Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
2.
Clin Epigenetics ; 15(1): 150, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37705055

RESUMEN

BACKGROUND: Acute myeloid leukaemia (AML) is a deadly disease characterised by the uncontrolled proliferation of immature myeloid cells within the bone marrow. Altered regulation of DNA methylation is an important epigenetic driver of AML, where the hypoxic bone marrow microenvironment can help facilitate leukaemogenesis. Thus, interactions between epigenetic regulation and hypoxia signalling will have important implications for AML development and treatment. MAIN BODY: This review summarises the importance of DNA methylation and the hypoxic bone marrow microenvironment in the development, progression, and treatment of AML. Here, we focus on the role hypoxia plays on signalling and the subsequent regulation of DNA methylation. Hypoxia is likely to influence DNA methylation through altered metabolic pathways, transcriptional control of epigenetic regulators, and direct effects on the enzymatic activity of epigenetic modifiers. DNA methylation may also prevent activation of hypoxia-responsive genes, demonstrating bidirectional crosstalk between epigenetic regulation and the hypoxic microenvironment. Finally, we consider the clinical implications of these interactions, suggesting that reduced cell cycling within the hypoxic bone marrow may decrease the efficacy of hypomethylating agents. CONCLUSION: Hypoxia is likely to influence AML progression through complex interactions with DNA methylation, where the therapeutic efficacy of hypomethylating agents may be limited within the hypoxic bone marrow. To achieve optimal outcomes for AML patients, future studies should therefore consider co-treatments that can promote cycling of AML cells within the bone marrow or encourage their dissociation from the bone marrow.


Asunto(s)
Metilación de ADN , Leucemia Mieloide Aguda , Humanos , Epigénesis Genética , Leucemia Mieloide Aguda/genética , Transducción de Señal , Hipoxia/genética , Microambiente Tumoral
3.
Epigenetics Chromatin ; 15(1): 26, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35843975

RESUMEN

Embryonic development is dependent on the maternal supply of proteins through the oocyte, including factors setting up the adequate epigenetic patterning of the zygotic genome. We previously reported that one such factor is the epigenetic repressor SMCHD1, whose maternal supply controls autosomal imprinted expression in mouse preimplantation embryos and mid-gestation placenta. In mouse preimplantation embryos, X chromosome inactivation is also an imprinted process. Combining genomics and imaging, we show that maternal SMCHD1 is required not only for the imprinted expression of Xist in preimplantation embryos, but also for the efficient silencing of the inactive X in both the preimplantation embryo and mid-gestation placenta. These results expand the role of SMCHD1 in enforcing the silencing of Polycomb targets. The inability of zygotic SMCHD1 to fully restore imprinted X inactivation further points to maternal SMCHD1's role in setting up the appropriate chromatin environment during preimplantation development, a critical window of epigenetic remodelling.


Asunto(s)
Proteínas Cromosómicas no Histona , ARN Largo no Codificante , Inactivación del Cromosoma X , Animales , Blastocisto/fisiología , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Desarrollo Embrionario , Impresión Genómica , Ratones , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cromosoma X
4.
Hemasphere ; 6(6): e734, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35651714

RESUMEN

Drug resistance and treatment failure in pediatric acute lymphoblastic leukemia (ALL) are in part driven by tumor heterogeneity and clonal evolution. Although bulk tumor genomic analyses have provided some insight into these processes, single-cell sequencing has emerged as a powerful technique to profile individual cells in unprecedented detail. Since the introduction of single-cell RNA sequencing, we now have the capability to capture not only transcriptomic, but also genomic, epigenetic, and proteomic variation between single cells separately and in combination. This rapidly evolving field has the potential to transform our understanding of the fundamental biology of pediatric ALL and guide the management of ALL patients to improve their clinical outcome. Here, we discuss the impact single-cell sequencing has had on our understanding of tumor heterogeneity and clonal evolution in ALL and provide examples of how single-cell technology can be integrated into the clinic to inform treatment decisions for children with high-risk disease.

5.
Sci Rep ; 12(1): 5776, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35388081

RESUMEN

Global changes in DNA methylation are observed in development and disease, and single-cell analyses are highlighting the heterogeneous regulation of these processes. However, technical challenges associated with single-cell analysis of DNA methylation limit these studies. We present single-cell transposable element methylation sequencing (scTEM-seq) for cost-effective estimation of average DNA methylation levels. By targeting high-copy SINE Alu elements, we achieve amplicon bisulphite sequencing with thousands of loci covered in each scTEM-seq library. Parallel transcriptome analysis is also performed to link global DNA methylation estimates with gene expression. We apply scTEM-seq to KG1a acute myeloid leukaemia (AML) cells, and primary AML cells. Our method reveals global DNA methylation heterogeneity induced by decitabine treatment of KG1a cells associated with altered expression of immune process genes. We also compare global DNA methylation estimates to expression of transposable elements and find a predominance of negative correlations. Finally, we observe co-ordinated upregulation of many transposable elements in a sub-set of decitabine treated cells. By linking global DNA methylation heterogeneity with transcription, scTEM-seq will refine our understanding of epigenetic regulation in cancer and beyond.


Asunto(s)
Elementos Transponibles de ADN , Leucemia Mieloide Aguda , Metilación de ADN , Elementos Transponibles de ADN/genética , Decitabina/farmacología , Epigénesis Genética , Humanos , Leucemia Mieloide Aguda/genética , Análisis de la Célula Individual
6.
Elife ; 92020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33186096

RESUMEN

Genomic imprinting establishes parental allele-biased expression of a suite of mammalian genes based on parent-of-origin specific epigenetic marks. These marks are under the control of maternal effect proteins supplied in the oocyte. Here we report epigenetic repressor Smchd1 as a novel maternal effect gene that regulates the imprinted expression of ten genes in mice. We also found zygotic SMCHD1 had a dose-dependent effect on the imprinted expression of seven genes. Together, zygotic and maternal SMCHD1 regulate three classic imprinted clusters and eight other genes, including non-canonical imprinted genes. Interestingly, the loss of maternal SMCHD1 does not alter germline DNA methylation imprints pre-implantation or later in gestation. Instead, what appears to unite most imprinted genes sensitive to SMCHD1 is their reliance on polycomb-mediated methylation as germline or secondary imprints, therefore we propose that SMCHD1 acts downstream of polycomb imprints to mediate its function.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Impresión Genómica/genética , Animales , Blastocisto , Proteínas Cromosómicas no Histona/genética , Metilación de ADN , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Genotipo , Proteínas Fluorescentes Verdes , Masculino , Ratones , Células-Madre Neurales
7.
Cancers (Basel) ; 12(11)2020 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-33114584

RESUMEN

Myelodysplastic syndrome (MDS) is a malignancy that disrupts normal blood cell production and commonly affects our ageing population. MDS patients are diagnosed using an invasive bone marrow biopsy and high-risk MDS patients are treated with hypomethylating agents (HMAs) such as decitabine and azacytidine. However, these therapies are only effective in 50% of patients, and many develop resistance to therapy, often resulting in bone marrow failure or leukemic transformation. Therefore, there is a strong need for less invasive, diagnostic tests for MDS, novel markers that can predict response to therapy and/or patient prognosis to aid treatment stratification, as well as new and effective therapeutics to enhance patient quality of life and survival. Epigenetic modifiers such as DNA methylation, long non-coding RNAs (lncRNAs) and micro-RNAs (miRNAs) are perturbed in MDS blasts and the bone marrow micro-environment, influencing disease progression and response to therapy. This review focusses on the potential utility of epigenetic modifiers in aiding diagnosis, prognosis, and predicting treatment response in MDS, and touches on the need for extensive and collaborative research using single-cell technologies and multi-omics to test the clinical utility of epigenetic markers for MDS patients in the future.

8.
Epigenomics ; 12(13): 1139-1151, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32790506

RESUMEN

Cancer is a disease of global epigenetic dysregulation. Mutations in epigenetic regulators are common events in multiple cancer types and epigenetic therapies are emerging as a treatment option in several malignancies. A major challenge for the clinical management of cancer is the heterogeneous nature of this disease. Cancers are composed of numerous cell types and evolve over time. This heterogeneity confounds decisions regarding treatment and promotes disease relapse. The emergence of single-cell epigenomic technologies has introduced the exciting possibility of linking genetic and transcriptional heterogeneity in the context of cancer biology. The next challenge is to leverage these tools for improved patient outcomes. Here we consider how single-cell epigenomic technologies may address the current challenges faced by cancer clinicians.


Asunto(s)
Epigénesis Genética , Epigenómica , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Análisis de la Célula Individual , Epigenoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Metástasis de la Neoplasia , Neoplasias/terapia
9.
Cancer Cell ; 36(6): 660-673.e11, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31821784

RESUMEN

Inhibition of the Menin (MEN1) and MLL (MLL1, KMT2A) interaction is a potential therapeutic strategy for MLL-rearranged (MLL-r) leukemia. Structure-based design yielded the potent, highly selective, and orally bioavailable small-molecule inhibitor VTP50469. Cell lines carrying MLL rearrangements were selectively responsive to VTP50469. VTP50469 displaced Menin from protein complexes and inhibited chromatin occupancy of MLL at select genes. Loss of MLL binding led to changes in gene expression, differentiation, and apoptosis. Patient-derived xenograft (PDX) models derived from patients with either MLL-r acute myeloid leukemia or MLL-r acute lymphoblastic leukemia (ALL) showed dramatic reductions of leukemia burden when treated with VTP50469. Multiple mice engrafted with MLL-r ALL remained disease free for more than 1 year after treatment. These data support rapid translation of this approach to clinical trials.


Asunto(s)
Cromatina/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cromatina/genética , Regulación Leucémica de la Expresión Génica/genética , Reordenamiento Génico/efectos de los fármacos , Reordenamiento Génico/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética
10.
Cell Syst ; 7(1): 63-76.e12, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30031774

RESUMEN

Pluripotency is accompanied by the erasure of parental epigenetic memory, with naïve pluripotent cells exhibiting global DNA hypomethylation both in vitro and in vivo. Exit from pluripotency and priming for differentiation into somatic lineages is associated with genome-wide de novo DNA methylation. We show that during this phase, co-expression of enzymes required for DNA methylation turnover, DNMT3s and TETs, promotes cell-to-cell variability in this epigenetic mark. Using a combination of single-cell sequencing and quantitative biophysical modeling, we show that this variability is associated with coherent, genome-scale oscillations in DNA methylation with an amplitude dependent on CpG density. Analysis of parallel single-cell transcriptional and epigenetic profiling provides evidence for oscillatory dynamics both in vitro and in vivo. These observations provide insights into the emergence of epigenetic heterogeneity during early embryo development, indicating that dynamic changes in DNA methylation might influence early cell fate decisions.


Asunto(s)
Metilación de ADN/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular , Reprogramación Celular , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Embrión de Mamíferos/citología , Epigénesis Genética/genética , Epigenómica , Genoma , Impresión Genómica , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología
11.
Nat Commun ; 9(1): 781, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29472610

RESUMEN

Parallel single-cell sequencing protocols represent powerful methods for investigating regulatory relationships, including epigenome-transcriptome interactions. Here, we report a single-cell method for parallel chromatin accessibility, DNA methylation and transcriptome profiling. scNMT-seq (single-cell nucleosome, methylation and transcription sequencing) uses a GpC methyltransferase to label open chromatin followed by bisulfite and RNA sequencing. We validate scNMT-seq by applying it to differentiating mouse embryonic stem cells, finding links between all three molecular layers and revealing dynamic coupling between epigenomic layers during differentiation.


Asunto(s)
Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Nucleosomas/metabolismo , Análisis de Secuencia de ADN/métodos , Transcripción Genética , Animales , Diferenciación Celular , Metilación de ADN , Femenino , Histonas/metabolismo , Masculino , Ratones , Nucleosomas/genética , Análisis de la Célula Individual
12.
Methods Mol Biol ; 1712: 87-95, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29224070

RESUMEN

DNA methylation is an epigenetic mark implicated in the regulation of key biological processes. Using high-throughput sequencing technologies and bisulfite-based approaches, it is possible to obtain comprehensive genome-wide maps of the mammalian DNA methylation landscape with a single-nucleotide resolution and absolute quantification. However, these methods were only applicable to bulk populations of cells. Here, we present a protocol to perform whole-genome bisulfite sequencing on single cells (scBS-Seq) using a post-bisulfite adapter tagging approach. In this method, bisulfite treatment is performed prior to library generation in order to both convert unmethylated cytosines and fragment DNA to an appropriate size. Then DNA fragments are pre-amplified with concomitant integration of the sequencing adapters, and libraries are subsequently amplified and indexed by PCR. Using scBS-Seq we can accurately measure DNA methylation at up to 50% of individual CpG sites and 70% of CpG islands.


Asunto(s)
Metilación de ADN/genética , Estudio de Asociación del Genoma Completo/métodos , Análisis de la Célula Individual/métodos , Sulfitos/química , Animales , Citosina/química , ADN/química , Epigenómica , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Análisis de Secuencia de ADN , Secuenciación Completa del Genoma
14.
Genome Biol ; 18(1): 67, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28395661

RESUMEN

Recent technological advances have enabled DNA methylation to be assayed at single-cell resolution. However, current protocols are limited by incomplete CpG coverage and hence methods to predict missing methylation states are critical to enable genome-wide analyses. We report DeepCpG, a computational approach based on deep neural networks to predict methylation states in single cells. We evaluate DeepCpG on single-cell methylation data from five cell types generated using alternative sequencing protocols. DeepCpG yields substantially more accurate predictions than previous methods. Additionally, we show that the model parameters can be interpreted, thereby providing insights into how sequence composition affects methylation variability.

15.
Nat Protoc ; 12(3): 534-547, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28182018

RESUMEN

DNA methylation (DNAme) is an important epigenetic mark in diverse species. Our current understanding of DNAme is based on measurements from bulk cell samples, which obscures intercellular differences and prevents analyses of rare cell types. Thus, the ability to measure DNAme in single cells has the potential to make important contributions to the understanding of several key biological processes, such as embryonic development, disease progression and aging. We have recently reported a method for generating genome-wide DNAme maps from single cells, using single-cell bisulfite sequencing (scBS-seq), allowing the quantitative measurement of DNAme at up to 50% of CpG dinucleotides throughout the mouse genome. Here we present a detailed protocol for scBS-seq that includes our most recent developments to optimize recovery of CpGs, mapping efficiency and success rate; reduce hands-on time; and increase sample throughput with the option of using an automated liquid handler. We provide step-by-step instructions for each stage of the method, comprising cell lysis and bisulfite (BS) conversion, preamplification and adaptor tagging, library amplification, sequencing and, lastly, alignment and methylation calling. An individual with relevant molecular biology expertise can complete library preparation within 3 d. Subsequent computational steps require 1-3 d for someone with bioinformatics expertise.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Genómica/métodos , Análisis de Secuencia de ADN/métodos , Análisis de la Célula Individual/métodos , Sulfitos/farmacología , Animales , Secuencia de Bases , Islas de CpG/genética , Ratones
16.
Development ; 144(7): 1221-1234, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28174249

RESUMEN

Mouse embryonic stem (ES) cells are locked into self-renewal by shielding from inductive cues. Release from this ground state in minimal conditions offers a system for delineating developmental progression from naïve pluripotency. Here, we examine the initial transition process. The ES cell population behaves asynchronously. We therefore exploited a short-half-life Rex1::GFP reporter to isolate cells either side of exit from naïve status. Extinction of ES cell identity in single cells is acute. It occurs only after near-complete elimination of naïve pluripotency factors, but precedes appearance of lineage specification markers. Cells newly departed from the ES cell state display features of early post-implantation epiblast and are distinct from primed epiblast. They also exhibit a genome-wide increase in DNA methylation, intermediate between early and late epiblast. These findings are consistent with the proposition that naïve cells transition to a distinct formative phase of pluripotency preparatory to lineage priming.


Asunto(s)
Rastreo Celular , Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Animales , Linaje de la Célula , Autorrenovación de las Células , Metilación de ADN/genética , Regulación hacia Abajo , Embrión de Mamíferos/citología , Células Madre Embrionarias/metabolismo , Genes Reporteros , Estratos Germinativos/citología , Cinética , Ratones , Células Madre Pluripotentes/metabolismo , Trasplante de Células Madre , Factores de Transcripción/metabolismo , Transcripción Genética
17.
Genome Biol ; 17: 72, 2016 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-27091476

RESUMEN

Emerging single-cell epigenomic methods are being developed with the exciting potential to transform our knowledge of gene regulation. Here we review available techniques and future possibilities, arguing that the full potential of single-cell epigenetic studies will be realized through parallel profiling of genomic, transcriptional, and epigenetic information.


Asunto(s)
Epigenómica/métodos , Análisis de la Célula Individual/métodos , Animales , Metilación de ADN , Regulación de la Expresión Génica , Humanos
18.
Nat Methods ; 13(3): 229-232, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26752769

RESUMEN

We report scM&T-seq, a method for parallel single-cell genome-wide methylome and transcriptome sequencing that allows for the discovery of associations between transcriptional and epigenetic variation. Profiling of 61 mouse embryonic stem cells confirmed known links between DNA methylation and transcription. Notably, the method revealed previously unrecognized associations between heterogeneously methylated distal regulatory elements and transcription of key pluripotency genes.


Asunto(s)
Células Madre Embrionarias/fisiología , Epigénesis Genética/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Elementos Reguladores de la Transcripción/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Células Cultivadas , Ratones , Datos de Secuencia Molecular
19.
Breast Cancer Res ; 18(1): 4, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26738740

RESUMEN

BACKGROUND: E74-like factor 5 (ELF5) is an epithelial-specific member of the E26 transforming sequence (ETS) transcription factor family and a critical regulator of cell fate in the placenta, pulmonary bronchi, and milk-producing alveoli of the mammary gland. ELF5 also plays key roles in malignancy, particularly in basal-like and endocrine-resistant forms of breast cancer. Almost all genes undergo alternative transcription or splicing, which increases the diversity of protein structure and function. Although ELF5 has multiple isoforms, this has not been considered in previous studies of ELF5 function. METHODS: RNA-sequencing data for 6757 samples from The Cancer Genome Atlas were analyzed to characterize ELF5 isoform expression in multiple normal tissues and cancers. Extensive in vitro analysis of ELF5 isoforms, including a 116-gene quantitative polymerase chain reaction panel, was performed in breast cancer cell lines. RESULTS: ELF5 isoform expression was found to be tissue-specific due to alternative promoter use but altered in multiple cancer types. The normal breast expressed one main isoform, while in breast cancer there were subtype-specific alterations in expression. Expression of other ETS factors was also significantly altered in breast cancer, with the basal-like subtype demonstrating a distinct ETS expression profile. In vitro inducible expression of the full-length isoforms 1 and 2, as well as isoform 3 (lacking the Pointed domain) had similar phenotypic and transcriptional effects. CONCLUSIONS: Alternative promoter use, conferring differential regulatory responses, is the main mechanism governing ELF5 action rather than differential transcriptional activity of the isoforms. This understanding of expression and function at the isoform level is a vital first step in realizing the potential of transcription factors such as ELF5 as prognostic markers or therapeutic targets in cancer.


Asunto(s)
Empalme Alternativo/genética , Proteínas de Unión al ADN/genética , Neoplasias/genética , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas c-ets/genética , Animales , Proteínas de Unión al ADN/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Glándulas Mamarias Humanas/patología , Neoplasias/patología , Especificidad de Órganos , Embarazo , Regiones Promotoras Genéticas , Isoformas de Proteínas/biosíntesis , Proteínas Proto-Oncogénicas c-ets/biosíntesis , Factores de Transcripción
20.
PLoS Biol ; 13(12): e1002330, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26717410

RESUMEN

During pregnancy, the ETS transcription factor ELF5 establishes the milk-secreting alveolar cell lineage by driving a cell fate decision of the mammary luminal progenitor cell. In breast cancer, ELF5 is a key transcriptional determinant of tumor subtype and has been implicated in the development of insensitivity to anti-estrogen therapy. In the mouse mammary tumor virus-Polyoma Middle T (MMTV-PyMT) model of luminal breast cancer, induction of ELF5 levels increased leukocyte infiltration, angiogenesis, and blood vessel permeability in primary tumors and greatly increased the size and number of lung metastasis. Myeloid-derived suppressor cells, a group of immature neutrophils recently identified as mediators of vasculogenesis and metastasis, were recruited to the tumor in response to ELF5. Depletion of these cells using specific Ly6G antibodies prevented ELF5 from driving vasculogenesis and metastasis. Expression signatures in luminal A breast cancers indicated that increased myeloid cell invasion and inflammation were correlated with ELF5 expression, and increased ELF5 immunohistochemical staining predicted much shorter metastasis-free and overall survival of luminal A patients, defining a group who experienced unexpectedly early disease progression. Thus, in the MMTV-PyMT mouse mammary model, increased ELF5 levels drive metastasis by co-opting the innate immune system. As ELF5 has been previously implicated in the development of antiestrogen resistance, this finding implicates ELF5 as a defining factor in the acquisition of the key aspects of the lethal phenotype in luminal A breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias Pulmonares/secundario , Pulmón/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/virología , Permeabilidad Capilar , Proliferación Celular , Proteínas de Unión al ADN , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hemorragia/etiología , Hemorragia/prevención & control , Humanos , Leucocitos/inmunología , Leucocitos/patología , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Depleción Linfocítica , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , Proteínas de Neoplasias/genética , Neovascularización Patológica/etiología , Neovascularización Patológica/prevención & control , Infiltración Neutrófila , Poliomavirus/patogenicidad , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Supervivencia , Factores de Transcripción , Carga Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA