Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Nat Commun ; 15(1): 5161, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886405

RESUMEN

Top emission can enhance luminance, color purity, and panel-manufacturing compatibility for emissive displays. Still, top-emitting quantum-dot light-emitting diodes (QLEDs) suffer from poor stability, low light outcoupling, and non-negligible viewing-angle dependence because, for QLEDs with non-red emission, the electrically optimum device structure is incompatible with single-mode optical microcavity. Here, we demonstrate that by improving the way of determining reflection penetration depths and creating refractive-index-lowering processes, the issues faced by green QLEDs can be overcome. This leads to advanced device performance, including a luminance exceeding 1.6 million nits, a current efficiency of 204.2 cd A-1, and a T95 operational lifetime of 15,600 hours at 1000 nits. Meanwhile, our design does not compromise light outcoupling as it offers an external quantum efficiency of 29.2% without implementing light extraction methods. Lastly, an angular color shift of Δu'v' = 0.0052 from 0° to 60° is achieved by narrowing the emission linewidth of quantum dots.

2.
J Immunother Cancer ; 12(1)2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38199607

RESUMEN

BACKGROUND: The extracellular matrix (ECM) and cancer-associated fibroblasts (CAFs) play major roles in tumor progression, metastasis, and the poor response of many solid tumors to immunotherapy. CAF-targeted chimeric antigen receptor-T cell therapy cannot infiltrate ECM-rich tumors such as osteosarcoma. METHOD: In this study, we used RNA sequencing to assess whether the recently invented membrane-anchored and tumor-targeted IL-12-armed (attIL12) T cells, which bind cell-surface vimentin (CSV) on tumor cells, could destroy CAFs to disrupt the ECM. We established an in vitro model of the interaction between osteosarcoma CAFs and attIL12-T cells to uncover the underlying mechanism by which attIL12-T cells penetrate stroma-enriched osteosarcoma tumors. RESULTS: RNA sequencing demonstrated that attIL12-T cell treatment altered ECM-related gene expression. Immunohistochemistry staining revealed disruption or elimination of high-density CAFs and ECM in osteosarcoma xenograft tumors following attIL12-T cell treatment, and CAF/ECM density was inversely correlated with T-cell infiltration. Other IL12-armed T cells, such as wild-type IL-12-targeted or tumor-targeted IL-12-T cells, did not disrupt the ECM because this effect depended on the engagement between CSV on the tumor cell and its ligand on the attIL12-T cells. Mechanistic studies found that attIL12-T cell treatment elevated IFNγ production on interacting with CSV+ tumor cells, suppressing transforming growth factor beta secretion and in turn upregulating FAS-mediated CAF apoptosis. CAF destruction reshaped the tumor stroma to favor T-cell infiltration and tumor inhibition. CONCLUSIONS: This study unveiled a novel therapy-attIL12-T cells-for targeting CAFs/ECM. These findings are highly relevant to humans because CAFs are abundant in human osteosarcoma.


Asunto(s)
Neoplasias Óseas , Fibroblastos Asociados al Cáncer , Osteosarcoma , Animales , Humanos , Interleucina-12 , Xenoinjertos , Osteosarcoma/terapia , Membrana Celular , Matriz Extracelular , Modelos Animales de Enfermedad , Neoplasias Óseas/terapia , Tratamiento Basado en Trasplante de Células y Tejidos
3.
Small ; : e2307498, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38059807

RESUMEN

A front surface gradient of the absorber valence band can effectively reduce the open-circuit voltage (VOC ) loss of perovskite solar cells by suppressing the minority carrier concentration near the front surface. However, the existing method is limited to the one-step fabrication process, resulting in underachieved photon harvesting and power conversion efficiency (PCE). To solve the problem, ZnCd-based alloy quantum dots (QDs) are utilized to create a valence-band-maximum gradient at the front surface of a two-step processed FAPbI3 absorber. This design significantly enhances VOC without requiring surface passivation. Furthermore, it is demonstrated that reducing the QD-perovskite lattice mismatch while maintaining QD's energy levels mitigates nonradiative recombination without compromising the front surface gradient effect. As a result, normal-structured perovskite solar cells achieve a VOC equivalent to 93% of the Schockley-Queisser limit and a PCE of 24.37%.

4.
Cancers (Basel) ; 15(15)2023 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-37568669

RESUMEN

Tumors of the central nervous system (CNS) are the most common and lethal childhood malignancy. Detection of residual disease and longitudinal monitoring of treatment response in patients are challenging and rely on serial imaging. This current standard of care fails to detect microscopic disease or provide molecular characteristics of residual tumors. As such, there is dire need for minimally invasive liquid biopsy techniques. We have previously shown the high specificity of using cell surface vimentin (CSV) to identify circulating tumor cells (CTCs) from patients bearing various types of cancers. Here, we describe the first report of CTCs captured from peripheral blood samples in 58 pediatric CNS tumor patients. In this study, we used a CSV-coated cell capture chip, the Abnova CytoQuest automated CTC isolation system, to boost the CTC capture from pediatric patients with CNS tumors. We successfully isolated CTCs in six glioma patients using immunostaining of histone H3 lysine27-to-methionine (H3K27M) mutations which are highly expressed by this tumor. We show that CSV is a viable marker for CNS CTC isolation and that this is a feasible method for detecting microscopic disease. Larger-scale studies focusing on CTCs in pediatric CNS tumors to explore their diagnostic and prognostic value are warranted.

5.
Nat Commun ; 14(1): 735, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36759517

RESUMEN

Although tissue-resident memory T (TRM) cells specific for previously encountered pathogens have been characterized, the induction and recruitment of brain TRM cells following immune therapy has not been observed in the context of glioblastoma. Here, we show that T cells expressing fibrinogen-like 2 (FGL2)-specific single-chain variable fragments (T-αFGL2) can induce tumor-specific CD8+ TRM cells that prevent glioblastoma recurrence. These CD8+ TRM cells display a highly expanded T cell receptor repertoire distinct from that found in peripheral tissue. When adoptively transferred to the brains of either immunocompetent or T cell-deficient naïve mice, these CD8+ TRM cells reject glioma cells. Mechanistically, T-αFGL2 cell treatment increased the number of CD69+CD8+ brain-resident memory T cells in tumor-bearing mice via a CXCL9/10 and CXCR3 chemokine axis. These findings suggest that tumor-specific brain-resident CD8+ TRM cells may have promising implications for the prevention of brain tumor recurrence.


Asunto(s)
Linfocitos T CD8-positivos , Glioblastoma , Animales , Ratones , Encéfalo , Glioblastoma/terapia , Memoria Inmunológica , Células T de Memoria , Recurrencia Local de Neoplasia , Linfocitos T/inmunología
6.
Front Public Health ; 11: 1289502, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38249379

RESUMEN

Background: Nursing care is essential for older adults with disabilities. Income plays a crucial role in determining the utilization of institutional care services. Pension benefit, as the main source of income for the older adults in China's cities and towns in their later years, is an important factor influencing the utilization of institutional care services. However, there have been no consistent findings on how pension benefits affect the utilization of institutional care services for the disabled older adults. Methods: This paper utilizes data from the 2017-2018 Chinese Longitudinal Healthy Longevity Survey. We select disabled older adults aged 65 and older, living in towns and cities, and use a probit regression model to investigate the impact of pension benefits on the utilization of institutional care services by urban disabled older adults empirically. Results: The study shows that a 1% increase in pension benefits raises the probability that the urban disabled older adults use institutional care services by 0.03. It also finds that for low-income urban disabled older adults, the effect is statistically significantly positive at the 1% level; but for high-income urban disabled older adults, the effect is not statistically significant. The pension benefits significantly increase the probability for the disabled older adults who are male, financially dependent, and live in townships. In addition, the pension benefits significantly reduce the probability that children will provide care and pay for care services for their older parents. Conclusion: Institutional care service is a normal good for the urban disabled older adults, especially for low-income older adults. Therefore, higher pension benefit raises the probability of utilizing institutional care services for the urban older adults with disabilities, and this positive effect is especially pronounced for older adults who are male, financially dependent, and reside in townships. In addition, increase in the pension benefits for the disabled older adults in towns and cities reduces the burden on children by reducing the probability that children will provide care and pay for care services for the older adults.


Asunto(s)
Personas con Discapacidad , Renta , Casas de Salud , Pensiones , Anciano , Femenino , Humanos , Masculino , Pueblo Asiatico , China , Estado de Salud , Población Urbana , Casas de Salud/economía , Casas de Salud/estadística & datos numéricos
7.
PLoS One ; 17(10): e0274601, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36201506

RESUMEN

Receptor-ligand binding has been analyzed at the protein level using isothermal titration calorimetry and surface plasmon resonance and at the cellular level using interaction-associated downstream gene induction/suppression. However, no currently available technique can characterize this interaction directly through visualization. In addition, all available assays require a large pool of cells; no assay capable of analyzing receptor-ligand interactions at the single-cell level is publicly available. Here, we describe a new microfluidic chip-based technique for analyzing and visualizing these interactions at the single-cell level. First, a protein is immobilized on a glass slide and a low-flow-rate pump is used to isolate cells that express receptors that bind to the immobilized ligand. Specifically, we demonstrate the efficacy of this technique by immobilizing biotin-conjugated FGL2 on an avidin-coated slide chip and passing a mixture of GFP-labeled wild-type T cells and RFP-labeled FcγRIIB-knockout T cells through the chip. Using automated scanning and counting, we found a large number of GFP+ T cells with binding activity but significantly fewer RFP+ FcγRIIB-knockout T cells. We further isolated T cells expressing a membrane-anchored, tumor-targeted IL-12 based on the receptor's affinity to vimentin to confirm the versatility of our technique. This protocol allows researchers to isolate receptor-expressing cells in about 4 hours for further downstream processing.


Asunto(s)
Avidina , Biotina , Biotina/metabolismo , Interleucina-12 , Ligandos , Microfluídica , Vimentina
8.
Clin Cancer Res ; 28(17): 3862-3873, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35727602

RESUMEN

PURPOSE: Chimeric antigen receptor (CAR) T-cell therapy has shown great promise for treating hematologic malignancies but requires a long duration of T-cell expansion, is associated with severe toxicity, and has limited efficacy for treating solid tumors. We designed experiments to address those challenges. EXPERIMENTAL DESIGN: We generated a cell membrane-anchored and tumor-targeted IL12 (attIL12) to arm peripheral blood mononuclear cells (PBMC) instead of T cells to omit the expansion phase for required CAR T cells. RESULTS: This IL12-based attIL12-PBMC therapy showed significant antitumor efficacy in both heterogeneous osteosarcoma patient-derived xenograft tumors and metastatic osteosarcoma tumors with no observable toxic effects. Mechanistically, attIL12-PBMC treatment resulted in tumor-restricted antitumor cytokine release and accumulation of attIL12-PBMCs in tumors. It also induced terminal differentiation of osteosarcoma cells into bone-like cells to impede tumor growth. CONCLUSIONS: In summary, attIL12-PBMC therapy is safe and effective against osteosarcoma. Our goal is to move this treatment into a clinical trial. Owing to the convenience of the attIL12-PBMC production process, we believe it will be feasible.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Neoplasias Óseas/terapia , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva/métodos , Interleucina-12 , Leucocitos Mononucleares , Osteosarcoma/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Immunother Cancer ; 10(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35027427

RESUMEN

BACKGROUND: Adoptive T-cell transfer has become an attractive therapeutic approach for hematological malignancies but shows poor activity against large and heterogeneous solid tumors. Interleukin-12 (IL-12) exhibits potent antitumor efficacy against solid tumors, but its clinical application has been stalled because of toxicity. Here, we aimed to develop a safe approach to IL-12 T-cell therapy for eliminating large solid tumors. METHODS: We generated a cell membrane-anchored IL-12 (aIL12), a tumor-targeted IL-12 (ttIL12), and a cell membrane-anchored and ttIL-12 (attIL12) and a cell membrane-anchored and tumor-targeted ttIL-12 (attIL12) armed T cells, chimeric antigen receptor-T cells, and T cell receptor-T (TCR-T) cells with each. We compared the safety and efficacy of these armed T cells in treating osteosarcoma patient-derived xenograft tumors and mouse melanoma tumors after intravenous infusions of the armed T cells. RESULTS: attIL12-T cell infusion showed remarkable antitumor efficacy in human and mouse large solid tumor models. Mechanistically, attIL12-T cells targeted tumor cells expressing cell-surface vimentin, enriching effector T cell and interferon γ production in tumors, which in turn stimulates dendritic cell maturation for activating secondary T-cell responses and tumor antigen spreading. Both attIL12- and aIL12-T-cell transfer eliminated peripheral cytokine release and the associated toxic effects. CONCLUSIONS: This novel approach sheds light on the safe application of IL-12-based T-cell therapy for large and heterogeneous solid tumors.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunoterapia/métodos , Interleucina-12/inmunología , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
10.
Nat Commun ; 12(1): 3500, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34108491

RESUMEN

WSX1, a receptor subunit for IL-27, is widely expressed in immune cells and closely involved in immune response, but its function in nonimmune cells remains unknown. Here we report that WSX1 is highly expressed in human hepatocytes but downregulated in hepatocellular carcinoma (HCC) cells. Using NRAS/AKT-derived spontaneous HCC mouse models, we reveal an IL-27-independent tumor-suppressive effect of WSX1 that largely relies on CD8+ T-cell immune surveillance via reducing neoplastic PD-L1 expression and the associated CD8+ T-cell exhaustion. Mechanistically, WSX1 transcriptionally downregulates an isoform of PI3K-PI3Kδ and thereby inactivates AKT, reducing AKT-induced GSK3ß inhibition. Activated GSK3ß then boosts PD-L1 degradation, resulting in PD-L1 reduction. Overall, we demonstrate that WSX1 is a tumor suppressor that reinforces hepatic immune surveillance by blocking the PI3Kδ/AKT/GSK3ß/PD-L1 pathway. Our results may yield insights into the host homeostatic control of immune response and benefit the development of cancer immunotherapies.


Asunto(s)
Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/inmunología , Receptores de Interleucina/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Vigilancia Inmunológica , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Persona de Mediana Edad , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Interleucina/metabolismo , Transducción de Señal/inmunología , Proteínas Supresoras de Tumor/metabolismo
11.
Cancer Lett ; 506: 83-94, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33676940

RESUMEN

Glioma stem cells (GSCs) are thought to underlie glioma initiation, evolution, resistance to therapies, and relapse. They are defined by their capacity to initiate glioma in immunocompromised mice which precludes analysis of their interaction with immune cells. Macrophages dominate the immune cell composition in glioma. We hypothesized that stemness and immune evasion induced by macrophages are closed intertwined in glioma. By using mass cytometry and RNA sequencing, we reveal that in immunocompetent mice, FGL2 promotes the stem-like phenotypes of glioma cells in an expression level-dependent manner. Mechanistically, FGL2-producing glioma cells recruit macrophages into the tumor microenvironment and induce the macrophages to secrete CXCL7 via the CD16/SyK/PI3K/HIF1α pathways. CXCL7, in turn, enhances the stem-like functionality of glioma cells, resulting in an increase in tumor incidence and progression that can be blocked with a neutralizing anti-CXCL7 antibody. Clinically, the FGL2-CXCL7 paracrine loop positively correlated with a higher macrophage signature and poorer prognosis in glioma patients. Thus, glioma cells' stem-like functionality is regulated by FGL2 in the presence of macrophages, and the FGL2-CXCL7 paracrine signaling axis is critical for regulating this function.


Asunto(s)
Neoplasias Encefálicas/etiología , Quimiocinas CXC/fisiología , Fibrinógeno/fisiología , Glioma/etiología , Células Madre Neoplásicas/fisiología , Macrófagos Asociados a Tumores/fisiología , Animales , Neoplasias Encefálicas/patología , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Glioma/patología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/fisiología , Transducción de Señal/fisiología
12.
Sci Adv ; 7(5)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33571109

RESUMEN

The invasiveness and high immune suppression of glioblastoma multiforme (GBM) produce poor survival of afflicted patients. Unfortunately, in the past decades, no therapeutic approach has remarkably improved the survival time of patients with GBM. Our analysis of the TCGA database and brain tumor tissue arrays indicated that CXCL1 and CXCL2 overexpression is closely associated with GBM's aggressiveness. Our results showed that elevation of CXCL1 or CXCL2 facilitated myeloid cell migration and simultaneously disrupted CD8+ T cell accumulation at tumor sites, causing accelerated tumor progression. Yet, blockade of CXCL1/2 significantly prevented myeloid-derived suppressor cell migration and thereby increased CD8+ T cell accumulation in vitro and in vivo. CXCL1/2 also promoted the paracrine factor S100A9 and further activated Erk1/2 and p70S60k, whereas blocking CXCL1/2 down-regulated these prosurvival factors. The combination of targeting CXCL1/2 and standard temozolomide chemotherapy improved upon the antitumor efficacy of chemotherapy alone, extending the overall survival time in GBM.


Asunto(s)
Neoplasias Encefálicas , Quimiocina CXCL1 , Quimiocina CXCL2 , Glioblastoma , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Escape del Tumor
13.
Cancer Lett ; 502: 143-153, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33279621

RESUMEN

Shedding, loss of expression, or internalization of natural killer group 2, member D (NKG2D) ligands from the tumor cell surface leads to immune evasion, which is associated with poor prognosis in patients with cancer. In many cancers, matrix metalloproteinases cause the proteolytic shedding of NKG2D ligands. However, it remained unclear how to protect NKG2D ligands from shedding. Here, we showed that the shedding of the mouse NKG2D ligand Rae-1 can be prevented by two critical acetyltransferases, GCN5 and PCAF, which acetylate the lysine residues of Rae-1 to avoid shedding both in vitro and in vivo. In contrast, mutations at lysines 80 and 87 of Rae-1 abrogated this acetylation and thereby desensitized tumor cells to NKG2D-dependent immune surveillance. Notably, the protein levels of GCN5 correlated with the expression levels of the human NKG2D ligand ULPB1 in a human tumor tissue microarray and, more importantly, with prolonged overall survival in many cancers. Our results suggest that the acetylation of Rae-1 protein at lysines 80 and 87 by GCN5 and PCAF protects Rae-1 from shedding so as to activate NKG2D-dependent immune surveillance. This discovery may shed light on new targets for NKG2D immunotherapy in cancer treatment.


Asunto(s)
Lisina/metabolismo , Mutación , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/metabolismo , Proteínas Asociadas a Matriz Nuclear/química , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/química , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Acetilación , Línea Celular Tumoral , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisina/genética , Trasplante de Neoplasias , Neoplasias/genética , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Estabilidad Proteica , Análisis de Supervivencia , Análisis de Matrices Tisulares , Escape del Tumor , Factores de Transcripción p300-CBP/metabolismo
14.
Oncogene ; 38(49): 7433-7446, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31427736

RESUMEN

The ligands for the natural killer group 2 (NKG2D) protein render tumor cells susceptible to NKG2D-dependent immune cell attack. However, cancer cells escape from immune surveillance by downregulating NKG2D ligands. We previously discovered that engagement of activated CD8+ T cells and tumor cells induces NKG2D ligands on tumor cells, but the underlying mechanism remains to be defined. Both in vivo mouse tumor models and in vitro cell assays were performed to study the downstream signaling. Our results supported the notion that, upon engagement with the cognate receptors, CD137 ligand and CD40 initiates activation of nuclear factor-kappa B (NF-κB) signaling in tumor cells even in the absence of CD8+ T cells. Like tumor and CD8+ T cell contact-dependent NKG2D ligand induction, this CD137L/CD40-mediated signaling activation was associated with elevated levels of acetyltransferase P300/CBP-associated factor (PCAF), whereas inhibition of phosphorylated NF-κB abrogated PCAF induction. Although stimulation of CD137L/CD40-mediated signaling is vital, inflammatory cytokines, including interferon gamma (IFNγ) and TNFα, also facilitate NKG2D ligand-induced immune surveillance via both facilitating T-cell chemotaxis and CD137L/CD40 induced NF-κB/PCAF activation. Collectively, our results unveil a novel mechanism of NKG2D ligand upregulation involving reverse signaling of CD40 and CD137L on tumor cells which, along with inflammatory cytokines IFNγ and TNFα, stimulate downstream NF-κB and PCAF activation. Understanding this mechanism may help in development of induced NKG2D ligand-dependent T-cell therapy against cancers.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , FN-kappa B/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Neoplasias/genética , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Factores de Transcripción p300-CBP/genética
15.
J Immunother Cancer ; 7(1): 154, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31208461

RESUMEN

BACKGROUND: Although accumulated evidence provides a strong scientific premise for using immune profiles to predict survival in patients with cancer, a universal immune profile across tumor types is still lacking, and how to achieve a survival-associated immune profile remains to be evaluated. METHODS: We analyzed datasets from The Cancer Genome Atlas to identify an immune profile associated with prolonged overall survival in multiple tumor types and tested the efficacy of tumor cell-surface vimentin-targeted interleukin 12 (ttIL-12) in inducing that immune profile and prolonging survival in both mouse and patient-derived xenograft tumor models. RESULTS: We identified an immune profile (IFNγHiCD8HiFOXP3LowCD33Low) associated with prolonged overall survival across several human tumor types. ttIL-12 in combination with surgical resection of the primary tumor transformed tumors to this immune profile. Intriguingly, this immune profile transformation led to inhibition of metastasis and to prolonged survival in both mouse and patient-derived xenograft malignant models. Wild-type IL-12 combined with surgery was significantly less effective. In the IL-12-sensitive C3H mouse strain, in fact, wild-type IL-12 and surgery resulted in shorter overall survival than in mice treated with control pDNA; this surprising result is believed to be attributable to IL-12 toxicity, which was absent in the mice treated with ttIL-12. The ttIL-12-induced immune profile associated with longer overall survival was also associated with a greater accumulation of CD8+ T cells and reduced infiltration of regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages. The underlying mechanism for this transformation by ttIL-12 treatment was induction of expression of CXCL9 and reduction of expression of CXCL2 and CCL22 in tumors. CONCLUSIONS: ttIL-12 when combined with surgery led to conversion to the IFNγHiCD8HiFOXP3LowCD33Low immune profile, eliminated relapse and metastasis, and prolonged overall survival.


Asunto(s)
Interleucina-12/farmacología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-12/inmunología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Terapia Molecular Dirigida , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Osteosarcoma/genética , Osteosarcoma/inmunología , Osteosarcoma/patología , Osteosarcoma/terapia , Análisis de Supervivencia , Vimentina/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nat Commun ; 10(1): 862, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30770835

RESUMEN

The original version of this Article contained errors in the author affiliations. Qingnan Zhao, Xueqing Xia, Longfei Huo and Shulin Li were incorrectly associated with Beijing Institute for Brain Disorders, 100069, Beijing, China.This has now been corrected in both the PDF and HTML versions of the Article.

17.
Nat Commun ; 10(1): 448, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683885

RESUMEN

Few studies implicate immunoregulatory gene expression in tumor cells in arbitrating brain tumor progression. Here we show that fibrinogen-like protein 2 (FGL2) is highly expressed in glioma stem cells and primary glioblastoma (GBM) cells. FGL2 knockout in tumor cells did not affect tumor-cell proliferation in vitro or tumor progression in immunodeficient mice but completely impaired GBM progression in immune-competent mice. This impairment was reversed in mice with a defect in dendritic cells (DCs) or CD103+ DC differentiation in the brain and in tumor-draining lymph nodes. The presence of FGL2 in tumor cells inhibited granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced CD103+ DC differentiation by suppressing NF-κB, STAT1/5, and p38 activation. These findings are relevant to GBM patients because a low level of FGL2 expression with concurrent high GM-CSF expression is associated with higher CD8B expression and longer survival. These data provide a rationale for therapeutic inhibition of FGL2 in brain tumors.


Asunto(s)
Antígenos CD/genética , Neoplasias Encefálicas/genética , Células Dendríticas/inmunología , Fibrinógeno/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Cadenas alfa de Integrinas/genética , Animales , Antígenos CD/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular , Línea Celular Tumoral , Células Dendríticas/patología , Progresión de la Enfermedad , Fibrinógeno/inmunología , Glioblastoma/inmunología , Glioblastoma/mortalidad , Glioblastoma/patología , Xenoinjertos , Humanos , Cadenas alfa de Integrinas/inmunología , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Neuroglía/inmunología , Neuroglía/patología , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología , Análisis de Supervivencia , Carga Tumoral , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
18.
Cancer Lett ; 433: 176-185, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29991446

RESUMEN

Glioblastoma multiforme (GBM) is the most prevalent and aggressive brain tumor. The current standard therapy, which includes radiation and chemotherapy, is frequently ineffective partially because of drug resistance and poor penetration of the blood-brain barrier. Reducing resistance and increasing sensitivity to chemotherapy may improve outcomes. Glioma stem cells (GSCs) are a source of relapse and chemoresistance in GBM; sensitization of GSCs to temozoliomide (TMZ), the primary chemotherapeutic agent used to treat GBM, is therefore integral for therapeutic efficacy. We previously discovered a unique tumor-specific target, cell surface vimentin (CSV), on patient-derived GSCs. In this study, we found that the anti-CSV monoclonal antibody 86C efficiently increased GSC sensitivity to TMZ. The combination TMZ+86C induced significantly greater antitumor effects than TMZ alone in eight of 12 GSC lines. TMZ+86C-sensitive GSCs had higher CSV expression overall and faster CSV resurfacing among CSV- GSCs compared with TMZ+86C-resistant GSCs. Finally, TMZ+86C increased apoptosis of tumor cells and prolonged survival compared with either drug alone in GBM mouse models. The combination of TMZ+86C represents a promising strategy to reverse GSC chemoresistance.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Temozolomida/administración & dosificación , Vimentina/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Glioblastoma/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Oncoimmunology ; 7(5): e1420450, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721368

RESUMEN

The clinical utility of circulating tumor cells (CTCs) has been investigated in numerous publications, but CTCs that express very typical immune cell markers have not been reported. Here we report a novel class of CTCs-CSV-positive macrophage-like CTCs (ML-CTCs). This nomenclature was based on the fact that this class of CTCs can be captured from blood samples of gastrointestinal stromal tumors (GISTs) patients using either the macrophage marker CD68 or our proprietary tumor-specific cell-surface vimentin (CSV) antibody 84-1; likewise, the captured ML-CTCs can be co-stained with both typical macrophage markers (CD14, CD68) and tumor cell markers (DOG-1, C-kit) but not CD45. Patients with metastatic GIST had significantly greater numbers of ML-CTCs than patients with localized GIST or cancer-free blood donors (P<0.0001). Unexpectedly, the classic CSV positive CTCs was abundant in metastatic disease but failed to predict GIST metastasis. Only CSV-positive ML-CTCs was able to serve as a solid and novel biomarker for prediction of metastatic risk in GIST patients.

20.
PLoS One ; 13(3): e0193485, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29494633

RESUMEN

Spondyloarthropathies, the second most frequently occurring form of chronic inflammatory arthritis, affects young adults in particular. However, a proper model with which to study the biology of this disease and to develop therapeutics is lacking. One of the most accepted animal models for this disease uses HLA-B27/Hu-ß2m transgenic rats; however, only 30%-50% of male HLA-B27/Hu-ß2m rats develop spontaneous, clinically apparent spondylitis and have a variable time until disease onset. Here, we report a high-incidence, low-variation spontaneous mouse model that delineates how the combination of inflammatory cytokine interleukin-27 (IL-27) signaling deficiency and mitogenic signaling (mutant p53R172H) in vivo, leads to bone loss in the vertebral bodies and ossification of the cartilage in the intervertebral discs. In this human disease-like mouse model, bone loss and pathogenic bone development are seen as early as 4 months of age in the absence of inflammatory aggregates in the enthesis or intervertebral disc.


Asunto(s)
Modelos Animales de Enfermedad , Receptores de Citocinas/genética , Espondiloartropatías/patología , Proteína p53 Supresora de Tumor/genética , Animales , Humanos , Masculino , Ratones , Mutación , Ratas , Receptores de Interleucina , Transducción de Señal , Espondiloartropatías/diagnóstico por imagen , Espondiloartropatías/genética , Espondiloartropatías/veterinaria , Microtomografía por Rayos X/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA