directed against the T-cell marker granzyme B. Phosphatidylserine (PS) exposure on the surface of Mreg and L-EVMreg was analyzed by fluorescence microscopy. Incubation of human lymphocytes with CD3/CD28 beads resulted in an increase of cell size, cell granularity, and number of granzyme B-positive cells (P < 0.05) which is indicative of T-cell activation. The presence of Mreg (0.5 × 106 Mreg/ml) led to a reduction of T-cell activation (number of granzyme B-positive cells; P < 0.001), and a similar but less pronounced effect was also observed when incubating activated T-cells with L-EVMreg (P < 0.05 for 3.2 × 106 L-EVMreg/ml). A differential analysis of the effects of Mreg and L-EVMreg on CD4+ and CD8+ T-cells showed an inhibition of CD4+ T-cells by Mreg (P < 0.01) and L-EVMreg (P < 0.05 for 1.6 × 106 L-EVMreg/ml; P < 0.01 for 3.2 × 106 L-EVMreg/ml). A moderate inhibition of CD8+ T-cells was observed by Mreg (P < 0.05) and by L-EVMreg (P < 0.01 for 1.6 × 106 L-EVMreg/ml and 3.2 × 106 L-EVMreg/ml). PS was restricted to confined regions of the Mreg surface, while L-EVMreg showed strong signals for PS in the exoplasmic leaflet. L-EVMreg attenuate CD3/CD28-mediated activation of CD4+ and CD8+ T-cells. L-EVMreg may have clinical relevance, particularly in the treatment of diseases associated with increased T-cell activity. KEY MESSAGES: Mreg release large extracellular vesicles (L-EVMreg) with an average size of 7.5 µm L-EVMreg exhibit phosphatidylserine positivity L-EVMreg suppress CD4+ and CD8+ T-cells L-EVMreg hold clinical potential in T-cell-related diseases.


Asunto(s)
Antígenos CD28 , Linfocitos T CD8-positivos , Humanos , Granzimas/farmacología , Fosfatidilserinas/farmacología , Macrófagos , Activación de Linfocitos , Linfocitos T CD4-Positivos
5.
J Allergy Clin Immunol ; 152(5): 1312-1320.e3, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37536509

RESUMEN

BACKGROUND: Eosinophils contribute to the pathology of several types of disorders, in particular of allergic nature, and strategies to limit their actions are therefore warranted. OBJECTIVE: We sought to evaluate the possibility of targeting the acidic, lysosome-like eosinophil granules as a potential means of inducing eosinophil cell death. METHODS: To this end, we used monensin, an ionophoric drug that has previously been shown to permeabilize the secretory granules of mast cells, thereby inducing cell death. RESULTS: Our findings reveal that monensin induces cell death in human eosinophils, whereas neutrophils were less affected. Blockade of granule acidification reduced the effect of monensin on the eosinophils, demonstrating that granule acidity is an important factor in the mechanism of cell death. Furthermore, monensin caused an elevation of the granule pH, which was accompanied by a decrease of the cytosolic pH, hence indicating that monensin caused leakage of acidic contents from the granules into the cytosol. In agreement with a granule-targeting mechanism, transmission electron microscopy analysis revealed that monensin caused extensive morphological alterations of the eosinophil granules, as manifested by a marked loss of electron density. Eosinophil cell death in response to monensin was caspase-independent, but dependent on granzyme B, a pro-apoptotic serine protease known to be expressed by eosinophils. CONCLUSIONS: We conclude that monensin causes cell death of human eosinophils through a granule-mediated mechanism dependent on granzyme B.


Asunto(s)
Eosinófilos , Monensina , Humanos , Monensina/farmacología , Monensina/metabolismo , Granzimas/metabolismo , Granzimas/farmacología , Vesículas Secretoras/metabolismo , Gránulos Citoplasmáticos
6.
Med Oncol ; 40(8): 228, 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37410214

RESUMEN

Research on immunotherapy in breast cancer treatment has recently gained importance. In this context, natural killer (NK) cells have been shown to kill cancer cells without affecting normal cells. Our study used the NK-92 cells that were stimulated with anti-CD226 antibodies (sNK-92) to increase their activity to target MDA-MB-231 triple-negative breast cancer cells. MCF-12A normal breast cells were used as the control in all experiments. The cytotoxic effects of NK-92 and sNK-92 cells on MDA-MB-231 cells were investigated using lactate dehydrogenase tests. The sNK-92 cells were more cytotoxic than NK-92 cells on MDA-MB-231 cells. In contrast, a significant cytotoxic change was not observed in MCF-12A cells cocultured with NK-92 and sNK-92 cells. An increase in granzyme B levels after coculturing with sNK-92 cells was investigated using the granzyme B enzyme-linked immunosorbent assay. The sNK-92 cells secreted more granzyme B than NK-92 cells against MDA-MB-231 cells. This increase was not observed in MCF-12A, indicating that sNK-92 cells specifically target cancer cells. In addition, immunostaining was used to investigate the synthesis level of BAX, CASP3, and CASP9 proteins to determine whether the observed cytotoxic effect was due to apoptosis. These proteins were synthesized more in MDA-MB-231 cells cocultured with sNK-92 than with NK-92 cells. However, no increase in their synthesis was observed in normal breast cells cocultured with NK-92 and sNK-92 cells. In conclusion, NK-92 cells stimulated with anti-CD226 antibodies secrete more granzyme B, resulting in a greater cytotoxic effect by inducing programmed cell death (apoptosis). The fact that the observed effects on breast cancer cells were not observed in normal breast cells indicates that sNK-92 cells specifically target breast cancer cells. These results indicate the potential use of CD226-stimulated NK-92 cells in immunotherapy.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Granzimas/farmacología , Granzimas/uso terapéutico , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Células Asesinas Naturales , Apoptosis , Proliferación Celular
7.
Hematology ; 28(1): 2166333, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36651499

RESUMEN

OBJECTIVE: This experiment will explore the role of TIGIT/PVR signaling pathway in the pathogenesis of MDS immune tolerance through in vitro co-culture of NK cells and MDSC cells. METHODS: Flow cytometry was used to detect the expression percentage of MDSCs and CD155 on MDSCs in the bone marrow of MDS patients and controls. The expression of NK cell surface receptors (NKG2D, NKp30, NKp46), secreted cytokines (perforin, granzyme B, CD107a, IFN-γ) and NK cell apoptosis rate were detected by flow cytometry to evaluate the effect of MDSCs on NK cell function. RESULTS: The number of MDSCs in bone marrow of MDS patients was notably higher than that of the control group (8.39 ± 7.01 vs 2.31 ± 1.65, P = 0.0001). Compared with the control group, the expression of CD155 on MDSCs in MDS group was increased (31.81 ± 21.33 vs. 10.49 ± 6.53, P < 0.0001). After NK cells were co-cultured with MDSCs, NKG2D, NKp30, NKp46, CD107a, IFN-γ, perforin and granzyme B were decreased, and the NK function partially recovered after the addition of inhibitors. CONCLUSION: Compared with the normal control, MDSCs and CD155 on MDSCs were highly expressed in MDS patients. After co-culture with MDSCs, the expression of NK cells' surface receptors decreased, the secretion of cytokines decreased and the apoptosis rate increased. After blocking TIGIT/CD155 pathway, NK cell function was reversed, but NK cell apoptosis was not reduced.


Asunto(s)
Síndromes Mielodisplásicos , Células Supresoras de Origen Mieloide , Humanos , Células Supresoras de Origen Mieloide/metabolismo , Granzimas/metabolismo , Granzimas/farmacología , Perforina/metabolismo , Perforina/farmacología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Células Asesinas Naturales , Síndromes Mielodisplásicos/metabolismo , Citocinas , Receptores Inmunológicos/metabolismo
8.
J Gerontol A Biol Sci Med Sci ; 78(3): 414-423, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36006802

RESUMEN

Natural killer (NK) cells are enriched in the central nervous system in aging-related atheriosclerotic cerebral small vessel disease (aCSVD), but their roles and underlying mechanism remain to be elucidated. To identify potential cytotoxic molecules released by NK cells in aCSVD lesions, proteomic analysis of cerebrospinal fluid (CSF), plasma, and peripheral NK cells from patients with aCSVD were performed. We found that integrin ß2 (ITGB2), cathepsin D (CTSD), and granzyme H (GZMH) were highly expressed in NK cells. ITGB2 interacted with intercellular adhesion molecule 1 in vascular endothelial cells. As assessed by immunofluorescence and scanning electron microscopy of the blood-brain barrier model, transwell membranes covered with primary human brain microvascular endothelial cells and astrocytes, we demonstrated that the CTSD-mediated degradation of collagen in the blood-brain barrier depended on the cytotoxicity of NK cells in aCSVD. With the immunostaining in vitro and in vivo, GZMH disruption of demyelinated nerve fibers was reversed by cotreatment with the inhibitor 3,4-DCIC during white matter hyperintensity (WMH) in aCSVD. Our results indicate that NK cells contribute to CTSD-induced damage to the blood-brain barrier and GZMH-induced disruption of nerve fibers during WMH in aCSVD.


Asunto(s)
Células Endoteliales , Proteómica , Humanos , Células Endoteliales/metabolismo , Granzimas/metabolismo , Granzimas/farmacología , Células Asesinas Naturales
9.
Biometals ; 36(3): 617-627, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36136256

RESUMEN

Lactoferrin (LTF) is a natural iron-binding protein with a potential for clinical utility in many human immune disorders, including cancer. A fusion of LTF with the Fc domain of IgG2 (FcLTF) was designed with inherent properties of an extended the half-life in circulation. Furthermore, the effects of LTF and FcLTF were assessed for influence on the activity of natural killer (NK) cells isolated from human peripheral blood, on the NK-92 cell line, and on human monocytes. The NK cytotoxic activity induced by LTF and FcLTF was determined against the human leukemia K562 cell line, and also for monocytes, by measuring TNFα and granzyme B production, and in an assay for Jurkat cell viability. Selected gene expression in NK-92 cells and monocytes, induced by LTF and FcLTF, was performed by Real Time PCR. No significant difference was observed in NK-92 cytotoxicity stimulated by LTF and FcLTF. The effects on NK cells isolated from the human peripheral blood were varied, possibly due to the immunoregulatory nature of LTF sensing the immune status of donors. Furthermore, only the FcLTF group strongly stimulated production of TNFα and granzyme B in isolated monocytes. In addition, only supernatants from the monocyte cultures treated with FcLTF decreased the viability of Jurkat cells. The ability of FcLTF to induce TNFα in monocytes was strongly inhibited by anti-CD32 and moderately inhibited by anti-CD14 antibody. Lastly, it was demonstrated that FcLTF, strongly induced expression of PI3K, with subsequent activation of AKT/mTOR signaling pathway. Overall, it was demonstrated that this novel fusion molecule may be a perferred choice for clinical utility than the wild type LTF.


Asunto(s)
Antineoplásicos , Lactoferrina , Humanos , Lactoferrina/farmacología , Lactoferrina/metabolismo , Granzimas/genética , Granzimas/metabolismo , Granzimas/farmacología , Factor de Necrosis Tumoral alfa , Antineoplásicos/farmacología , Monocitos , Inmunoglobulina G/farmacología , Inmunoglobulina G/metabolismo
10.
Biomed Phys Eng Express ; 8(6)2022 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-36327448

RESUMEN

Granzymes-based immunotherapy for the treatment of solid tumors has gained great success and played more and more important effect in clinical studies. However, the antitumor effect of Granzymes still requires improvements owing to the cell evasion and metastasis of cancer. To overcome these limitations, synergistic combinatorial anti-tumor effect of Granzyme B (GrB) and miR-21 inhibitor (miR-21i) for breast cancer therapy through a new co-delivery system was investigated in present study. GrB was covalently bonded with miR-21i by disulfide bond and encapsulated in a nanocapsule formed byin situpolymerization of N -(3-aminopropyl) methacrylamide (APM), ethylene glycol dimethacrylate (EGDMA) and 2-Methacryloyloxyethyl phosphorylcholine (MPC). The nanocapsules possessed spherical and uniform diameter size as well as pH responsiveness in various environments. MTT and flow cytometry analysis showed that a synergistic anti-proliferation and promoting apoptosis effect was achieved when the nanocapsules were added into breast cancer cell lines. More importantly, the cell evasion ability was markedly inhibited using the nanocapusles detected through transwell invasion assay. Also thein vivoanti-tumor therapeutic efficacy of GrB-miR-21i nanocapusles was evaluated in a mouse tumor model. In conclusion, the nanocapsules for simultaneously delivery of GrB and miR-21i produce a synergistic effect in human breast cancer therapy.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Nanocápsulas , Humanos , Ratones , Animales , Femenino , Granzimas/genética , Granzimas/metabolismo , Granzimas/farmacología , MicroARNs/genética , Línea Celular Tumoral , Neoplasias de la Mama/terapia
11.
Life Sci Alliance ; 5(11)2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36271507

RESUMEN

CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4- but not of 5T4-IFNGR1- tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Humanos , Anticuerpos Biespecíficos/farmacología , Linfocitos T CD8-positivos , Granzimas/farmacología , Complejo CD3/farmacología , Citotoxicidad Inmunológica , Perforina/farmacología , Receptor de Muerte Celular Programada 1 , Neoplasias/tratamiento farmacológico , Citocinas
12.
Int J Biol Macromol ; 219: 1034-1046, 2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-35963357

RESUMEN

Chinese yam polysaccharides (CYP) exhibit superior adjuvant activity and modulate the immune response, but the low bioavailability limits their clinical application. Pickering emulsions have been proven as an efficient vaccine delivery system to enhance the immune response. Here, we used the Chinese yam polysaccharides PLGA-stabilized Pickering emulsion adjuvant system (CYP-PPAS) loaded with Porcine circovirus 2 as a vaccine and focused on investigating its adjuvant activity on humoral and cellular immunity in mice. The CYP-PPAS increased PCV-2 antigen loading efficiency and showed a high antigen uptake efficiency by macrophages in vitro. In vivo, CYP-PPAS significantly facilitated DCs maturation in draining lymph nodes than CYP or PPAS alone group. The CYP-PPAS also induced an increased proliferation index and a CD4+/CD8+ ratio. Meanwhile, in contrast to the CYP and PPAS groups, CYP-PPAS elicited a stronger anti-PCV-2 IgG and mixed Th1/Th2 immune response. Specifically, the CYP-PPAS group displayed the high expression of CD107a, FasL, and Granzyme B secretion to augment a strong cytotoxic lymphocyte response. Overall, the CYP-PPAS was a successful adjuvant system for promoting humoral and cellular immune responses, which opens up an avenue for the development of effective adjuvants against infectious diseases.


Asunto(s)
Dioscorea , Vacunas , Adyuvantes Inmunológicos/farmacología , Adyuvantes Farmacéuticos/farmacología , Animales , Emulsiones/farmacología , Granzimas/farmacología , Inmunidad Celular , Inmunoglobulina G/farmacología , Ratones , Polisacáridos/farmacología , Porcinos
13.
Biotechnol Prog ; 38(5): e3279, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35661450

RESUMEN

As a water-soluble macromolecule polysaccharide, xanthan gum (XG) has several biological activities, such as antitumor, antiviral, and immunomodulatory function. However, the effect of XG on the proliferation and cytotoxicity of cytokines induced killer (CIK) cells is rarely studied. In this study, the effect of XG on CIK cells derived from peripheral blood was investigated by analyzing the expansion fold of total cells, phenotype, cytotoxicity, degranulation, and apoptosis in serum-free medium. The results showed that the expansion fold of total cells with 100 µg/ml XG which molecule weight is 2.95 × 106 Da reached 4534.0 folds, significantly higher than that without XG (1299.0 folds, p < 0.05). The percentage of main effector cells-CD3+ CD56+ cells increased to 25.5% and the cytotoxic activity of CIK cells increased to 45.3%. The cell proportions of expression granzyme B and perforin that related to cytotoxicity in CIK cells reached 53.6% and 48.3%, respectively, significantly higher than 27.5% and 37.5% in the group without XG (p < 0.05). Collectively, XG could stimulate the ex vivo expansion of CIK cells and enhance the cytotoxicity of expanded CIK cells. The above results provide technical support for optimizing the expansion process of CIK cells ex vivo.


Asunto(s)
Células Asesinas Inducidas por Citocinas , Antivirales/farmacología , Células Cultivadas , Células Asesinas Inducidas por Citocinas/metabolismo , Granzimas/metabolismo , Granzimas/farmacología , Perforina/metabolismo , Perforina/farmacología , Polisacáridos Bacterianos , Factor de Necrosis Tumoral alfa , Agua/metabolismo
14.
Front Immunol ; 12: 712678, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34413857

RESUMEN

Mycobacterium tuberculosis (Mtb), the pathological agent that causes tuberculosis (TB) is the number one infectious killer worldwide with one fourth of the world's population currently infected. Data indicate that γ9δ2 T cells secrete Granzyme A (GzmA) in the extracellular space triggering the infected monocyte to inhibit growth of intracellular mycobacteria. Accordingly, deletion of GZMA from γ9δ2 T cells reverses their inhibitory capacity. Through mechanistic studies, GzmA's action was investigated in monocytes from human PBMCs. The use of recombinant human GzmA expressed in a mammalian system induced inhibition of intracellular mycobacteria to the same degree as previous human native protein findings. Our data indicate that: 1) GzmA is internalized within mycobacteria-infected cells, suggesting that GzmA uptake could prevent infection and 2) that the active site is not required to inhibit intracellular replication. Global proteomic analysis demonstrated that the ER stress response and ATP producing proteins were upregulated after GzmA treatment, and these proteins abundancies were confirmed by examining their expression in an independent set of patient samples. Our data suggest that immunotherapeutic host interventions of these pathways may contribute to better control of the current TB epidemic.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Estrés del Retículo Endoplásmico/inmunología , Granzimas/fisiología , Monocitos/microbiología , Mycobacterium bovis/fisiología , Subgrupos de Linfocitos T/inmunología , Western Blotting , División Celular , Granzimas/biosíntesis , Granzimas/genética , Granzimas/farmacología , Células HEK293 , Humanos , Células T de Memoria/inmunología , Células T de Memoria/metabolismo , Proteoma , Receptores de Antígenos de Linfocitos T gamma-delta/análisis , Proteínas Recombinantes/farmacología , Subgrupos de Linfocitos T/metabolismo , Electroforesis Bidimensional Diferencial en Gel
15.
Nat Nanotechnol ; 16(10): 1130-1140, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34385682

RESUMEN

Despite the enormous therapeutic potential of immune checkpoint blockade (ICB), it benefits only a small subset of patients. Some chemotherapeutics can switch 'immune-cold' tumours to 'immune-hot' to synergize with ICB. However, safe and universal therapeutic platforms implementing such immune effects remain scarce. We demonstrate that sphingomyelin-derived camptothecin nanovesicles (camptothesomes) elicit potent granzyme-B- and perforin-mediated cytotoxic T lymphocyte (CTL) responses, potentiating PD-L1/PD-1 co-blockade to eradicate subcutaneous MC38 adenocarcinoma with developed memory immunity. In addition, camptothesomes improve the pharmacokinetics and lactone stability of camptothecin, avoid systemic toxicities, penetrate deeply into the tumour and outperform the antitumour efficacy of Onivyde. Camptothesome co-load the indoleamine 2,3-dioxygenase inhibitor indoximod into its interior using the lipid-bilayer-crossing capability of the immunogenic cell death inducer doxorubicin, eliminating clinically relevant advanced orthotopic CT26-Luc tumours and late-stage B16-F10-Luc2 melanoma, and achieving complete metastasis remission when combined with ICB and folate targeting. The sphingomyelin-derived nanotherapeutic platform and doxorubicin-enabled transmembrane transporting technology are generalizable to various therapeutics, paving the way for transformation of the cancer immunochemotherapy paradigm.


Asunto(s)
Camptotecina/farmacología , Quimioterapia , Inmunoterapia , Nanopartículas/química , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Camptotecina/química , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Granzimas/química , Granzimas/farmacología , Humanos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/farmacología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Perforina/química , Perforina/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Esfingomielinas/química , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
16.
Molecules ; 25(13)2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32646038

RESUMEN

The human immune system is a complex system which protects against invaders and maintains tissue homeostasis. It is broadly divided into the innate and adaptive branches. Granzyme B is serine protease that plays an important role in both and can serve as a biomarker for cellular activation. Because of this, a granzyme B PET agent (GZP) has recently been developed and has been shown to be able to monitor response to immunotherapy. Here, we evaluated the utility of granzyme B PET imaging to assess the innate immune response. We subcutaneously administered LPS to mice to induce inflammation and performed granzyme B PET imaging after 24 and 120 h. We dissected out tissue in the region of injection and performed granzyme B immunofluorescence (IF) to confirm specificity of the GZP radiotracer. Granzyme B PET imaging demonstrated increased uptake in the region of LPS injection after 24 h, which normalized at 120 h. Granzyme B immunofluorescence showed specific staining in tissue from the 24 h time point compared to the PBS-injected control. These findings support the use of granzyme B PET for imaging innate immunity. In certain clinical contexts, the use of GZP PET imaging may be superior to currently available agents, and we therefore suggest further preclinical studies with the ultimate goal of translation to clinical use.


Asunto(s)
Granzimas , Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos , Tomografía de Emisión de Positrones , Animales , Granzimas/química , Granzimas/farmacología , Inflamación/inducido químicamente , Inflamación/diagnóstico por imagen , Inflamación/inmunología , Ratones , Ratones Mutantes
17.
Cell Death Dis ; 10(10): 695, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31541080

RESUMEN

Cytotoxic T lymphocytes (CTL) and natural killer cells (NK)-mediated elimination of tumor cells is mostly dependent on Granzyme B apoptotic pathway, which is regulated by the wild type (wt) p53 protein. Because TP53 inactivating mutations, frequently found in human tumors, could interfere with Granzyme B-mediated cell death, the use of small molecules developed to reactivate wtp53 function in p53-mutated tumor cells could optimize their lysis by CTL or NK cells. Here, we show that the pharmalogical reactivation of a wt-like p53 function in p53-mutated breast cancer cells using the small molecule CP-31398 increases their sensitivity to NK-mediated lysis. This potentiation is dependent on p53-mediated induction of autophagy via the sestrin-AMPK-mTOR pathway and the ULK axis. This CP31398-induced autophagy sequestrates in autophagosomes several anti-apoptotic proteins, including Bcl-XL and XIAP, facilitating Granzyme B-mediated mitochondrial outer membrane permeabilization, caspase-3 activation and Granzyme B- or NK cell-induced apoptosis. Together, our results define a new way to increase cytotoxic lymphocyte-mediated lysis of p53-mutated breast cancer cell, through a p53-dependent autophagy induction, with potential applications in combined immunotherapeutic approaches.


Asunto(s)
Granzimas/farmacología , Células Asesinas Naturales/inmunología , Proteína p53 Supresora de Tumor/inmunología , Animales , Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Granzimas/metabolismo , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Ratones , Perforina/farmacología , Pirimidinas/farmacología , Transducción de Señal , Transfección , Proteína p53 Supresora de Tumor/metabolismo
18.
Front Immunol ; 10: 1450, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293597

RESUMEN

Granzyme A (GzmA), together with perforin, are well-known for their cytotoxic activity against tumor or virus-infected cells. In addition to this cytotoxic function, GzmA stimulates several immune cell types and induces inflammation in the absence of perforin, however, its effect on the dendritic cell (DC) is unknown. In the current study, we showed that recombinant GzmA induced the phenotypic maturation of plasmacytoid DCs (pDCs) and conventional DCs (cDCs), but not their apoptosis. Particularly, GzmA made pDCs more functional, thus leading to production of type I interferon (IFN) via the TLR9-MyD88 pathway. We also demonstrated that GzmA binds TLR9 and co-localizes with it in endosomes. When co-administered with antigen, GzmA acted as a powerful adjuvant for eliciting antigen-specific cytotoxic CD8+ T lymphocytes (CTLs) that protected mice from tumor challenge. The induction of CTL was completely abolished in XCR1+ DC-depleted mice, whereas it was reduced to less than half in pDC-depleted or IFN-α/ß receptor knockout mice. Thus, CTL cross-priming was dependent on XCR1+cDC and also type I IFN, which was produced by GzmA-activated pDCs. These results indicate that GzmA -stimulated pDCs enhance the cross-priming activity of cDCs in situ. We also showed that the adjuvant effect of GzmA is superior to CpG-ODN and LPS. Our findings highlight the ability of GzmA to bridge innate and adaptive immune responses via pDC help and suggest that GzmA may be useful as a vaccine adjuvant.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Granzimas/farmacología , Inmunidad Celular/efectos de los fármacos , Células Plasmáticas/inmunología , Animales , Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Granzimas/genética , Granzimas/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Células Plasmáticas/citología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
19.
Proc Natl Acad Sci U S A ; 116(4): 1361-1369, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30545915

RESUMEN

Interleukin-1ß (IL-1ß) is abundant in the tumor microenvironment, where this cytokine can promote tumor growth, but also antitumor activities. We studied IL-1ß during early tumor progression using a model of orthotopically introduced 4T1 breast cancer cells. Whereas there is tumor progression and spontaneous metastasis in wild-type (WT) mice, in IL-1ß-deficient mice, tumors begin to grow but subsequently regress. This change is due to recruitment and differentiation of inflammatory monocytes in the tumor microenvironment. In WT mice, macrophages heavily infiltrate tumors, but in IL-1ß-deficient mice, low levels of the chemokine CCL2 hamper recruitment of monocytes and, together with low levels of colony-stimulating factor-1 (CSF-1), inhibit their differentiation into macrophages. The low levels of macrophages in IL-1ß-deficient mice result in a relatively high percentage of CD11b+ dendritic cells (DCs) in the tumors. In WT mice, IL-10 secretion from macrophages is dominant and induces immunosuppression and tumor progression; in contrast, in IL-1ß-deficient mice, IL-12 secretion by CD11b+ DCs prevails and supports antitumor immunity. The antitumor immunity in IL-1ß-deficient mice includes activated CD8+ lymphocytes expressing IFN-γ, TNF-α, and granzyme B; these cells infiltrate tumors and induce regression. WT mice with 4T1 tumors were treated with either anti-IL-1ß or anti-PD-1 Abs, each of which resulted in partial growth inhibition. However, treating mice first with anti-IL-1ß Abs followed by anti-PD-1 Abs completely abrogated tumor progression. These data define microenvironmental IL-1ß as a master cytokine in tumor progression. In addition to reducing tumor progression, blocking IL-1ß facilitates checkpoint inhibition.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Antígeno CD11b/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Factores Estimulantes de Colonias/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Granzimas/farmacología , Humanos , Terapia de Inmunosupresión/métodos , Inflamación/metabolismo , Interferón gamma/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Microambiente Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
20.
Clin Cancer Res ; 25(4): 1196-1205, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30327313

RESUMEN

PURPOSE: The lack of a timely and reliable measure of response to cancer immunotherapy has confounded understanding of mechanisms of resistance and subsequent therapeutic advancement. We hypothesized that PET imaging of granzyme B using a targeted peptide, GZP, could be utilized for early response assessment across many checkpoint inhibitor combinations, and that GZP uptake could be compared between therapeutic regimens and dosing schedules as an early biomarker of relative efficacy. EXPERIMENTAL DESIGN: Two models, MC38 and CT26, were treated with a series of checkpoint inhibitors. GZP PET imaging was performed to assess tumoral GZP uptake, and tumor volume changes were subsequently monitored to determine response. The average GZP PET uptake and response of each treatment group were correlated to evaluate the utility of GZP PET for comparing therapeutic efficacy. RESULTS: In both tumor models, GZP PET imaging was highly accurate for predicting response, with 93% sensitivity and 94% negative predictive value. Mean tumoral GZP signal intensity of treatment groups linearly correlated with percent response across all therapies and schedules. Moreover, GZP PET correctly predicted that sequential dose scheduling of PD-1 and CTLA-4 targeted therapies demonstrates comparative efficacy to concurrent administration. CONCLUSIONS: Granzyme B quantification is a highly sensitive and specific early measure of therapeutic efficacy for checkpoint inhibitor regimens. This work provides evidence that GZP PET imaging may be useful for rapid assessment of therapeutic efficacy in the context of clinical trials for both novel drugs as well as dosing regimens.


Asunto(s)
Neoplasias del Colon/diagnóstico por imagen , Genes cdc/efectos de los fármacos , Granzimas/farmacología , Inmunoterapia , Animales , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Granzimas/genética , Xenoinjertos , Humanos , Ratones , Tomografía de Emisión de Positrones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...