Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 13(18)2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34572851

RESUMEN

The role of the proangiogenic factor olfactomedin-like 3 (OLFML3) in cancer is unclear. To characterize OLFML3 expression in human cancer and its role during tumor development, we undertook tissue expression studies, gene expression analyses of patient tumor samples, in vivo studies in mouse cancer models, and in vitro coculture experiments. OLFML3 was expressed at high levels, mainly in blood vessels, in multiple human cancers. We focused on colorectal cancer (CRC), as elevated expression of OLFML3 mRNA correlated with shorter relapse-free survival, higher tumor grade, and angiogenic microsatellite stable consensus molecular subtype 4 (CMS4). Treatment of multiple in vivo tumor models with OLFML3-blocking antibodies and deletion of the Olfml3 gene from mice decreased lymphangiogenesis, pericyte coverage, and tumor growth. Antibody-mediated blockade of OLFML3 and deletion of host Olfml3 decreased the recruitment of tumor-promoting tumor-associated macrophages and increased infiltration of the tumor microenvironment by NKT cells. Importantly, targeting OLFML3 increased the antitumor efficacy of anti-PD-1 checkpoint inhibitor therapy. Taken together, the results demonstrate that OLFML3 is a promising candidate therapeutic target for CRC.

2.
Cancers (Basel) ; 13(18)2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34572887

RESUMEN

Although immune checkpoint inhibitors improve median overall survival in patients with metastatic urothelial cancer (mUC), only a minority of patients benefit from it. Early blood-based response biomarkers may provide a reliable way to assess response weeks before imaging is available, enabling an early switch to other therapies. We conducted an exploratory study aimed at the identification of early markers of response to anti-PD-1 in patients with mUC. Whole blood RNA sequencing and phenotyping of peripheral blood mononuclear cells were performed on samples of 26 patients obtained before and after 2 to 6 weeks of anti-PD-1. Between baseline and on-treatment samples of patients with clinical benefit, 51 differentially expressed genes (DEGs) were identified, of which 37 were upregulated during treatment. Among the upregulated genes was PDCD1, the gene encoding PD-1. STRING network analysis revealed a cluster of five interconnected DEGs which were all involved in DNA replication or cell cycle regulation. We hypothesized that the upregulation of DNA replication/cell cycle genes is a result of T cell proliferation and we were able to detect an increase in Ki-67+ CD8+ T cells in patients with clinical benefit (median increase: 1.65%, range -0.63 to 7.06%, p = 0.012). In patients without clinical benefit, no DEGs were identified and no increase in Ki-67+ CD8+ T cells was observed. In conclusion, whole blood transcriptome profiling identified early changes in DNA replication and cell cycle regulation genes as markers of clinical benefit to anti-PD-1 in patients with urothelial cancer. Although promising, our findings require further validation before implementation in the clinic.

3.
Nat Commun ; 11(1): 1571, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32218432

RESUMEN

Estrogens and progesterone control breast development and carcinogenesis via their cognate receptors expressed in a subset of luminal cells in the mammary epithelium. How they control the extracellular matrix, important to breast physiology and tumorigenesis, remains unclear. Here we report that both hormones induce the secreted protease Adamts18 in myoepithelial cells by controlling Wnt4 expression with consequent paracrine canonical Wnt signaling activation. Adamts18 is required for stem cell activation, has multiple binding partners in the basement membrane and interacts genetically with the basal membrane-specific proteoglycan, Col18a1, pointing to the basement membrane as part of the stem cell niche. In vitro, ADAMTS18 cleaves fibronectin; in vivo, Adamts18 deletion causes increased collagen deposition during puberty, which results in impaired Hippo signaling and reduced Fgfr2 expression both of which control stem cell function. Thus, Adamts18 links luminal hormone receptor signaling to basement membrane remodeling and stem cell activation.


Asunto(s)
Proteínas ADAMTS/metabolismo , Hormonas/farmacología , Glándulas Mamarias Animales/citología , Nicho de Células Madre , Células Madre/metabolismo , Proteínas ADAMTS/deficiencia , Proteínas ADAMTS/genética , Animales , Antígenos CD/metabolismo , Línea Celular , Autorrenovación de las Células/efectos de los fármacos , Epitelio/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Fibronectinas/metabolismo , Glicoproteínas/metabolismo , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Progesterona/metabolismo , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
4.
Bioinformatics ; 35(18): 3339-3347, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30753284

RESUMEN

MOTIVATION: Unbiased clustering methods are needed to analyze growing numbers of complex datasets. Currently available clustering methods often depend on parameters that are set by the user, they lack stability, and are not applicable to small datasets. To overcome these shortcomings we used topological data analysis, an emerging field of mathematics that discerns additional feature and discovers hidden insights on datasets and has a wide application range. RESULTS: We have developed a topology-based clustering method called Two-Tier Mapper (TTMap) for enhanced analysis of global gene expression datasets. First, TTMap discerns divergent features in the control group, adjusts for them, and identifies outliers. Second, the deviation of each test sample from the control group in a high-dimensional space is computed, and the test samples are clustered using a new Mapper-based topological algorithm at two levels: a global tier and local tiers. All parameters are either carefully chosen or data-driven, avoiding any user-induced bias. The method is stable, different datasets can be combined for analysis, and significant subgroups can be identified. It outperforms current clustering methods in sensitivity and stability on synthetic and biological datasets, in particular when sample sizes are small; outcome is not affected by removal of control samples, by choice of normalization, or by subselection of data. TTMap is readily applicable to complex, highly variable biological samples and holds promise for personalized medicine. AVAILABILITY AND IMPLEMENTATION: TTMap is supplied as an R package in Bioconductor. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Perfilación de la Expresión Génica , Programas Informáticos , Algoritmos , Análisis por Conglomerados , Expresión Génica , Tamaño de la Muestra
5.
Cancer Cell ; 29(3): 407-422, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26947176

RESUMEN

Seventy-five percent of breast cancers are estrogen receptor α positive (ER⁺). Research on these tumors is hampered by lack of adequate in vivo models; cell line xenografts require non-physiological hormone supplements, and patient-derived xenografts (PDXs) are hard to establish. We show that the traditional grafting of ER⁺ tumor cells into mammary fat pads induces TGFß/SLUG signaling and basal differentiation when they require low SLUG levels to grow in vivo. Grafting into the milk ducts suppresses SLUG; ER⁺ tumor cells develop, like their clinical counterparts, in the presence of physiological hormone levels. Intraductal ER⁺ PDXs are retransplantable, predictive, and appear genomically stable. The model provides opportunities for translational research and the study of physiologically relevant hormone action in breast carcinogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Glándulas Mamarias Humanas/patología , Microambiente Tumoral/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/genética
6.
Endocrinology ; 156(10): 3442-50, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26241069

RESUMEN

Worldwide, breast cancer incidence has been increasing for decades. Exposure to reproductive hormones, as occurs with recurrent menstrual cycles, affects breast cancer risk, and can promote disease progression. Exogenous hormones and endocrine disruptors have also been implicated in increasing breast cancer incidence. Numerous in vitro studies with hormone-receptor-positive cell lines have provided insights into the complexities of hormone receptor signaling at the molecular level; in vivo additional layers of complexity add on to this. The combined use of mouse genetics and tissue recombination techniques has made it possible to disentangle hormone action in vivo and revealed that estrogens, progesterone, and prolactin orchestrate distinct developmental stages of mammary gland development. The 2 ovarian steroids that fluctuate during menstrual cycles act on a subset of mammary epithelial cells, the hormone-receptor-positive sensor cells, which translate and amplify the incoming systemic signals into local, paracrine stimuli. Progesterone has emerged as a major regulator of cell proliferation and stem cell activation in the adult mammary gland. Two progesterone receptor targets, receptor activator of NfκB ligand and Wnt4, serve as downstream paracrine mediators of progesterone receptor-induced cell proliferation and stem cell activation, respectively. Some of the findings in the mouse have been validated in human ex vivo models and by next-generation whole-transcriptome sequencing on healthy donors staged for their menstrual cycles. The implications of these insights into the basic control mechanisms of mammary gland development for breast carcinogenesis and the possible role of endocrine disruptors, in particular bisphenol A in this context, will be discussed below.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/fisiopatología , Sistema Endocrino/fisiología , Progesterona/fisiología , Animales , Compuestos de Bencidrilo/efectos adversos , Mama/metabolismo , Mama/patología , Proliferación Celular , Modelos Animales de Enfermedad , Disruptores Endocrinos/efectos adversos , Femenino , Hormonas/fisiología , Humanos , Glándulas Mamarias Animales/metabolismo , Ciclo Menstrual , Ratones , Ratones Endogámicos C57BL , Fenoles/efectos adversos , Ratas , Factores de Riesgo , Transducción de Señal , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...