Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(5): e0303171, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38768113

RESUMEN

Tumor microenvironment (TME) is a complex dynamic system with many tumor-interacting components including tumor-infiltrating leukocytes (TILs), cancer associated fibroblasts, blood vessels, and other stromal constituents. It intrinsically affects tumor development and pharmacology of oncology therapeutics, particularly immune-oncology (IO) treatments. Accurate measurement of TME is therefore of great importance for understanding the tumor immunity, identifying IO treatment mechanisms, developing predictive biomarkers, and ultimately, improving the treatment of cancer. Here, we introduce a mouse-IO NGS-based (NGSmIO) assay for accurately detecting and quantifying the mRNA expression of 1080 TME related genes in mouse tumor models. The NGSmIO panel was shown to be superior to the commonly used microarray approach by hosting 300 more relevant genes to better characterize various lineage of immune cells, exhibits improved mRNA and protein expression correlation to flow cytometry, shows stronger correlation with mRNA expression than RNAseq with 10x higher sequencing depth, and demonstrates higher sensitivity in measuring low-expressed genes. We describe two studies; firstly, detecting the pharmacodynamic change of interferon-γ expression levels upon anti-PD-1: anti-CD4 combination treatment in MC38 and Hepa 1-6 tumors; and secondly, benchmarking baseline TILs in 14 syngeneic tumors using transcript level expression of lineage specific genes, which demonstrate effective and robust applications of the NGSmIO panel.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Microambiente Tumoral , Animales , Ratones , Microambiente Tumoral/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Interferón gamma/genética , Interferón gamma/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , ARN Mensajero/genética , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Femenino , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Perfilación de la Expresión Génica/métodos
2.
Curr Protoc ; 3(8): e852, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37552031

RESUMEN

Breakthroughs in cancer treatment with immunotherapeutics have provided long-term patient benefits for many different types of cancer. However, complete response is not achieved in many patients and tumor types, and the mechanisms underlying this lack of response are poorly understood. Despite this, numerous new targets, therapeutics, and drug combinations are being developed and tested in clinical trials. Preclinical models that recapitulate the complex human tumor microenvironment and the interplay between tumor and immune cells within the cancer-immunity cycle are needed to improve our understanding and screen new therapeutics for efficacy and safety/toxicity. Humanized mice, encompassing human tumors and human immune cells engrafted on immunodeficient mice, have been widely used for many years in immuno-oncology, with developments to improve both the humanization and the translational value central to the next generation of models. In this overview, we discuss recent advances in humanized models relevant to immuno-oncology drug discovery, the advantages and limitations of such models, the application of humanized models for efficacy and safety assessments of immunotherapeutics, and the potential opportunities. © 2023 Crown Bioscience. Current Protocols published by Wiley Periodicals LLC.


Asunto(s)
Neoplasias , Humanos , Ratones , Animales , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Microambiente Tumoral
3.
PLoS One ; 18(1): e0279821, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36602988

RESUMEN

Patient-derived tumor xenograft (PDX)/organoid (PDO), driven by cancer stem cells (CSC), are considered the most predictive models for translational oncology. Large PDX collections reflective of patient populations have been created and used extensively to test various investigational therapies, including population-trials as surrogate subjects in vivo. PDOs are recognized as in vitro surrogates for patients amenable for high-throughput screening (HTS). We have built a biobank of carcinoma PDX-derived organoids (PDXOs) by converting an existing PDX library and confirmed high degree of similarities between PDXOs and parental PDXs in genomics, histopathology and pharmacology, suggesting "biological equivalence or interchangeability" between the two. Here we demonstrate the applications of PDXO biobank for HTS "matrix" screening for both lead compounds and indications, immune cell co-cultures for immune-therapies and engineering enables in vitro/in vivo imaging. This large biobank of >550 matched pairs of PDXs/PDXOs across different cancers could become powerful tools for the future cancer drug discovery.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Humanos , Bancos de Muestras Biológicas , Xenoinjertos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Antineoplásicos/farmacología , Modelos Animales de Enfermedad , Organoides , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Curr Protoc ; 2(7): e431, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35789132

RESUMEN

Patient-derived cancer disease models conserve many key features of the original human cancers, potentially allowing higher predictive power than traditional cell line models. Accordingly, in vivo patient-derived xenografts (PDX) are frequently utilized in preclinical and translational oncology studies as patient surrogates for population-based screens ("mouse clinical trials"), for which large PDX biobanks have been generated over the last decade from various cancer types. In vitro patient-derived organoids (PDO) have recently emerged as a disruptive technology, enabling early "patient in a dish" clinical trials. Like PDX, PDOs retain the histology/genomics of the original tumor and are highly predictive of the clinical response. Organoids derived from adult stem cells (ASC) in patient tissue can function as mini-organs. They have greater advantages over other 3D in vitro systems, making them highly predictive, reliable, and consistent in vitro models. Large biobanks enable the adoption of organoids in early drug screening and patient selection. PDX biobanks, as a source of human material, have been used to create 3D in vitro screens, but with limitations. However, creating organoids from the ASCs residing in PDXs has been successfully used as a rapid and cost-effective way to enable higher throughput in vitro screens and generate matched in vitro/in vivo model pairs that retain genomic, histopathological, and pharmacology profiles. This overview summarizes the generation of matched in vitro/in vivo models from patient material, the advantages over other systems, and the applications to drug discovery. © 2022 Wiley Periodicals LLC.


Asunto(s)
Neoplasias , Organoides , Animales , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Humanos , Ratones , Neoplasias/tratamiento farmacológico
5.
Sci Rep ; 12(1): 3278, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35228603

RESUMEN

Cancers are immunologically heterogeneous. A range of immunotherapies target abnormal tumor immunity via different mechanisms of actions (MOAs), particularly various tumor-infiltrate leukocytes (TILs). We modeled loss of function (LOF) in four common anti-PD-1 antibody-responsive syngeneic tumors, MC38, Hepa1-6, CT-26 and EMT-6, by systematical depleting a series of TIL lineages to explore the mechanisms of tumor immunity and treatment. CD8+-T-cells, CD4+-T-cells, Treg, NK cells and macrophages were individually depleted through either direct administration of anti-marker antibodies/reagents or using DTR (diphtheria toxin receptor) knock-in mice, for some syngeneic tumors, where specific subsets were depleted following diphtheria toxin (DT) administration. These LOF experiments revealed distinctive intrinsic tumor immunity and thus different MOAs in their responses to anti-PD-1 antibody among different syngeneic tumors. Specifically, the intrinsic tumor immunity and the associated anti-PD-1 MOA were predominately driven by CD8+ cytotoxic TILs (CTL) in all syngeneic tumors, excluding Hepa1-6 where CD4+ Teff TILs played a key role. TIL-Treg also played a critical role in supporting tumor growth in all four syngeneic models as well as M2-macrophages. Pathway analysis using pharmacodynamic readouts of immuno-genomics and proteomics on MC38 and Hepa1-6 also revealed defined, but distinctive, immune pathways of activation and suppression between the two, closely associated with the efficacy and consistent with TIL-pharmacodynamic readouts. Understanding tumor immune-pathogenesis and treatment MOAs in the different syngeneic animal models, not only assists the selection of the right model for evaluating new immunotherapy of a given MOA, but also can potentially help to understand the potential disease mechanisms and strategize optimal immune-therapies in patients.


Asunto(s)
Antineoplásicos , Inmunoterapia , Animales , Antineoplásicos/metabolismo , Linfocitos T CD8-positivos , Línea Celular Tumoral , Humanos , Linfocitos Infiltrantes de Tumor , Ratones , Linfocitos T Reguladores , Microambiente Tumoral
6.
Curr Opin Pharmacol ; 61: 49-61, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34619442

RESUMEN

Chimeric antigen receptor (CAR) T cells have largely been successful in treating hematological malignancies in the clinic but have not been as effective in treating solid tumors, in part, owing to poor access and the immunosuppressive tumor microenvironment. In addition, CAR-T therapy can cause potentially life-threatening side effects, including cytokine release syndrome and neurotoxicity. Current preclinical testing of CAR-T therapy efficacy is typically performed in mouse tumor models, which often fails to predict toxicity. Recent developments in humanized models and transgenic mice as well as in vitro three-dimensional organoids in early development and nonhuman primate models are being adopted for CAR-T cell efficacy and toxicity assessment. However, because no single model perfectly recapitulates the human immune system and tumor microenvironment, careful model selection based on their respective pros and cons is crucial for adequate evaluation of different CAR-T treatments, so that their clinical development can be better supported.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Animales , Inmunoterapia Adoptiva , Ratones , Neoplasias/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T , Microambiente Tumoral
7.
Mol Cancer Ther ; 15(4): 753-63, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26873730

RESUMEN

There is a growing recognition that current preclinical models do not reflect the tumor microenvironment in cellular, biological, and biophysical content and this may have a profound effect on drug efficacy testing, especially in the era of molecular-targeted agents. Here, we describe a method to directly embed low-passage patient tumor-derived tissue into basement membrane extract, ensuring a low proportion of cell death to anoikis and growth complementation by coculture with patient-derived cancer-associated fibroblasts (CAF). A range of solid tumors proved amenable to growth and pharmacologic testing in this 3D assay. A study of 30 early-stage non-small cell lung cancer (NSCLC) specimens revealed high levels of de novo resistance to a large range of standard-of-care agents, while histone deacetylase (HDAC) inhibitors and their combination with antineoplastic drugs displayed high levels of efficacy. Increased resistance was seen in the presence of patient-derived CAFs for many agents, highlighting the utility of the assay for tumor microenvironment-educated drug testing. Standard-of-care agents showed similar responses in the 3D ex vivo and patient-matched in vivo models validating the 3D-Tumor Growth Assay (3D-TGA) as a high-throughput screen for close-to-patient tumors using significantly reduced animal numbers. Mol Cancer Ther; 15(4); 753-63. ©2016 AACR.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Inhibidores de Histona Desacetilasas/uso terapéutico , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Nivel de Atención , Células del Estroma/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Biomarcadores , Transición Epitelial-Mesenquimal/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Técnicas In Vitro , Neoplasias Pulmonares/cirugía , Estadificación de Neoplasias , Fenotipo , Células del Estroma/metabolismo , Técnicas de Cultivo de Tejidos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Biomater Sci ; 3(11): 1439-48, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26351701

RESUMEN

Materials for delivery of oligonucleotides need to be simple to produce yet effective in vivo to be considered for clinical applications. Formulations of biomaterials based on combinations of existing demonstrated polymeric gene carriers with targeted derivatives are potential candidates for rapid translation but have not been fully explored for siRNA applications. Here we investigated formulations based on derivatised PEI for delivery of siRNA to gastrointestinal cancer cells. siRNA was complexed with linear PEI alone or with a mixture of linear PEI and transferrin-conjugated branched PEI (TfPEI), and knockdown of reporter genes was investigated. Overall, the in vitro use of complexes containing TfPEI resulted in up to 93% knockdown at 72 h post-transfection. Sustained knockdown was also achieved in a bioluminescent xenograft model. When complexes were delivered intratumorally, a 43% reduction in luminescence was achieved in the treated group compared with the control group 48 h after treatment. For systemic administration, only the intraperitoneal route, and not the intravenous route was effective, with 49% knockdown achieved at 72 h and sustained up to 144 h (44%) after a single administration of TfPEI-complexed siRNA. No toxicity or induction of the interferon response was observed. These findings demonstrate that simple formulations of transferrin-conjugated PEI with a 'parent' polymer such as linear PEI have potential as a method for therapeutic delivery of siRNA when administered either intratumorally or systemically.


Asunto(s)
Polietileneimina/administración & dosificación , ARN Interferente Pequeño/química , Transferrina/administración & dosificación , Línea Celular Tumoral , Portadores de Fármacos , Silenciador del Gen , Humanos , Polietileneimina/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transferrina/química
9.
Methods Mol Biol ; 1070: 117-29, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24092436

RESUMEN

Metastatic spread is generally responsible for the mortality of colorectal cancer patients. There are no adequate treatments for advanced colorectal cancer, and novel therapeutic modalities are urgently required. To this end, valid metastatic models, which accurately mimic the disease process, are needed. When deciding upon a metastasis model, the goals of the investigation will dictate the complexity of the model chosen. If biological mechanisms are being investigated, only a small number of experimental animals may be required, and a more complex, surgically intensive model may be used. If a therapeutic agent is being evaluated, owing to group sizes required to generate statistically significant effects, a less complex, less surgically intensive model may be preferable. The latter, however, may encompass only a particular phase of metastasis rather than reflecting all aspects of the metastatic cascade.


Asunto(s)
Ensayos de Migración Celular/métodos , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Metástasis de la Neoplasia/patología , Animales , Diagnóstico por Imagen , Femenino , Humanos , Neoplasias Hepáticas/patología , Mediciones Luminiscentes , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Invasividad Neoplásica
10.
J Biol Chem ; 287(47): 39967-81, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-22992725

RESUMEN

VEGF plays a central role in angiogenesis in cancer. Non-small cell lung cancer (NSCLC) tumors have increased microvascular density, localized hypoxia, and high VEGF expression levels; however, there is a lack of understanding of how oncogenic and tumor microenvironment changes such as hypoxia lead to greater VEGF expression in lung and other cancers. We show that NSCLC cells secreted higher levels of VEGF than normal airway epithelial cells. Actinomycin D inhibited all NSCLC VEGF secretion, and VEGF minimal promoter-luciferase reporter constructs were constitutively active until the last 85 base pairs before the transcription start site containing three SP-1 transcription factor-binding sites; mutation of these VEGF promoter SP-1-binding sites eliminated VEGF promoter activity. Furthermore, dominant negative SP-1, mithramycin A, and SP-1 shRNA decreased VEGF promoter activity, whereas overexpression of SP-1 increased VEGF promoter activity. Chromatin immunoprecipitation assays demonstrated SP-1, p300, and PCA/F histone acetyltransferase binding and histone H4 hyperacetylation at the VEGF promoter in NSCLC cells. Cultured NSCLC cells expressed higher levels of SP-1 protein than normal airway epithelial cells, and double-fluorescence immunohistochemistry showed a strong correlation between SP-1 and VEGF in human NSCLC tumors. In addition, hypoxia-driven VEGF expression in NSCLC cells was SP-1-dependent, with hypoxia increasing SP-1 activity and binding to the VEGF promoter. These studies are the first to demonstrate that overexpression of SP-1 plays a central role in hypoxia-induced VEGF secretion.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación Neoplásica de la Expresión Génica , Hipoxia/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/biosíntesis , Factor de Transcripción Sp1/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Hipoxia de la Célula , Línea Celular Tumoral , Dactinomicina/farmacología , Femenino , Histonas/genética , Histonas/metabolismo , Humanos , Hipoxia/genética , Hipoxia/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Regiones Promotoras Genéticas/genética , Unión Proteica , Factor de Transcripción Sp1/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
11.
J Ethnopharmacol ; 140(3): 545-9, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22353429

RESUMEN

UNLABELLED: ETHNO-PHARMACOLOGICAL RELEVANCE: The scientific proof and clinical validation of Chinese herbal medicine (CHM) require a rigorous approach that includes chemical standardization, biological assays, animal studies and clinical trials. AIM OF THE STUDY: To assess the experimental design of animal studies on the activity of CHM by selection and scrutinizing of a series of papers in some major disease areas. MATERIALS AND METHODS: We have analyzed the English publications reported in MEDLINE (ISI web of knowledge). RESULTS: Our data showed that (i) research of CHM during the last 10 years had been highly intensified and become more accessible worldwide through increased publications in English, although still most authors had Chinese names; (ii) English journals publishing animal research of CHM were comparable to those publishing animal studies of non-Chinese phytotherapy in terms of impact factor; and (iii) published data on authentication and quality control of CHM, as well as research design of animal studies were far from sufficient to meet the criteria needed to support their reproducibility and reliability. CONCLUSIONS AND PERSPECTIVES: The recent decade witnessed an increase in CHM research activities and CHM English publications. Based on common problems identified in publications on CHM animal studies, we have proposed a checklist that could help in preliminary selection of publications lacking the most common problems and thus would be useful for a quick search of reproducible CHM regimens that are likely to be effective in a given context. The second application of this checklist is to help avoid the most common problems when designing experiments.


Asunto(s)
Investigación Biomédica/métodos , Medicamentos Herbarios Chinos , Medicina de Hierbas , Medicina Tradicional China , Fitoterapia , Plantas Medicinales , Proyectos de Investigación/normas , Animales , Lenguaje , MEDLINE , Edición , Control de Calidad , Reproducibilidad de los Resultados
12.
Mol Cancer ; 10(1): 7, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21232089

RESUMEN

BACKGROUND: Breast cancer is a disease characterised by both genetic and epigenetic alterations. Epigenetic silencing of tumour suppressor genes is an early event in breast carcinogenesis and reversion of gene silencing by epigenetic reprogramming can provide clues to the mechanisms responsible for tumour initiation and progression. In this study we apply the reprogramming capacity of oocytes to cancer cells in order to study breast oncogenesis. RESULTS: We show that breast cancer cells can be directly reprogrammed by amphibian oocyte extracts. The reprogramming effect, after six hours of treatment, in the absence of DNA replication, includes DNA demethylation and removal of repressive histone marks at the promoters of tumour suppressor genes; also, expression of the silenced genes is re-activated in response to treatment. This activity is specific to oocytes as it is not elicited by extracts from ovulated eggs, and is present at very limited levels in extracts from mouse embryonic stem cells. Epigenetic reprogramming in oocyte extracts results in reduction of cancer cell growth under anchorage independent conditions and a reduction in tumour growth in mouse xenografts. CONCLUSIONS: This study presents a new method to investigate tumour reversion by epigenetic reprogramming. After testing extracts from different sources, we found that axolotl oocyte extracts possess superior reprogramming ability, which reverses epigenetic silencing of tumour suppressor genes and tumorigenicity of breast cancer cells in a mouse xenograft model. Therefore this system can be extremely valuable for dissecting the mechanisms involved in tumour suppressor gene silencing and identifying molecular activities capable of arresting tumour growth. These applications can ultimately shed light on the contribution of epigenetic alterations in breast cancer and advance the development of epigenetic therapies.


Asunto(s)
Ambystoma mexicanum , Neoplasias de la Mama/genética , Extractos Celulares/farmacología , Oocitos/química , Xenopus , Animales , Neoplasias de la Mama/patología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina , Digitonina/farmacología , Células Madre Embrionarias/química , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Histonas/metabolismo , Humanos , Metilación , Ratones , Proteínas de Microfilamentos/genética , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Activación Transcripcional , Trasplante Heterólogo , Células Tumorales Cultivadas
13.
Neuro Oncol ; 13(1): 70-83, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20978004

RESUMEN

Reliable model systems are needed to elucidate the role cancer stem cells (CSCs) play in pediatric brain tumor drug resistance. The majority of studies to date have focused on clinically distinct adult tumors and restricted tumor types. Here, the CSC component of 7 newly established primary pediatric cell lines (2 ependymomas, 2 medulloblastomas, 2 gliomas, and a CNS primitive neuroectodermal tumor) was thoroughly characterized. Comparison of DNA copy number with the original corresponding tumor demonstrated that genomic changes present in the original tumor, typical of that particular tumor type, were retained in culture. In each case, the CSC component was approximately 3-4-fold enriched in neurosphere culture compared with monolayer culture, and a higher capacity for multilineage differentiation was observed for neurosphere-derived cells. DNA content profiles of neurosphere-derived cells expressing the CSC marker nestin demonstrated the presence of cells in all phases of the cell cycle, indicating that not all CSCs are quiescent. Furthermore, neurosphere-derived cells demonstrated an increased resistance to etoposide compared with monolayer-derived cells, having lower initial DNA damage, potentially due to a combination of increased drug extrusion by ATP-binding cassette multidrug transporters and enhanced rates of DNA repair. Finally, orthotopic xenograft models reflecting the tumor of origin were established from these cell lines. In summary, these cell lines and the approach taken provide a robust model system that can be used to develop our understanding of the biology of CSCs in pediatric brain tumors and other cancer types and to preclinically test therapeutic agents.


Asunto(s)
Neoplasias Encefálicas/patología , Ciclo Celular , Reparación del ADN , Etopósido/farmacología , Glioblastoma/patología , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Adolescente , Animales , Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Niño , Preescolar , Aberraciones Cromosómicas , Ensayo Cometa , Resistencia a Antineoplásicos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Células Madre Neoplásicas/trasplante , Polimorfismo de Nucleótido Simple/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Stem Cell Res ; 6(2): 158-67, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21095172

RESUMEN

There are two critical stages in the retroviral reprogramming of somatic cells to produce human induced pluripotent stem cell (hiPSC) lines. One is the production of high titer virus required to reprogram somatic cells; the other is identification of true hiPSC colonies from heterogeneous cell populations, and their isolation and expansion to generate a sustainable, pluripotent stem cell line. Here we describe simple, time-saving methods to address the current difficulties at these two critical junctures. First, we have developed a method to increase the number of infectious viral units 600-fold. Second, we have developed a TRA-1-81-based positive selection column method for isolating "true" hiPSCs from the heterogeneous cell populations, which overcomes the labor-intensive and highly subjective method of manual selection of hiPSC colonies. We have used these techniques to produce 8 hiPSC lines from human fibroblasts and we believe that they are of considerable utility to researchers in the hiPSC field.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Línea Celular , Separación Celular/métodos , Células Madre Pluripotentes Inducidas/fisiología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias
15.
Gut ; 59(8): 1037-45, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20584780

RESUMEN

BACKGROUND AND AIMS: Helicobacter pylori (H pylori) infection is a major risk factor in the development of distal gastric adenocarcinoma. Development of the invasive phenotype is associated with the phenomenon of epithelial:mesenchymal transition (EMT). Soluble heparin-binding epidermal growth factor (HB-EGF) has been implicated in this process. A study was undertaken to investigate the possibility that matrix metalloproteinase (MMP)-7 is upregulated in H pylori infection as a result of hypergastrinaemia, which may enhance shedding of HB-EGF and contribute towards EMT in gastric adenocarcinoma cell lines. METHODS: Three gastric epithelial cell lines (AGS, MGLVA1 and ST16) were co-cultured with the pathogenic H pylori strain 60190 and non-pathogenic strain Tx30a in an in vitro infection model. Gene expression was quantified by real-time PCR, HB-EGF shedding by ELISA and protein expression by immunofluorescence or immunohistochemistry. The INS-GAS mouse, a transgenic mouse model of gastric carcinogenesis which overexpresses amidated gastrin, was used to investigate the in vivo relationship between HB-EGF, MMP-7, gastrin and EMT. RESULTS: The pathogenic strain of H pylori significantly upregulated EMT-associated genes Snail, Slug and vimentin in all three gastric cell lines to a greater degree than the non-pathogenic strain. Pathogenic H pylori also upregulated HB-EGF shedding, a factor implicated in EMT, which was partially dependent on both gastrin and MMP-7 expression. Gastrin and MMP-7 siRNAs and MMP-7 neutralising antibody significantly reduced upregulation of HB-EGF shedding in H pylori infected gastric cell lines and reduced EMT gene expression. The effect of H pylori on EMT was also reversed by gastrin siRNA. Neutralisation of gastrin in the INS-GAS mouse model reduced expression of MMP-7, HB-EGF and key EMT proteins. CONCLUSION: The upregulation of MMP-7 by pathogenic H pylori is partially dependent on gastrin and may have a role in the development of gastric cancer, potentially through EMT, by indirectly increasing levels of soluble HB-EGF.


Asunto(s)
Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Proteínas de Neoplasias/biosíntesis , Neoplasias Gástricas/microbiología , Animales , Transformación Celular Neoplásica/genética , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Gastrinas/biosíntesis , Gastrinas/genética , Gastrinas/fisiología , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/patología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/fisiología , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/fisiología , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Células Tumorales Cultivadas , Regulación hacia Arriba , Virulencia
16.
Mol Cancer Ther ; 7(10): 3223-36, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18852126

RESUMEN

All four members of the human epidermal growth factor (EGF) receptor (HER) family are implicated in human cancers. Although efficacious in a subset of patients, resistance to single-targeted anti-HER therapy [i.e., cetuximab (Erbitux) and trastuzumab (Herceptin)] is often associated with coexpression of other HER family members. This may be overcome by a HER ligand binding molecule that sequesters multiple EGF-like ligands, preventing ligand-dependent receptor activation. Toward this end, we have combined the HER-1/EGFR and HER-3 ligand binding domains, dimerized with fusion of an Fc fragment of human IgG1. This resulted in a mixture of HER-1/Fc homodimer (HFD100), HER-3/Fc homodimer (HFD300), and HER-1/Fc:HER-3/Fc heterodimer (RB200), also termed Hermodulins. The purified first-generation RB200 bound EGF and neuregulin 1 (NRG1)-beta1 ligands, determined by cross-linking and direct binding studies. The binding affinity for both was approximately 10 nmol/L by dissociation-enhanced lanthanide fluorescence immunoassay using europium (Eu)-labeled ligands. Competition studies with RB200 using Eu-EGF or Eu-NRG1-beta1 revealed that RB200 bound HER-1 ligands, including transforming growth factor-alpha and heparin-binding EGF, and HER-3 ligands NRG1-alpha and NRG1-beta3. RB200 inhibited EGF- and NRG1-beta1-stimulated tyrosine phosphorylation of HER family proteins, proliferation of a diverse range of tumor cells in monolayer cell growth assays, tumor cell proliferation as a single agent and in synergy with tyrosine kinase inhibitors, lysophosphatidic acid-stimulated cell proliferation, and tumor growth in two human tumor xenograft nude mouse models. Taken together, the data reveal that RB200 has the potential to sequester multiple HER ligands and interfere with signaling by HER-1, HER-2, and HER-3.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/metabolismo , Fragmentos Fc de Inmunoglobulinas/farmacología , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Proliferación Celular/efectos de los fármacos , Clonación Molecular , Dimerización , Sinergismo Farmacológico , Factor de Crecimiento Epidérmico/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Ligandos , Ratones , Neurregulina-1/metabolismo , Fosfoserina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Receptor ErbB-2/química , Receptor ErbB-3/química , Trastuzumab
17.
Int J Cancer ; 119(1): 49-59, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16450371

RESUMEN

Parathyroid-hormone related protein (PTHrP) is the primary factor in humoral hypercalcemia of malignancy and is highly secreted by breast cancers. The pro-hormone undergoes post-translational processing and cleavage to give rise to mature secretory peptides, one of which is midregion PTHrP (38-94/95/101) containing a nuclear localisation sequence (NLS) in amino acids (87-106). The current study investigates whether the NLS in midregion PTHrP is important in breast cancer growth. PTHrP-(67-101), a midregion PTHrP fragment containing NLS-(87-101) significantly increased growth of MCF-7 and MDA-MB231 cells (126.3 and 121.3% of control respectively in serum conditions), independent of PTHR1 whereas PTHrP-(67-86), which lacks the NLS did not. Fluorescent-labelled PTHrP-(67-101) translocated to the nucleus, whereas PTHrP-(67-86) remained cytosolic and a scrambled(+NLS) peptide was not internalised. In comparison, no growth influence or uptake was seen in non-tumour breast cells (Hs578Bst). Increases in intracellular calcium mobilisation were observed in breast cancer cells stimulated with both PTHrP-(67-101) and PTHrP-(67-86) (EC(50) of 3.2 pM and 2.2 pM respectively for MCF-7 cells), whereas inositide turnover was not detected. Both nuclear uptake and calcium signalling were attenuated in the presence of EGTA, but not with U73122 or N-terminal PTHrP peptides. Our studies indicate that the NLS-containing midregion PTHrP peptide is dependent on both internalisation and nuclear translocation to induce growth in breast cancer cells. These findings highlight the importance of midregion PTHrP and its receptor in breast cancer growth and may provide potential targets for future therapeutic intervention.


Asunto(s)
Neoplasias de la Mama/metabolismo , Calcio/metabolismo , Núcleo Celular/metabolismo , Señales de Localización Nuclear/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Western Blotting , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Microscopía Confocal , Fragmentos de Péptidos/metabolismo , Transducción de Señal , Factores de Tiempo
18.
Br J Pharmacol ; 140(7): 1169-76, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14597595

RESUMEN

1. It has previously been shown that ATP and UTP stimulate P2Y receptors in vascular smooth muscle cells (VSMCs), but the nature of these receptors, in particular the contribution of P2Y2 and P2Y4 subtypes, has not been firmly established. Here we undertake a further pharmacological analysis of [3H]inositol polyphosphate responses to nucleotides in cultured rat VSMCs. 2. ATP generated a response that was partial compared to UTP, as reported earlier. 3. In the presence of a creatine phosphokinase (CPK) system for regenerating nucleoside triphosphates, the response to ATP was increased, the response to UTP was unchanged, and the difference between UTP and ATP concentration-response curves disappeared. Chromatographic analysis showed that ATP was degraded slightly faster than UTP. 4. The response to UDP was always smaller than that to UTP, but with a shallow slope and a high potency component. In the presence of hexokinase (which prevents the accumulation of ATP/UTP from ADP/UDP), the maximum response to UDP was reduced and the high-potency component of the curve was retained. By contrast, the response to ADP was weaker throughout in the presence of hexokinase. 5. ATP gamma S was an effective agonist with a similar EC50 to UTP, but with a lower maximum. ITP was a weak agonist compared with UTP. 6. Suramin was an effective antagonist of the response to UTP (pA2=4.48), but not when ATP was the agonist. However, suramin was an effective antagonist (pA2=4.45) when stimulation with ATP was in the presence of the CPK regenerating system. 7. Taken together with the results of others, these findings indicate that the response of cultured rat VSMCs to UTP and to ATP is predominantly at the P2Y2 receptor, and that there is also a response to UDP at the P2Y6 receptor.


Asunto(s)
Adenosina Trifosfato/farmacología , Músculo Liso Vascular/efectos de los fármacos , Receptores Purinérgicos P2/efectos de los fármacos , Uridina Trifosfato/farmacología , Adenosina Trifosfato/metabolismo , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Células Cultivadas , Células Clonales , Relación Dosis-Respuesta a Droga , Agonistas del Receptor Purinérgico P2 , Antagonistas del Receptor Purinérgico P2 , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Especificidad de la Especie , Suramina/farmacología , Uridina Trifosfato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA