Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(1): 199-204, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30559191

RESUMEN

Cysteinyl leukotrienes (cys-LTs) are proinflammatory mediators that enhance vascular permeability through distinct receptors (CysLTRs). We found that CysLT2R regulates angiogenesis in isolated mouse endothelial cells (ECs) and in Matrigel implants in WT mice and enhances EC contraction and permeability via the Rho-dependent myosin light chain 2 and vascular endothelial (VE)-cadherin axis. Since solid tumors utilize aberrant angiogenesis for their growth and metastasis and their vessels exhibit vascular hyperpermeability, we hypothesized that CysLT2R, via its actions on the endothelium, might regulate tumor growth. Both tumor growth and metastases of adoptively transferred syngeneic Lewis lung carcinoma (LLC) cells are significantly reduced in CysLT2R-null mice (Cysltr2-/-) compared with WT and CysLT1R-null mice (Cysltr1-/-). In WT recipients of LLC cells, CysLT2R expression is significantly increased in the tumor vasculature, compared with CysLT1R. Further, the tumor vasculature in Cysltr2-/- recipients exhibited significantly improved integrity, as revealed by increased pericyte coverage and decreased leakage of i.v.-administered Texas Red-conjugated dextran. Administration of a selective CysLT2R antagonist significantly reduced LLC tumor volume, vessel density, dextran leakage, and metastases in WT mice, highlighting CysLT2R as a VEGF-independent regulator of the vasculature promoting risk of metastasis. Thus, both genetic and pharmacological findings establish CysLT2R as a gateway for angiogenesis and EC dysregulation in vitro and ex vivo and in an in vivo model with a mouse tumor. Our data suggest CysLT2R as a possible target for intervention.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Neovascularización Patológica/inducido químicamente , Receptores de Leucotrienos/metabolismo , Animales , Permeabilidad Capilar/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/farmacología , Técnicas de Inactivación de Genes , Antagonistas de Leucotrieno/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/tratamiento farmacológico , Trasplante de Neoplasias , Neoplasias Experimentales , Neovascularización Patológica/tratamiento farmacológico , Ácidos Ftálicos/farmacología , Receptores de Leucotrienos/efectos de los fármacos
2.
J Immunol ; 197(3): 691-8, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27431984

RESUMEN

Microenvironments that tumor cells encounter are different during the stages of cancer progression-primary tumor, metastasis, and at the metastatic site. This suggests potential differences in immune surveillance of primary tumor and metastasis. Epithelial-mesenchymal transition (EMT) is a key reversible process in which cancer cells transition into highly motile and invasive cells for dissemination. Only a tiny proportion successfully metastasize, supporting the notion of metastasis-specific immune surveillance. EMT involves extensive molecular reprogramming of cells conferring many clinically relevant features to cancer cells and affects tumor cell interactions within the tumor microenvironment. We review the impact of tumor immune infiltrates on tumor cell EMT and the consequences of EMT in shaping the immune microenvironment of tumors. The usefulness of EMT as a model to investigate metastasis-specific immune surveillance mechanisms are also explored. Finally, we discuss potential implications of EMT for tumor immunogenicity, as well as current immunotherapies and future strategies.


Asunto(s)
Transición Epitelial-Mesenquimal/inmunología , Neoplasias/patología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Animales , Progresión de la Enfermedad , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología
3.
Biochem Biophys Res Commun ; 489(2): 130-134, 2017 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-28536076

RESUMEN

Arginine, a cationic amino acid is known to stabilize proteins under harsh conditions. It is widely used to stabilize protein aggregation, and to correct protein folding during protein production. Hence it would be a good therapeutic candidate for treating protein aggregation related diseases. Recent reports suggest, that the aggregation of tumor suppressor protein p53 is one of the leading causes of tumor progression. When mutated, p53 protein aggregates, loses its function leading to unwanted cell growth and ultimately results in tumor. Here in this study we focus on the inhibitory effects of polyarginine and its analogues polyornithine, canavanine, and citrulline on the inhibition of p53 mutant peptide aggregation, and p53 mutant cancer cell proliferation inhibition in vitro. Biochemical assays and cell toxicity studies were used to characterize the study. The results show that polyarginine, and polyornithine, in micromolar concentrations, significantly inhibits p53 conserved peptide aggregation, and the cell proliferation of p53 mutant cancer cells. Hence they could be promising candidates for treating p53 mutant/misfolded protein aggregation associated cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/genética , Neoplasias/patología , Péptidos/farmacología , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/prevención & control , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Ensayos Analíticos de Alto Rendimiento , Humanos , Mutación , Péptidos/administración & dosificación , Péptidos/química , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/patología , Relación Estructura-Actividad
4.
Am J Pathol ; 185(4): 969-86, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25681733

RESUMEN

Myofibroblasts are crucial to the pathogenesis of tissue fibrosis. Their formation of stress fibers results in the release of myocardin-related transcription factor (MRTF), a transcriptional coactivator of serum response factor (SRF). MRTF-A (Mkl1)-deficient mice are protected from lung fibrosis. We hypothesized that the SRF/MRTF pathway inhibitor CCG-203971 would modulate myofibroblast function in vitro and limit lung fibrosis in vivo. Normal and idiopathic pulmonary fibrosis lung fibroblasts were treated with/without CCG-203971 (N-[4-chlorophenyl]-1-[3-(2-furanyl)benzoyl]-3-piperidine carboxamide) and/or Fas-activating antibody in the presence/absence of transforming growth factor (TGF)-ß1, and apoptosis was assessed. In vivo studies examined the effect of therapeutically administered CCG-203971 on lung fibrosis in two distinct murine models of fibrosis induced by bleomycin or targeted type II alveolar epithelial injury. In vitro, CCG-203971 prevented nuclear localization of MRTF-A; increased the apoptotic susceptibility of normal and idiopathic pulmonary fibrosis fibroblasts; blocked TGF-ß1-induced myofibroblast differentiation; and inhibited TGF-ß1-induced expression of fibronectin, X-linked inhibitor of apoptosis, and plasminogen activator inhibitor-1. TGF-ß1 did not protect fibroblasts or myofibroblasts from apoptosis in the presence of CCG-203971. In vivo, CCG-203971 significantly reduced lung collagen content in both murine models while decreasing alveolar plasminogen activator inhibitor-1 and promoting myofibroblast apoptosis. These data support a central role of the SRF/MRTF pathway in the pathobiology of lung fibrosis and suggest that its inhibition can help resolve lung fibrosis by promoting fibroblast apoptosis.


Asunto(s)
Apoptosis , Pulmón/metabolismo , Pulmón/patología , Mesodermo/patología , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Adulto , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoprotección/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis , Humanos , Inflamación/patología , Mesodermo/efectos de los fármacos , Ratones Endogámicos C57BL , Miofibroblastos/patología , Ácidos Nipecóticos/administración & dosificación , Ácidos Nipecóticos/farmacología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sus scrofa , Factor de Crecimiento Transformador beta1/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Receptor fas/metabolismo
5.
BMC Cancer ; 16: 614, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27501846

RESUMEN

BACKGROUND: Circulating tumor cells (CTCs) have shown prognostic relevance in many cancer types. However, the majority of current CTC capture methods rely on positive selection techniques that require a priori knowledge about the surface protein expression of disseminated CTCs, which are known to be a dynamic population. METHODS: We developed a microfluidic CTC capture chip that incorporated a nanoroughened glass substrate for capturing CTCs from blood samples. Our CTC capture chip utilized the differential adhesion preference of cancer cells to nanoroughened etched glass surfaces as compared to normal blood cells and thus did not depend on the physical size or surface protein expression of CTCs. RESULTS: The microfluidic CTC capture chip was able to achieve a superior capture yield for both epithelial cell adhesion molecule positive (EpCAM+) and EpCAM- cancer cells in blood samples. Additionally, the microfluidic CTC chip captured CTCs undergoing transforming growth factor beta-induced epithelial-to-mesenchymal transition (TGF-ß-induced EMT) with dynamically down-regulated EpCAM expression. In a mouse model of human breast cancer using EpCAM positive and negative cell lines, the number of CTCs captured correlated positively with the size of the primary tumor and was independent of their EpCAM expression. Furthermore, in a syngeneic mouse model of lung cancer using cell lines with differential metastasis capability, CTCs were captured from all mice with detectable primary tumors independent of the cell lines' metastatic ability. CONCLUSIONS: The microfluidic CTC capture chip using a novel nanoroughened glass substrate is broadly applicable to capturing heterogeneous CTC populations of clinical interest independent of their surface marker expression and metastatic propensity. We were able to capture CTCs from a non-metastatic lung cancer model, demonstrating the potential of the chip to collect the entirety of CTC populations including subgroups of distinct biological and phenotypical properties. Further exploration of the biological potential of metastatic and presumably non-metastatic CTCs captured using the microfluidic chip will yield insights into their relevant differences and their effects on tumor progression and cancer outcomes.


Asunto(s)
Separación Celular/métodos , Molécula de Adhesión Celular Epitelial/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Neoplasias/metabolismo , Células Neoplásicas Circulantes/patología , Factor de Crecimiento Transformador beta/farmacología , Células A549 , Animales , Adhesión Celular , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Heterogeneidad Genética , Humanos , Células MCF-7 , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias/patología , Células Neoplásicas Circulantes/efectos de los fármacos , Células Neoplásicas Circulantes/metabolismo
6.
Carcinogenesis ; 35(6): 1292-300, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24510113

RESUMEN

In cancer cells, the process of epithelial-mesenchymal transition (EMT) confers migratory and invasive capacity, resistance to apoptosis, drug resistance, evasion of host immune surveillance and tumor stem cell traits. Cells undergoing EMT may represent tumor cells with metastatic potential. Characterizing the EMT secretome may identify biomarkers to monitor EMT in tumor progression and provide a prognostic signature to predict patient survival. Utilizing a transforming growth factor-ß-induced cell culture model of EMT, we quantitatively profiled differentially secreted proteins, by GeLC-tandem mass spectrometry. Integrating with the corresponding transcriptome, we derived an EMT-associated secretory phenotype (EASP) comprising of proteins that were differentially upregulated both at protein and mRNA levels. Four independent primary tumor-derived gene expression data sets of lung cancers were used for survival analysis by the random survival forests (RSF) method. Analysis of 97-gene EASP expression in human lung adenocarcinoma tumors revealed strong positive correlations with lymph node metastasis, advanced tumor stage and histological grade. RSF analysis built on a training set (n = 442), including age, sex and stage as variables, stratified three independent lung cancer data sets into low-, medium- and high-risk groups with significant differences in overall survival. We further refined EASP to a 20 gene signature (rEASP) based on variable importance scores from RSF analysis. Similar to EASP, rEASP predicted survival of both adenocarcinoma and squamous carcinoma patients. More importantly, it predicted survival in the early-stage cancers. These results demonstrate that integrative analysis of the critical biological process of EMT provides mechanism-based and clinically relevant biomarkers with significant prognostic value.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Fenotipo , Adulto , Anciano , Línea Celular Tumoral , Análisis por Conglomerados , Biología Computacional , Transición Epitelial-Mesenquimal/genética , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Proteómica
8.
Ann Am Thorac Soc ; 20(8): 1077-1087, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37526479

RESUMEN

Rationale: To identify barriers and opportunities for Ph.D., basic and translational scientists to be fully integrated into clinical units. Objectives: In 2022, an ad hoc committee of the American Thoracic Society developed a project proposal and workshop to identify opportunities and barriers for scientists who do not practice medicine to develop successful careers and achieve tenure-track faculty positions in clinical departments and divisions within academic medical centers (AMCs) in the United States. Methods: This document focuses on results from a survey of adult and pediatric pulmonary, critical care, and sleep medicine division chiefs as well as a survey of workshop participants, including faculty in departmental and school leadership roles in both basic science and clinical units within U.S. AMCs. Results: We conclude that full integration of non-clinically practicing basic and translational scientists into the clinical units, in addition to their traditional placements in basic science units, best serves the tripartite mission of AMCs to provide care, perform research, and educate the next generation. Evidence suggests clinical units do employ Ph.D. scientists in large numbers, but these faculty are often hired into non-tenure track positions, which do not provide the salary support, start-up funds, research independence, or space often associated with hiring in basic science units within the same institution. These barriers to success of Ph.D. faculty in clinical units are largely financial. Conclusions: Our recommendation is for AMCs to consider and explore some of our proposed strategies to accomplish the goal of integrating basic and translational scientists into clinical units in a meaningful way.


Asunto(s)
Centros Médicos Académicos , Médicos , Adulto , Estados Unidos , Humanos , Niño , Selección de Personal , Liderazgo , Docentes Médicos
9.
Am J Pathol ; 178(6): 2461-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21641374

RESUMEN

Fibrotic obliteration of the small airways leading to progressive airflow obstruction, termed bronchiolitis obliterans syndrome (BOS), is the major cause of poor outcomes after lung transplantation. We recently demonstrated that a donor-derived population of multipotent mesenchymal stem cells (MSCs) can be isolated from the bronchoalveolar lavage (BAL) fluid of human lung transplant recipients. Herein, we study the organ specificity of these cells and investigate the role of local mesenchymal progenitors in fibrogenesis after lung transplantation. We demonstrate that human lung allograft-derived MSCs uniquely express embryonic lung mesenchyme-associated transcription factors with a 35,000-fold higher expression of forkhead/winged helix transcription factor forkhead box (FOXF1) noted in lung compared with bone marrow MSCs. Fibrotic differentiation of MSCs isolated from normal lung allografts was noted in the presence of profibrotic mediators associated with BOS, including transforming growth factor-ß and IL-13. MSCs isolated from patients with BOS demonstrated increased expression of α-SMA and collagen I when compared with non-BOS controls, consistent with a stable in vivo fibrotic phenotype. FOXF1 mRNA expression in the BAL cell pellet correlated with the number of MSCs in the BAL fluid, and myofibroblasts present in the fibrotic lesions expressed FOXF1 by in situ hybridization. These data suggest a key role for local tissue-specific, organ-resident, mesenchymal precursors in the fibrogenic processes in human adult lungs.


Asunto(s)
Trasplante de Pulmón , Pulmón/patología , Células Madre Mesenquimatosas/patología , Actinas/metabolismo , Biomarcadores/metabolismo , Biopsia , Células de la Médula Ósea/patología , Bronquiolitis Obliterante/patología , Líquido del Lavado Bronquioalveolar , Recuento de Células , Diferenciación Celular , Separación Celular , Colágeno/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Humanos , Pulmón/embriología , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Miofibroblastos/patología , Especificidad de Órganos , Fenotipo , Receptores de Interleucina-13/metabolismo , Trasplante Homólogo
10.
Blood ; 115(22): 4403-11, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20130237

RESUMEN

One of the more insidious outcomes of patients who survive severe sepsis is profound immunosuppression. In this study, we addressed the hypothesis that post septic immune defects were due, in part, to the presence and/or expansion of regulatory T cells (Tregs). After recovery from severe sepsis, mice exhibited significantly higher numbers of Tregs, which exerted greater in vitro suppressive activity compared with controls. The expansion of Tregs was not limited to CD25(+) cells, because Foxp3 expression was also detected in CD25(-) cells from post septic mice. This latter group exhibited a significant increase of chromatin remodeling at the Foxp3 promoter, because a marked increase in acetylation at H3K9 was associated with an increase in Foxp3 transcription. Post septic splenic dendritic cells promoted Treg conversion in vitro. Using a solid tumor model to explore the function of Tregs in an in vivo setting, we found post septic mice showed an increase in tumor growth compared with sham-treated mice with a syngeneic tumor model. This observation could mechanistically be related to the ability of post septic Tregs to impair the antitumor response mediated by CD8(+) T cells. Together, these data show that the post septic immune system obstructs tumor immunosurveillance, in part, by augmented Treg expansion and function.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Tolerancia Inmunológica , Sepsis/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Secuencia de Bases , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Sepsis/patología , Linfocitos T Reguladores/patología
11.
Bioinformatics ; 26(4): 456-63, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20007254

RESUMEN

MOTIVATION: The elucidation of biological concepts enriched with differentially expressed genes has become an integral part of the analysis and interpretation of genomic data. Of additional importance is the ability to explore networks of relationships among previously defined biological concepts from diverse information sources, and to explore results visually from multiple perspectives. Accomplishing these tasks requires a unified framework for agglomeration of data from various genomic resources, novel visualizations, and user functionality. RESULTS: We have developed ConceptGen, a web-based gene set enrichment and gene set relation mapping tool that is streamlined and simple to use. ConceptGen offers over 20,000 concepts comprising 14 different types of biological knowledge, including data not currently available in any other gene set enrichment or gene set relation mapping tool. We demonstrate the functionalities of ConceptGen using gene expression data modeling TGF-beta-induced epithelial-mesenchymal transition and metabolomics data comparing metastatic versus localized prostate cancers.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Reconocimiento de Normas Patrones Automatizadas/métodos , Programas Informáticos , Animales , Biología Computacional , Bases de Datos Genéticas , Redes Reguladoras de Genes , Humanos , Masculino , Metástasis de la Neoplasia/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
12.
Adv Sci (Weinh) ; 8(6): 2003747, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33747745

RESUMEN

As the recognition between natural killer (NK) cells and cancer cells does not require antigen presentation, NK cells are being actively studied for use in adoptive cell therapies in the rapidly evolving armamentarium of cancer immunotherapy. In addition to utilizing NK cells, recent studies have shown that exosomes derived from NK cells also exhibit antitumor properties. Furthermore, these NK cell-derived exosomes exhibit higher stability, greater modification potentials and less immunogenicity compared to NK cells. Therefore, technologies that allow highly sensitive and specific isolation of NK cells and NK cell-derived exosomes can enable personalized NK-mediated cancer therapeutics in the future. Here, a novel microfluidic system to collect patient-specific NK cells and on-chip biogenesis of NK-exosomes is proposed. In a small cohort of non-small cell lung cancer (NSCLC) patients, both NK cells and circulating tumor cells (CTCs) were isolated, and it is found NSCLC patients have high numbers of NK and NK-exosomes compared with healthy donors, and these concentrations show a trend of positive and negative correlations with bloodborne CTC numbers, respectively. It is further demonstrated that the NK-exosomes harvested from NK-graphene oxide chip exhibit cytotoxic effect on CTCs. This versatile system is expected to be used for patient-specific NK-based immunotherapies along with CTCs for potential prognostic/diagnostic applications.

13.
Cell Chem Biol ; 28(12): 1716-1727.e6, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34289376

RESUMEN

GAS41 is an emerging oncogene overexpressed and implicated in multiple cancers, including non-small cell lung cancer (NSCLC). GAS41 is a dimeric protein that contains the YEATS domain, which is involved in the recognition of lysine-acylated histones. Here, we report the development of GAS41 YEATS inhibitors by employing a fragment-based screening approach. These inhibitors bind to GAS41 YEATS domain in a channel constituting a recognition site for acylated lysine on histone proteins. To enhance inhibitory activity, we developed a dimeric analog with nanomolar activity that blocks interactions of GAS41 with acetylated histone H3. Our lead compound engages GAS41 in cells, blocks proliferation of NSCLC cells, and modulates expression of GAS41-dependent genes, validating on-target mechanism of action. This study demonstrates that disruption of GAS41 protein-protein interactions may represent an attractive approach to target lung cancer cells. This work exemplifies the use of bivalent inhibitors as a general strategy to block challenging protein-protein interactions.


Asunto(s)
Amidas/farmacología , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Tiofenos/farmacología , Factores de Transcripción/antagonistas & inhibidores , Amidas/química , Antineoplásicos/química , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Estructura Molecular , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Tiofenos/química , Factores de Transcripción/metabolismo
14.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33497367

RESUMEN

Limitations of checkpoint inhibitor cancer immunotherapy include induction of autoimmune syndromes and resistance of many cancers. Since CD318, a novel CD6 ligand, is associated with the aggressiveness and metastatic potential of human cancers, we tested the effect of an anti-CD6 monoclonal antibody, UMCD6, on killing of cancer cells by human lymphocytes. UMCD6 augmented killing of breast, lung, and prostate cancer cells through direct effects on both CD8+ T cells and NK cells, increasing cancer cell death and lowering cancer cell survival in vitro more robustly than monoclonal antibody checkpoint inhibitors that interrupt the programmed cell death 1 (PD-1)/PD-1 ligand 1 (PD-L1) axis. UMCD6 also augmented in vivo killing by human peripheral blood lymphocytes of a human breast cancer line xenotransplanted into immunodeficient mice. Mechanistically, UMCD6 upregulated the expression of the activating receptor NKG2D and downregulated expression of the inhibitory receptor NKG2A on both NK cells and CD8+ T cells, with concurrent increases in perforin and granzyme B production. The combined capability of an anti-CD6 monoclonal antibody to control autoimmunity through effects on CD4+ lymphocyte differentiation while enhancing killing of cancer cells through distinct effects on CD8+ and NK cells opens a potential new approach to cancer immunotherapy that would suppress rather than instigate autoimmunity.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Humanos , Células Asesinas Naturales/citología , Ratones , Ratones SCID
15.
Blood ; 112(10): 4250-8, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18535203

RESUMEN

Neutrophils (polymorphonuclear leukocytes [PMNs]) are critical to the immune response, including clearance of infectious pathogens. Sepsis is associated with impaired PMN function, including chemotaxis. PMNs express peroxisome proliferator-activated receptor-gamma (PPAR-gamma), a ligand-activated nuclear transcription factor involved in immune and inflammatory regulation. The role of PPAR-gamma in PMN responses, however, is not well characterized. We report that freshly isolated human PMNs constitutively express PPAR-gamma, which is up-regulated by the sepsis-induced cytokines TNF-alpha and IL-4. PMN chemotactic responses to formylmethionyl-leucyl-phenylalanine (fMLP) and IL-8 were dose-dependently inhibited by treatment with the PPAR-gamma ligands troglitazone and 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) and by transfection of PMN-like HL-60 cells with a constitutively active PPAR-gamma construct. Inhibition of chemotaxis by PPAR-gamma ligands correlated with decreases in extracellular signal-regulated kinase-1 and -2 activation, actin polymerization, and adherence to a fibrinogen substrate. Furthermore, PMN expression of PPAR-gamma was increased in sepsis patients and mice with either of 2 models of sepsis. Finally, treatment with the PPAR-gamma antagonist GW9662 significantly reversed the inhibition of PMN chemotaxis and increased peritoneal PMN recruitment in murine sepsis. This study indicates that PPAR-gamma activation is involved in PMN chemotactic responses in vitro and may play a role in the migration of these cells in vivo.


Asunto(s)
Quimiotaxis/inmunología , PPAR gamma/inmunología , Sepsis/inmunología , Regulación hacia Arriba/inmunología , Actinas/inmunología , Anilidas/farmacología , Animales , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Quimiotaxis/efectos de los fármacos , Cromanos/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Fibrinógeno/inmunología , Células HL-60 , Humanos , Inflamación/inmunología , Interleucina-4/inmunología , Interleucina-8/inmunología , Interleucina-8/farmacología , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/inmunología , N-Formilmetionina Leucil-Fenilalanina/inmunología , N-Formilmetionina Leucil-Fenilalanina/farmacología , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacología , Tiazolidinedionas/farmacología , Troglitazona , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba/efectos de los fármacos
16.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31619584

RESUMEN

Lung cancer remains the leading cause of cancer-related death in the United States. Although the alveolar macrophage (AM) comprises the major resident immune cell in the lung, few studies have investigated its role in lung cancer development. We recently discovered a potentially novel mechanism wherein AMs regulate STAT-induced inflammatory responses in neighboring epithelial cells (ECs) via secretion and delivery of suppressors of cytokine signaling 3 (SOCS3) within extracellular vesicles (EVs). Here, we explored the impact of SOCS3 transfer on EC tumorigenesis and the integrity of AM SOCS3 secretion during development of lung cancer. AM-derived EVs containing SOCS3 inhibited STAT3 activation as well as proliferation and survival of lung adenocarcinoma cells. Levels of secreted SOCS3 were diminished in lungs of patients with non-small cell lung cancer and in a mouse model of lung cancer, and the impaired ability of murine AMs to secrete SOCS3 within EVs preceded the development of lung tumors. Loss of this homeostatic brake on tumorigenesis prompted our effort to "rescue" it. Provision of recombinant SOCS3 loaded within synthetic liposomes inhibited proliferation and survival of lung adenocarcinoma cells in vitro as well as malignant transformation of normal ECs. Intratumoral injection of SOCS3 liposomes attenuated tumor growth in a lung cancer xenograft model. This work identifies AM-derived vesicular SOCS3 as an endogenous antitumor mechanism that is disrupted within the tumor microenvironment and whose rescue by synthetic liposomes can be leveraged as a potential therapeutic strategy for lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Neoplasias Pulmonares/inmunología , Macrófagos Alveolares/inmunología , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Células A549 , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/inmunología , Carcinogénesis/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Inyecciones Intralesiones , Liposomas , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Ratones , Cultivo Primario de Células , Ratas , Proteínas Recombinantes/administración & dosificación , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/administración & dosificación , Proteína 3 Supresora de la Señalización de Citocinas/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Clin Invest ; 115(10): 2801-10, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16151532

RESUMEN

Human noncollagenous domain 1 of the alpha1 chain of type IV collagen [alpha1(IV)NC1], or arresten, is derived from the carboxy terminal of type IV collagen. It was shown to inhibit angiogenesis and tumor growth in vivo; however, the mechanisms involved are not known. In the present study we demonstrate that human alpha1(IV)NC1 binds to alpha1beta1 integrin, competes with type IV collagen binding to alpha1beta1 integrin, and inhibits migration, proliferation, and tube formation by ECs. Also, alpha1(IV)NC1 pretreatment inhibited FAK/c-Raf/MEK/ERK1/2/p38 MAPK activation in ECs but had no effect on the PI3K/Akt pathway. In contrast, alpha1(IV)NC1 did not affect proliferation, migration, or the activation of FAK/c-Raf/MEK1/2/p38/ERK1 MAPK pathway in alpha1 integrin receptor knockout ECs. Consistent with this, alpha1(IV)NC1 elicited significant antiangiogenic effects and tumor growth inhibition in vivo but failed to do the same in alpha1 integrin receptor knockout mice. This suggests a highly specific, alpha1beta1 integrin-dependent antiangiogenic activity of alpha1(IV)NC1. In addition, alpha1(IV)NC1 inhibited hypoxia-induced expression of hypoxia-inducible factor 1alpha and VEGF in ECs cultured on type IV collagen by inhibiting ERK1/2 and p38 activation. This unravels a hitherto unknown function of human alpha1(IV)NC1 and suggests a critical role for integrins in hypoxia and hypoxia-induced angiogenesis. Collectively, the above data indicate that alpha1(IV)NC1 is a potential therapeutic candidate for targeting tumor angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Colágeno Tipo IV/metabolismo , Células Endoteliales/metabolismo , Sangre Fetal/metabolismo , Integrina alfa1beta1/metabolismo , Sistema de Señalización de MAP Quinasas , Neovascularización Patológica/metabolismo , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/farmacología , Hipoxia de la Célula/efectos de los fármacos , Colágeno Tipo IV/genética , Colágeno Tipo IV/farmacología , Células Endoteliales/citología , Activación Enzimática , Sangre Fetal/citología , Eliminación de Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Integrina alfa1beta1/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína/genética , Factor A de Crecimiento Endotelial Vascular/biosíntesis
18.
J Clin Invest ; 128(4): 1384-1396, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29324443

RESUMEN

During epithelial-mesenchymal transition (EMT) epithelial cancer cells transdifferentiate into highly motile, invasive, mesenchymal-like cells, giving rise to disseminating tumor cells. Few of these disseminated cells successfully metastasize. Immune cells and inflammation in the tumor microenvironment were shown to drive EMT, but few studies investigated the consequences of EMT for tumor immunosurveillance. In addition to initiating metastasis, we demonstrate that EMT confers increased susceptibility to natural killer (NK) cells and contributes, in part, to the inefficiency of the metastatic process. Depletion of NK cells allowed spontaneous metastasis without affecting primary tumor growth. EMT-induced modulation of E-cadherin and cell adhesion molecule 1 (CADM1) mediated increased susceptibility to NK cytotoxicity. Higher CADM1 expression correlates with improved patient survival in 2 lung and 1 breast adenocarcinoma patient cohorts and decreased metastasis. Our observations reveal a novel NK-mediated, metastasis-specific immunosurveillance in lung cancer and present a window of opportunity for preventing metastasis by boosting NK cell activity.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Transición Epitelial-Mesenquimal/inmunología , Inmunidad Celular , Vigilancia Inmunológica , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Microambiente Tumoral/inmunología , Células A549 , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Molécula 1 de Adhesión Celular/genética , Molécula 1 de Adhesión Celular/inmunología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Células Asesinas Naturales/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología
19.
Oncogene ; 21(25): 4000-8, 2002 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-12037682

RESUMEN

The estrogen receptor alpha (ERalpha) signaling plays an essential role in breast cancer progression and endocrine therapy. Mitogen-activated protein kinase (MAPK/Erk1/2) has been implicated in ligand-independent activation of ER, resulting in the cross-talk between growth factor and ER mediated signaling. In this study, we examined the effect of the cross-talk on estradiol (E(2))-mediated signaling, tumor growth and its effect on anti-estrogen therapy. Our findings demonstrate that expression of constitutively activated mitogen activated kinase kinase (MEK1), an immediate upstream activator of MAPK in estrogen receptor positive MCF-7 breast cancer cells (MEK/MCF-7), showed an increase in ERalpha-driven transcriptional activation. In MEK/MCF-7 cells maximal transactivation levels were achieved in response to treatment with much lower E(2) concentrations (10(-10) M E(2)) when compared to MCF-7 control cells (10(-8) M E(2)). Furthermore, we have seen an increased association between ERalpha and its nuclear coactivators AIB1 or TIF-2, in MEK/MCF-7 cells relative to those seen in MCF-7 control cells. In addition, in vivo studies show that MEK/MCF-7 cell tumors are approximately threefold larger than those of MCF-7 cell, in the presence of E(2). Immunohistochemical staining demonstrates that progesterone receptor (PR) and pS2, two E(2)-regulated gene products, are significantly increased in MEK/MCF-7 cell tumors compared to those of MCF-7 control tumors, suggesting that activation of ERalpha by MAPK enhances the expression of E(2)-regulated genes and accelerates tumor growth. Remarkably, the antiestrogens tamoxifen and ICI 182,780, were shown both in vitro and in vivo studies to efficiently antagonize the stimulatory effects of E(2) on ER regulated transactivation and tumor growth in MEK/MCF-7 as well as MCF-7 cell lines. Taken together, these data suggest that MAPK/ER cross-talk enhances ERalpha-mediated signaling and accelerates E(2)-dependent tumor growth without diminishing sensitivity to the inhibitory effects of anti-estrogens.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Estrógenos/metabolismo , Tamoxifeno/farmacología , Animales , Neoplasias de la Mama/metabolismo , División Celular , Cartilla de ADN/química , Receptor alfa de Estrógeno , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Humanos , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Ratones , Ratones SCID , Pruebas de Precipitina , Receptores de Estrógenos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Transfección , Células Tumorales Cultivadas
20.
Oncogene ; 23(1): 100-8, 2004 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-14712215

RESUMEN

The peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a member of the nuclear hormone receptor superfamily of ligand-activated transcription factors and a crucial regulator of cellular differentiation. Differentiation-inducing and antiproliferative effects of PPAR-gamma suggest that PPAR-gamma agonists might be useful as effective anticancer agents. Few studies have examined the efficacy of these agonists in animal models of tumorigenesis, and their mechanism(s) of action are still not clear. Our studies indicate higher PPAR-gamma expression in primary tumors from non-small-cell lung cancer (NSCLC) patients when compared to normal surrounding tissue. The expression of PPAR-gamma was also observed in several NSCLC lines. The treatment of lung adenocarcinoma cells (A549) with troglitazone (Tro), a PPAR-gamma ligand, enhanced PPAR-gamma transcriptional activity and induced a dose-dependent inhibition of A549 cell growth. The observed growth arrest was predominantly due to the inhibition of cell proliferation without significant induction of apoptosis. Cell cycle analysis of Tro-treated cells revealed a cell cycle arrest at G(0)/G(1) with concomitant downregulation of G(0)/G(1) cyclins D and E. In addition, Tro treatment stimulated sustained Erk1/2 activation in A549 cells, suggesting the activation of a differentiation-inducing pathway. Furthermore, treatment of A549 tumor-bearing SCID mice with Tro or Pio inhibited primary tumor growth by 66.7% and significantly inhibited the number of spontaneous lung metastatic lesions. Collectively, our data demonstrate that activation of PPAR-gamma impedes lung tumor progression and suggest that PPAR-gamma ligands may serve as potential therapeutic agents for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/prevención & control , Neoplasias Pulmonares/prevención & control , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Carcinoma de Pulmón de Células no Pequeñas/patología , División Celular , Línea Celular Tumoral , Activación Enzimática , Fase G1 , Humanos , Neoplasias Pulmonares/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores Citoplasmáticos y Nucleares/análisis , Fase de Descanso del Ciclo Celular , Factores de Transcripción/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA