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.
Blood Cancer J ; 12(7): 110, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35853853

RESUMEN

Bromodomain-containing protein 9 (BRD9), an essential component of the SWI/SNF chromatin remodeling complex termed ncBAF, has been established as a therapeutic target in a subset of sarcomas and leukemias. Here, we used novel small molecule inhibitors and degraders along with RNA interference to assess the dependency on BRD9 in the context of diverse hematological malignancies, including acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), and multiple myeloma (MM) model systems. Following depletion of BRD9 protein, AML cells undergo terminal differentiation, whereas apoptosis was more prominent in ALL and MM. RNA-seq analysis of acute leukemia and MM cells revealed both unique and common signaling pathways affected by BRD9 degradation, with common pathways including those associated with regulation of inflammation, cell adhesion, DNA repair and cell cycle progression. Degradation of BRD9 potentiated the effects of several chemotherapeutic agents and targeted therapies against AML, ALL, and MM. Our findings support further development of therapeutic targeting of BRD9, alone or combined with other agents, as a novel strategy for acute leukemias and MM.


Asunto(s)
Antineoplásicos , Leucemia Mieloide Aguda , Mieloma Múltiple , Factores de Transcripción , Antineoplásicos/farmacología , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Interferencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Cancer Res ; 82(4): 681-694, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34916221

RESUMEN

Blood-borne metastasis of breast cancer involves a series of tightly regulated sequential steps, including the growth of a primary tumor lesion, intravasation of circulating tumor cells (CTC), and adaptation in various distant metastatic sites. The genes orchestrating each of these steps are poorly understood in physiologically relevant contexts, owing to the rarity of experimental models that faithfully recapitulate the biology, growth kinetics, and tropism of human breast cancer. Here, we conducted an in vivo loss-of-function CRISPR screen in newly derived CTC xenografts, unique in their ability to spontaneously mirror the human disease, and identified specific genetic dependencies for each step of the metastatic process. Validation experiments revealed sensitivities to inhibitors that are already available, such as PLK1 inhibitors, to prevent CTC intravasation. Together, these findings present a new tool to reclassify driver genes involved in the spread of human cancer, providing insights into the biology of metastasis and paving the way to test targeted treatment approaches. SIGNIFICANCE: A loss-of-function CRISPR screen in human CTC-derived xenografts identifies genes critical for individual steps of the metastatic cascade, suggesting novel drivers and treatment opportunities for metastatic breast cancers.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Células Neoplásicas Circulantes/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , RNA-Seq/métodos , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Quinasa Tipo Polo 1
3.
Nature ; 566(7745): 553-557, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30728496

RESUMEN

A better understanding of the features that define the interaction between cancer cells and immune cells is important for the development of new cancer therapies1. However, focus is often given to interactions that occur within the primary tumour and its microenvironment, whereas the role of immune cells during cancer dissemination in patients remains largely uncharacterized2,3. Circulating tumour cells (CTCs) are precursors of metastasis in several types of cancer4-6, and are occasionally found within the bloodstream in association with non-malignant cells such as white blood cells (WBCs)7,8. The identity and function of these CTC-associated WBCs, as well as the molecular features that define the interaction between WBCs and CTCs, are unknown. Here we isolate and characterize individual CTC-associated WBCs, as well as corresponding cancer cells within each CTC-WBC cluster, from patients with breast cancer and from mouse models. We use single-cell RNA sequencing to show that in the majority of these cases, CTCs were associated with neutrophils. When comparing the transcriptome profiles of CTCs associated with neutrophils against those of CTCs alone, we detect a number of differentially expressed genes that outline cell cycle progression, leading to more efficient metastasis formation. Further, we identify cell-cell junction and cytokine-receptor pairs that define CTC-neutrophil clusters, representing key vulnerabilities of the metastatic process. Thus, the association between neutrophils and CTCs drives cell cycle progression within the bloodstream and expands the metastatic potential of CTCs, providing a rationale for targeting this interaction in treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Ciclo Celular , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/patología , Neutrófilos/patología , Animales , Neoplasias de la Mama/terapia , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Exones/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Uniones Intercelulares , Ratones , Mutación/genética , Metástasis de la Neoplasia/genética , Células Neoplásicas Circulantes/metabolismo , Neutrófilos/metabolismo , Análisis de Secuencia de ARN , Secuenciación del Exoma
4.
Cell ; 176(1-2): 98-112.e14, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30633912

RESUMEN

The ability of circulating tumor cells (CTCs) to form clusters has been linked to increased metastatic potential. Yet biological features and vulnerabilities of CTC clusters remain largely unknown. Here, we profile the DNA methylation landscape of single CTCs and CTC clusters from breast cancer patients and mouse models on a genome-wide scale. We find that binding sites for stemness- and proliferation-associated transcription factors are specifically hypomethylated in CTC clusters, including binding sites for OCT4, NANOG, SOX2, and SIN3A, paralleling embryonic stem cell biology. Among 2,486 FDA-approved compounds, we identify Na+/K+ ATPase inhibitors that enable the dissociation of CTC clusters into single cells, leading to DNA methylation remodeling at critical sites and metastasis suppression. Thus, our results link CTC clustering to specific changes in DNA methylation that promote stemness and metastasis and point to cluster-targeting compounds to suppress the spread of cancer.


Asunto(s)
Neoplasias de la Mama/genética , Metástasis de la Neoplasia/genética , Células Neoplásicas Circulantes/patología , Animales , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Proteína Homeótica Nanog/metabolismo , Metástasis de la Neoplasia/fisiopatología , Células Neoplásicas Circulantes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción SOXB1/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3
5.
Breast Cancer Res ; 20(1): 141, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30458879

RESUMEN

BACKGROUND: The presence of circulating tumor cells (CTCs) in patients with breast cancer correlates to a bad prognosis. Yet, CTCs are detectable in only a minority of patients with progressive breast cancer, and factors that influence the abundance of CTCs remain elusive. METHODS: We conducted CTC isolation and enumeration in a selected group of 73 consecutive patients characterized by progressive invasive breast cancer, high tumor load and treatment discontinuation at the time of CTC isolation. CTCs were quantified with the Parsortix microfluidic device. Clinicopathological variables, blood counts at the time of CTC isolation and detailed treatment history prior to blood sampling were evaluated for each patient. RESULTS: Among 73 patients, we detected at least one CTC per 7.5 ml of blood in 34 (46%). Of these, 22 (65%) had single CTCs only, whereas 12 (35%) featured both single CTCs and CTC clusters. Treatment with the monoclonal antibody denosumab correlated with the absence of CTCs, both when considering all patients and when considering only those with bone metastasis. We also found that low red blood cell count was associated with the presence of CTCs, whereas high CA 15-3 tumor marker, high mean corpuscular volume, high white blood cell count and high mean platelet volume associated specifically with CTC clusters. CONCLUSIONS: In addition to blood count correlatives to single and clustered CTCs, we found that denosumab treatment associates with most patients lacking CTCs from their peripheral circulation. Prospective studies will be needed to validate the involvement of denosumab in the prevention of CTC generation.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Denosumab/farmacología , Eritrocitos , Células Neoplásicas Circulantes/efectos de los fármacos , Anciano , Antineoplásicos/uso terapéutico , Mama/patología , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Recuento de Células/métodos , Denosumab/uso terapéutico , Progresión de la Enfermedad , Femenino , Humanos , Técnicas Analíticas Microfluídicas/métodos , Persona de Mediana Edad , Invasividad Neoplásica/patología , Pronóstico , Estudios Retrospectivos
6.
Br J Cancer ; 119(4): 487-491, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30065256

RESUMEN

Human glioblastoma (GBM) is a highly aggressive, invasive and hypervascularised malignant brain cancer. Individual circulating tumour cells (CTCs) are sporadically found in GBM patients, yet it is unclear whether multicellular CTC clusters are generated in this disease and whether they can bypass the physical hurdle of the blood-brain barrier.  Here, we assessed CTC presence and composition at multiple time points in 13 patients with progressing GBM during an open-label phase 1/2a study with the microtubule inhibitor BAL101553. We observe CTC clusters ranging from 2 to 23 cells and present at multiple sampling time points in a GBM patient with pleomorphism and extensive necrosis, throughout disease progression. Exome sequencing of GBM CTC clusters highlights variants in 58 cancer-associated genes including ATM, PMS2, POLE, APC, XPO1, TFRC, JAK2, ERBB4 and ALK. Together, our findings represent the first evidence of the presence of CTC clusters in GBM.


Asunto(s)
Bencimidazoles/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Células Neoplásicas Circulantes/patología , Oxadiazoles/administración & dosificación , Animales , Bencimidazoles/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Recuento de Células , Análisis por Conglomerados , Progresión de la Enfermedad , Femenino , Redes Reguladoras de Genes/efectos de los fármacos , Variación Genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , Masculino , Ratones , Mutación , Células Neoplásicas Circulantes/química , Células Neoplásicas Circulantes/efectos de los fármacos , Oxadiazoles/farmacología , Secuenciación del Exoma , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Diagnostics (Basel) ; 8(2)2018 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-29747380

RESUMEN

The field of cancer diagnostics has recently been impacted by new and exciting developments in the area of liquid biopsy. A liquid biopsy is a minimally invasive alternative to surgical biopsies of solid tissues, typically achieved through the withdrawal of a blood sample or other body fluids, allowing the interrogation of tumor-derived material including circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) fragments that are present at a given time point. In this short review, we discuss a few studies that summarize the state-of-the-art in the liquid biopsy field from a diagnostic perspective, and speculate on current challenges and expectations of implementing liquid biopsy testing for cancer diagnosis and monitoring in the clinical setting.

8.
Stem Cell Reports ; 9(1): 329-341, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28579394

RESUMEN

Primordial germ cells (PGCs) are the earliest embryonic progenitors in the germline. Correct formation of PGCs is critical to reproductive health as an adult. Recent work has shown that primate PGCs can be differentiated from pluripotent stem cells; however, a bioassay that supports their identity as transplantable germ cells has not been reported. Here, we adopted a xenotransplantation assay by transplanting single-cell suspensions of human and nonhuman primate embryonic Macaca mulatta (rhesus macaque) testes containing PGCs into the seminiferous tubules of adult busulfan-treated nude mice. We discovered that both human and nonhuman primate embryonic testis are xenotransplantable, generating colonies while not generating tumors. Taken together, this work provides two critical references (molecular and functional) for defining transplantable primate PGCs. These results provide a blueprint for differentiating pluripotent stem cells to transplantable PGC-like cells in a species that is amenable to transplantation and fertility studies.


Asunto(s)
Células Germinativas/trasplante , Túbulos Seminíferos/cirugía , Testículo/embriología , Testículo/trasplante , Animales , Busulfano/uso terapéutico , Femenino , Humanos , Inmunosupresores/uso terapéutico , Macaca mulatta , Masculino , Ratones Desnudos , Trasplante Heterólogo/métodos
9.
ESMO Open ; 1(4): e000078, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27843628

RESUMEN

The development of a metastatic disease is recognised as the cause of death of over 90% of patients diagnosed with cancer. Understanding the biological features of metastasis has been hampered for a long time by the difficulties to study widespread cancerous lesions in patients, and by the absence of reliable methods to isolate viable metastatic cells during disease progression. These difficulties negatively impact on our ability to develop new agents that are tailored to block the spread of cancer. Yet, recent advances in specialised devices for the isolation of circulating tumour cells (CTCs), hand-in-hand with technologies that enable single cell resolution interrogation of their genome and transcriptome, are now paving the way to understanding those molecular mechanisms that drive the formation of metastasis. In this review, we aim to summarise some of the latest discoveries in CTC biology in the context of several types of cancer, and to highlight those findings that have a potential to improve the clinical management of patients with metastatic cancer.

10.
Biol Direct ; 11: 33, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27457474

RESUMEN

UNLABELLED: More than 90 % of cancer-related deaths are due to the development of a systemic metastatic disease. Clearly, much remains to be understood about the biological principles that govern human cancer metastasis, aiming at the ambitious objective to decrease metastasis-related mortality in patients. For many years, research on metastasis has been conducted in great part on experimental mouse models, mainly because of the difficulties in sampling, longitudinal studies, and molecular interrogation of a human metastatic disease. However, recently, extraordinary advances in microfluidic technologies are allowing the isolation and characterization of human circulating tumor cells (CTCs) that escaped a primary tumor mass and are in the process of seeding a distant metastasis. Analysis of human CTCs has now revealed important features of cancer metastasis, such as the high metastatic potential of CTC-clusters compared to single CTCs, the dynamic expression of epithelial and mesenchymal markers on CTCs during treatment, and the possibility to culture CTCs from patients for a real-time and individualized testing of drug susceptibility. Nevertheless, several aspects of CTC biology remain unsolved, such as the characterization of the stem-like cell population among human CTCs. Here, we focus on describing the latest findings in the CTC field, and discuss them in the context of cancer stem cell biology. Defining the molecular features of those few metastasis-initiating, stem-like CTCs holds the exceptional promise to develop metastasis-tailored therapies for patients with cancer. REVIEWERS: This article was reviewed by Elisa Cimetta, Luca Pellegrini and Sirio Dupont (nominated by LP).


Asunto(s)
Células Neoplásicas Circulantes/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Biomarcadores de Tumor/análisis , Humanos , Ratones
11.
Cell ; 161(6): 1425-36, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26004067

RESUMEN

Global DNA demethylation in humans is a fundamental process that occurs in pre-implantation embryos and reversion to naive ground state pluripotent stem cells (PSCs). However, the extent of DNA methylation reprogramming in human germline cells is unknown. Here, we performed whole-genome bisulfite sequencing (WGBS) and RNA-sequencing (RNA-seq) of human prenatal germline cells from 53 to 137 days of development. We discovered that the transcriptome and methylome of human germline is distinct from both human PSCs and the inner cell mass (ICM) of human blastocysts. Using this resource to monitor the outcome of global DNA demethylation with reversion of primed PSCs to the naive ground state, we uncovered hotspots of ultralow methylation at transposons that are protected from demethylation in the germline and ICM. Taken together, the human germline serves as a valuable in vivo tool for monitoring the epigenome of cells that have emerged from a global DNA demethylation event.


Asunto(s)
Blastocisto/metabolismo , Metilación de ADN , Embrión de Mamíferos/metabolismo , Células Germinativas/metabolismo , Masa Celular Interna del Blastocisto , Células Madre Embrionarias/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino
12.
EMBO J ; 34(6): 748-58, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25519955

RESUMEN

PRMT5 is a type II protein arginine methyltransferase with roles in stem cell biology, reprograming, cancer and neurogenesis. During embryogenesis in the mouse, it was hypothesized that PRMT5 functions with the master germline determinant BLIMP1 to promote primordial germ cell (PGC) specification. Using a Blimp1-Cre germline conditional knockout, we discovered that Prmt5 has no major role in murine germline specification, or the first global epigenetic reprograming event involving depletion of cytosine methylation from DNA and histone H3 lysine 9 dimethylation from chromatin. Instead, we discovered that PRMT5 functions at the conclusion of PGC reprograming I to promote proliferation, survival and expression of the gonadal germline program as marked by MVH. We show that PRMT5 regulates gene expression by promoting methylation of the Sm spliceosomal proteins and significantly altering the spliced repertoire of RNAs in mammalian embryonic cells and primordial cells.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Germinativas/citología , Proteína Metiltransferasas/metabolismo , Factores de Transcripción/genética , Animales , Western Blotting , Biología Computacional , Metilación de ADN , Cartilla de ADN/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Genotipo , Células Germinativas/enzimología , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteína-Arginina N-Metiltransferasas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Empalmosomas/metabolismo
13.
Cell Stem Cell ; 15(4): 393-394, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25280211

RESUMEN

In stem cell biology, the dynamic addition and removal of 5-methylcytosines (5mCs) are necessary for lineage differentiation, nuclear reprogramming, and embryonic development. Recent investigations have sought to understand the mechanisms of how 5mCs are added and in particular how 5mCs are removed from DNA during embryogenesis.


Asunto(s)
Núcleo Celular/metabolismo , Metilación de ADN/genética , Mamíferos/genética , Animales , Femenino , Masculino
14.
Stem Cell Rev Rep ; 10(2): 230-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24477620

RESUMEN

Human pluripotent stem cells (PSCs) are critical in vitro tools for understanding mechanisms that regulate lineage differentiation in the human embryo as well as a potentially unlimited supply of stem cells for regenerative medicine. Pluripotent human and mouse embryonic stem cells (ESCs) derived from the inner cell mass of blastocysts share a similar transcription factor network to maintain pluripotency and self-renewal, yet there are considerable molecular differences reflecting the diverse environments in which mouse and human ESCs are derived. In the current study we evaluated the role of Protein arginine methyltransferase 5 (PRMT5) in human ESC (hESC) self-renewal and pluripotency given its critical role in safeguarding mouse ESC pluripotency. Unlike the mouse, we discovered that PRMT5 has no role in hESC pluripotency. Using microarray analysis we discovered that a significant depletion in PRMT5 RNA and protein from hESCs changed the expression of only 78 genes, with the majority being repressed. Functionally, we discovered that depletion of PRMT5 had no effect on expression of OCT4, NANOG or SOX2, and did not prevent teratoma formation. Instead, we show that PRMT5 functions in hESCs to regulate proliferation in the self-renewing state by regulating the fraction of cells in Gap 1 (G1) of the cell cycle and increasing expression of the G1 cell cycle inhibitor P57. Taken together our data unveils a distinct role for PRMT5 in hESCs and identifies P57 as new target.


Asunto(s)
Proliferación Celular , Células Madre Embrionarias/fisiología , Proteína-Arginina N-Metiltransferasas/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre Embrionarias/trasplante , Puntos de Control de la Fase G1 del Ciclo Celular , Expresión Génica , Humanos , Ratones , Ratones SCID , Teratoma/patología
15.
Bio Protoc ; 3(12)2013 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-27441203

RESUMEN

High throughput RNA Sequencing has revolutionized transcriptome analyses. However, most available protocols require micrograms of RNA rendering this technique not feasible for analyzing small numbers of cells, including precious rare cell types isolated from human tissues or organs. Here, we used an RNA Amplification System and describe a method for preparing RNA sense-strand cDNA libraries compatible with an Illumina sequencing platform starting from limited numbers of human fetal germ cells as well as human embryonic stem cells (hESCs) isolated using Fluorescence Activated Cell Sorting (FACS). With this protocol we generated seven RNA-Seq libraries starting from 4,000 germ cells sorted from fetal ovaries (n = 2) and fetal testes (n = 2) at 16-16.5 weeks of development and 4,000 sorted hESCs (n = 3). We predict that multiplexed libraries can also be generated by replacing the single-plex 3' adapter used here with a multiplexing compatible 3' adapter and indexed PCR primers.

16.
Nat Cell Biol ; 15(1): 113-22, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23242216

RESUMEN

The generation of research-quality, clinically relevant cell types in vitro from human pluripotent stem cells requires a detailed understanding of the equivalent human cell types. Here we analysed 134 human embryonic and fetal samples from 6 to 20 developmental weeks and identified the stages at which cKIT(+) primordial germ cells (PGCs), the precursors of gametes, undergo whole-genome epigenetic reprogramming with global depletion of 5mC, H3K27me3 and H2A.Z, and the time at which imprint erasure is initiated and 5hmC is present. Using five alternative in vitro differentiation strategies combined with single-cell microfluidic analysis and a bona fide human cKIT(+) PGC signature, we show the stage of cKIT(+) PGC formation in the first 16 days of differentiation. Taken together, our study creates a resource of human germ line ontogeny that is essential for future studies aimed at in vitro differentiation and unveiling the mechanisms necessary to pass human DNA from one generation to the next.


Asunto(s)
Diferenciación Celular , Impresión Genómica , Células Germinativas/metabolismo , Células Germinativas/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Cultivadas , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Metilación de ADN , Embrión de Mamíferos/citología , Células Madre Embrionarias/metabolismo , Femenino , Feto/citología , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Humanos , Masculino , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ovario/citología , Ovario/embriología , Ovario/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Análisis de la Célula Individual , Testículo/citología , Testículo/embriología , Testículo/metabolismo , Transcriptoma
17.
PLoS One ; 6(12): e28960, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22194959

RESUMEN

The cell intrinsic programming that regulates mammalian primordial germ cell (PGC) development in the pre-gonadal stage is challenging to investigate. To overcome this we created a transgene-free method for generating PGCs in vitro (iPGCs) from mouse embryonic stem cells (ESCs). Using labeling for SSEA1 and cKit, two cell surface molecules used previously to isolate presumptive iPGCs, we show that not all SSEA1+/cKit+ double positive cells exhibit a PGC identity. Instead, we determined that selecting for cKit(bright) cells within the SSEA1+ fraction significantly enriches for the putative iPGC population. Single cell analysis comparing SSEA1+/cKit(bright) iPGCs to ESCs and embryonic PGCs demonstrates that 97% of single iPGCs co-express PGC signature genes Blimp1, Stella, Dnd1, Prdm14 and Dazl at similar levels to e9.5-10.5 PGCs, whereas 90% of single mouse ESC do not co-express PGC signature genes. For the 10% of ESCs that co-express PGC signature genes, the levels are significantly lower than iPGCs. Microarray analysis shows that iPGCs are transcriptionally distinct from ESCs and repress gene ontology groups associated with mesoderm and heart development. At the level of chromatin, iPGCs contain 5-methyl cytosine bases in their DNA at imprinted and non-imprinted loci, and are enriched in histone H3 lysine 27 trimethylation, yet do not have detectable levels of Mvh protein, consistent with a Blimp1-positive pre-gonadal PGC identity. In order to determine whether iPGC formation is dependent upon Blimp1, we generated Blimp1 null ESCs and found that loss of Blimp1 significantly depletes SSEA1/cKit(bright) iPGCs. Taken together, the generation of Blimp1-positive iPGCs from ESCs constitutes a robust model for examining cell-intrinsic regulation of PGCs during the Blimp1-positive stage of development.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Análisis de la Célula Individual/métodos , Factores de Transcripción/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Separación Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Gónadas/citología , Antígeno Lewis X/metabolismo , Mesodermo/metabolismo , Ratones , Modelos Biológicos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transcripción Genética , Transgenes
18.
Nucleic Acids Res ; 37(20): 6950-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19755500

RESUMEN

Human Adenovirus type 5 encodes two short RNA polymerase III transcripts, the virus-associated (VA) RNAI and VA RNAII, which can adopt stable hairpin structures that resemble micro-RNA precursors. The terminal stems of the VA RNAs are processed into small RNAs (mivaRNAs) that are incorporated into RISC. It has been reported that VA RNAI has two transcription initiation sites, which produce two VA RNAI species; a major species, VA RNAI(G), which accounts for 75% of the VA RNAI pool, and a minor species, VA RNAI(A), which initiates transcription three nucleotides upstream compared to VA RNAI(G). We show that this 5'-heterogeneity results in a dramatic difference in RISC assembly. Thus, both VA RNAI(G) and VA RNAI(A) are processed by Dicer at the same position in the terminal stem generating the same 3'-strand mivaRNA. This mivaRNA is incorporated into RISC with 200-fold higher efficiency compared to the 5'-strand of mivaRNAI. Of the small number of 5'-strands used in RISC assembly only VA RNAI(A) generated active RISC complexes. We also show that the 3'-strand of mivaRNAI, although being the preferred substrate for RISC assembly, generates unstable RISC complexes with a low in vitro cleavage activity, only around 2% compared to RISC assembled on the VA RNAI(A) 5'-strand.


Asunto(s)
Adenovirus Humanos/genética , Interferencia de ARN , ARN Viral/química , ARN Viral/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Mutación , Fosforilación , Ribonucleasa III/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...