Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Eur Respir J ; 55(2)2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31744834

RESUMEN

Stratification of asthmatic patients based on relevant biomarkers enables the prediction of responsiveness against immune-targeted therapies in patients with asthma. Individualised therapy in patients with eosinophilic asthma has yielded improved clinical outcomes; similar approaches in patients with neutrophilic asthma have yet to be developed. We determined whether colony-stimulating factors (CSFs) in the airway reflect the inflammatory phenotypes of asthma and contribute to disease progression of neutrophilic asthma.We analysed three different mouse models of asthma and assessed cytokine profiles in sputum from human patients with asthma stratified according to inflammatory phenotype. In addition, we evaluated the therapeutic efficacy of various cytokine blockades in a mouse model of neutrophilic asthma.Among the CSFs, airway granulocyte CSF (G-CSF) contributes to airway neutrophilia by promoting neutrophil development in bone marrow and thereby distinguishes neutrophilic inflammation from eosinophilic inflammation in mouse models of asthma. G-CSF is produced by concurrent stimulation of the lung epithelium with interleukin (IL)-17A and tumour necrosis factor (TNF)-α; therefore, dual blockade of upstream stimuli using monoclonal antibodies or genetic deficiency of the cytokines in IL-17A×TNF-α double-knockout mice reduced the serum level of G-CSF, leading to alleviation of neutrophilic inflammation in the airway. In humans, the sputum level of G-CSF can be used to stratify patients with asthma with neutrophil-dominated inflammation.Our results indicated that myelopoiesis-promoting G-CSF and cytokines as the upstream inducing factors are potential diagnostic and therapeutic targets in patients with neutrophilic asthma.


Asunto(s)
Asma , Asma/tratamiento farmacológico , Humanos , Inflamación , Pulmón , Neutrófilos , Esputo
2.
J Virol ; 93(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31341044

RESUMEN

Proteoglycans function not only as structural components of the extracellular compartment but also as regulators of various cellular events, including cell migration, inflammation, and infection. Many microbial pathogens utilize proteoglycans to facilitate adhesion and invasion into host cells. Here we report a secreted form of a novel heparan sulfate proteoglycan that functions against virus infection. The expression of SPOCK2/testican-2 was significantly induced in virus-infected lungs or in interferon (IFN)-treated alveolar lung epithelial cells. Overexpression from a SPOCK2 expression plasmid alone or the treatment of cells with recombinant SPOCK2 protein efficiently blocked influenza virus infection at the step of viral attachment to the host cell and entry. Moreover, mice treated with purified SPOCK2 were protected against virus infection. Sialylated glycans and heparan sulfate chains covalently attached to the SPOCK2 core protein were critical for its antiviral activity. Neuraminidase (NA) of influenza virus cleaves the sialylated moiety of SPOCK2, thereby blocking its binding to the virus. Our data suggest that IFN-induced SPOCK2 functions as a decoy receptor to bind and block influenza virus infection, thereby restricting entry of the infecting virus into neighboring cells.IMPORTANCE Here we report a novel proteoglycan protein, testican-2/SPOCK2, that prevents influenza virus infection. Testican-2/SPOCK2 is a complex type of secreted proteoglycan with heparan sulfate GAG chains attached to the core protein. SPOCK2 expression is induced upon virus infection or by interferons, and the protein is secreted to an extracellular compartment, where it acts directly to block virus-cell attachment and entry. Treatment with purified testican-2/SPOCK2 protein can efficiently block influenza virus infection in vitro and in vivo We also identified the heparan sulfate moiety as a key regulatory module for this inhibitory effect. Based on its mode of action (cell attachment/entry blocker) and site of action (extracellular compartment), we propose testican-2/SPOCK2 as a potential antiviral agent that can efficiently control influenza virus infection.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Resistencia a la Enfermedad/genética , Interacciones Huésped-Patógeno/genética , Proteoglicanos/genética , Virosis/etiología , Virosis/metabolismo , Animales , Antivirales/farmacología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Proteoglicanos de Heparán Sulfato , Humanos , Virus de la Influenza A/efectos de los fármacos , Ratones , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , Proteínas Recombinantes/farmacología , Acoplamiento Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
3.
J Synchrotron Radiat ; 21(Pt 2): 424-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24562565

RESUMEN

Key parameters of two-phase flows, such as void fraction and microscale bubble size, shape and velocity, were simultaneously measured using time-resolved X-ray imaging. X-ray phase-contrast imaging was employed to obtain those parameters on microbubbles. The void fraction was estimated from X-ray absorption. The radii of the measured microbubbles were mostly smaller than 20 µm, and the maximum velocity was 39.442 mm s(-1), much higher than that in previous studies. The spatial variations of the void fraction were consecutively obtained with a small time interval. This technique would be useful in the experimental analysis of bubbly flows in which microbubbles move at high speed.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Microburbujas , Sincrotrones , Espectroscopía de Absorción de Rayos X/métodos , Algoritmos , Diagnóstico por Imagen/métodos , Calor , Reología , Tiempo , Agua , Difracción de Rayos X
4.
J Synchrotron Radiat ; 21(Pt 5): 1160-6, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25178007

RESUMEN

X-ray imaging techniques have been employed to visualize various biofluid flow phenomena in a non-destructive manner. X-ray particle image velocimetry (PIV) was developed to measure velocity fields of blood flows to obtain hemodynamic information. A time-resolved X-ray PIV technique that is capable of measuring the velocity fields of blood flows under real physiological conditions was recently developed. However, technical limitations still remained in the measurement of blood flows with high image contrast and sufficient biocapability. In this study, CO2 microbubbles as flow-tracing contrast media for X-ray PIV measurements of biofluid flows was developed. Human serum albumin and CO2 gas were mechanically agitated to fabricate CO2 microbubbles. The optimal fabricating conditions of CO2 microbubbles were found by comparing the size and amount of microbubbles fabricated under various operating conditions. The average size and quantity of CO2 microbubbles were measured by using a synchrotron X-ray imaging technique with a high spatial resolution. The quantity and size of the fabricated microbubbles decrease with increasing speed and operation time of the mechanical agitation. The feasibility of CO2 microbubbles as a flow-tracing contrast media was checked for a 40% hematocrit blood flow. Particle images of the blood flow were consecutively captured by the time-resolved X-ray PIV system to obtain velocity field information of the flow. The experimental results were compared with a theoretically amassed velocity profile. Results show that the CO2 microbubbles can be used as effective flow-tracing contrast media in X-ray PIV experiments.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Dióxido de Carbono , Medios de Contraste , Hemorreología/fisiología , Microburbujas , Estudios de Factibilidad , Hematócrito , Humanos , Albúmina Sérica , Rayos X
5.
J Synchrotron Radiat ; 20(Pt 3): 498-503, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23592630

RESUMEN

X-ray imaging is used to visualize the biofluid flow phenomena in a nondestructive manner. A technique currently used for quantitative visualization is X-ray particle image velocimetry (PIV). Although this technique provides a high spatial resolution (less than 10 µm), significant hemodynamic parameters are difficult to obtain under actual physiological conditions because of the limited temporal resolution of the technique, which in turn is due to the relatively long exposure time (~10 ms) involved in X-ray imaging. This study combines an image intensifier with a high-speed camera to reduce exposure time, thereby improving temporal resolution. The image intensifier amplifies light flux by emitting secondary electrons in the micro-channel plate. The increased incident light flux greatly reduces the exposure time (below 200 µs). The proposed X-ray PIV system was applied to high-speed blood flows in a tube, and the velocity field information was successfully obtained. The time-resolved X-ray PIV system can be employed to investigate blood flows at beamlines with insufficient X-ray fluxes under specific physiological conditions. This method facilitates understanding of the basic hemodynamic characteristics and pathological mechanism of cardiovascular diseases.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Fotograbar/instrumentación , Radiografía/instrumentación , Reología/instrumentación , Diseño de Equipo , Análisis de Falla de Equipo
6.
Sci Adv ; 7(3)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523913

RESUMEN

Cancer immunotherapy with 4-1BB agonists has limited further clinical development because of dose-limiting toxicity. Here, we developed a bispecific antibody (bsAb; B7-H3×4-1BB), targeting human B7-H3 (hB7-H3) and mouse or human 4-1BB, to restrict the 4-1BB stimulation in tumors. B7-H3×m4-1BB elicited a 4-1BB-dependent antitumor response in hB7-H3-overexpressing tumor models without systemic toxicity. BsAb primarily targets CD8 T cells in the tumor and increases their proliferation and cytokine production. Among the CD8 T cell population in the tumor, 4-1BB is solely expressed on PD-1+Tim-3+ "terminally differentiated" subset, and bsAb potentiates these cells for eliminating the tumor. Furthermore, the combination of bsAb and PD-1 blockade synergistically inhibits tumor growth accompanied by further increasing terminally differentiated CD8 T cells. B7-H3×h4-1BB also shows antitumor activity in h4-1BB-expressing mice. Our data suggest that B7-H3×4-1BB is an effective and safe therapeutic agent against B7-H3-positive cancers as monotherapy and combination therapy with PD-1 blockade.


Asunto(s)
Anticuerpos Biespecíficos , Linfocitos T CD8-positivos , Linfocitos Infiltrantes de Tumor , Neoplasias , Animales , Anticuerpos Biespecíficos/farmacología , Linfocitos T CD8-positivos/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1
7.
Clin Transl Immunology ; 9(9): e1168, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32994996

RESUMEN

OBJECTIVES: Emerging oncotherapeutic strategies require the induction of an immunostimulatory tumor microenvironment (TME) containing numerous tumor-reactive CD8+ T cells. Interleukin-7 (IL-7), a T-cell homeostatic cytokine, induces an antitumor response; however, the detailed mechanisms underlying the contributions of the IL-7 to TME remain unclear. Here, we aimed to investigate the mechanism underlying the induction of antitumor response by hybrid Fc-fused long-acting recombinant human IL-7 (rhIL-7-hyFc) through regulation of both adaptive and innate immune cells in the TME. METHODS: We evaluated rhIL-7-hyFc-mediated antitumor responses in murine syngeneic tumor models. We analysed the cellular and molecular features of tumor-infiltrating lymphocytes (TILs) and changes in the TME after rhIL-7-hyFc treatment. Furthermore, we evaluated the antitumor efficacy of rhIL-7-hyFc combined with chemotherapy and checkpoint inhibitors (CPIs). RESULTS: Systemic delivery of rhIL-7-hyFc induced significant therapeutic benefits by expanding CD8+ T cells with enhanced tumor tropism. In tumors, rhIL-7-hyFc increased both tumor-reactive and bystander CD8+ TILs, all of which displayed enhanced effector functions but less exhausted phenotypes. Moreover, rhIL-7-hyFc suppressed the generation of immunosuppressive myeloid cells in the bone marrow of tumor-bearing mice, resulting in the immunostimulatory TME. Combination therapy with chemotherapy and CPIs, rhIL-7-hyFc elicited a strong antitumor response and even under a T lymphopenic condition by restoring CD8+ T cells. When combined with chemotherapy and CPIs, rhIL-7-hyFc administration enhanced antitumor response under intact andlymphopenic conditions by restoring CD8+ T cells. CONCLUSION: Taken together, these data demonstrate that rhIL-7-hyFc induces antitumor responses by generating T-cell-inflamed TME and provide a preclinical proof of concept of immunotherapy with rhIL-7-hyFc to enhance therapeutic responses in the clinic.

8.
Immune Netw ; 19(6): e43, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31921473

RESUMEN

A full-length translational product of the trophinin gene, KIAA1114, is a distinctive marker of cancer stem cells in human hepatocellular carcinoma, and a mAb, Kiatomab, is specific to KIAA1114 antigen. In this study, we addressed the therapeutic potential of Kiatomab for treating both metastatic and solid tumors in mouse models. Kiatomab recognizes the linear epitope of KIAA1114, which is expressed on cell surfaces of various murine cancer cell lines. Kiatomab treatment induced potent antitumor responses in pulmonary metastasis models. Antitumor activity was mediated by the fragment crystallizable portion of Kiatomab and dependent on the host immune system. The use of Kiatomab alone as an antitumor therapy was ineffective in solid tumor models. However, in combination with cyclophosphamide, or by switching the isotype of the mAb, improved antitumor effects of Kiatomab were observed. These results suggest that Kiatomab can be used as a novel mAb for cancer immunotherapy.

9.
PLoS One ; 14(5): e0217691, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31150476

RESUMEN

By comparing and measuring covariations of viral protein sequences from isolates of the 2009 pH1N1 influenza A virus (IAV), specific substitutions that co-occur in the NP-NA pair were identified. To investigate the effect of these co-occurring substitution pairs, the V100I substitution in NP and the D248N substitution in NA were introduced into laboratory-adapted WSN IAVs. The recombinant WSN with the covarying NPV100I-NAD248N pair exhibited enhanced pathogenicity, as characterized by increased viral production, increased death and inflammation of host cells, and high mortality in infected mice. Although direct interactions between the NPV100I and NAD248N proteins were not detected, the RNA-binding ability of NPV100I was increased, which was further strengthened by NAD248N, in expression-plasmid-transfected cells. Additionally, the NAD248N protein was frequently recruited within lipid rafts, indirectly affecting the RNA-binding ability of NP as well as viral release. Altogether, our data indicate that the covarying NPV100I-NAD248N pair obtained from 2009 pH1N1 IAV sequence information function together to synergistically augment viral assembly and release, which may explain the observed enhanced viral pathogenicity.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/genética , Proteínas del Núcleo Viral/genética , Proteínas Virales/genética , Animales , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Ratones , Replicación Viral/genética
10.
Immune Netw ; 17(5): 343-351, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29093655

RESUMEN

Developing a novel vaccine that can be applied against multiple strains of influenza virus is of utmost importance to human health. Previously, we demonstrated that the intranasal introduction of Fc-fused IL-7 (IL-7-mFc), a long-acting cytokine fusion protein, confers long-lasting prophylaxis against multiple strains of influenza A virus (IAV) by inducing the development of lung-resident memory-like T cells, called TRM-like cells. Here, we further investigated the mechanisms of IL-7-mFc-mediated protective immunity to IAVs. First, we found that IL-7-mFc treatment augments the accumulation of pulmonary T cells in 2 ways: recruiting blood circulating T cells into the lung and expanding T cells at the lung parenchyma. Second, the blockade of T cell migration from the lymph nodes (LNs) with FTY720 treatment was not required for mounting the protective immunity to IAV with IL-7-mFc, suggesting a more important role of IL-7 in T cells in the lungs. Third, IL-7-mFc treatment also recruited various innate immune cells into the lungs. Among these cells, plasmacytoid dendritic cells (pDCs) play an important role in IL-7-mFc-mediated protective immunity through reducing the immunopathology and increasing IAV-specific cytotoxic T lymphocyte (CTL) responses. In summary, our results show that intranasal treatment with IL-7-mFc modulates pulmonary immune responses to IAV, affecting both innate and adaptive immune cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA