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
2.
Cell Death Dis ; 15(1): 98, 2024 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-38286983

RESUMEN

Extracellular matrix (ECM) remodeling has been implicated in the tumor malignant progression and immune escape in glioblastoma (GBM). Runt-related transcription factor 1 (RUNX1) is a vital transcriptional factor for promoting tumorigenesis and invasion in mesenchymal subtype of GBM. But the correlation between RUNX1 and ECM genes expression and regulatory mechanism of RUNX1 on ECM genes expression remain poorly understood to date. In this study, by using integral analysis of chromatin immunoprecipitation-sequencing and RNA sequencing, we reported that RUNX1 positively regulated the expression of various ECM-related genes, including Fibronectin 1 (FN1), Collagen type IV alpha 1 chain (COL4A1), and Lumican (LUM), in GBM. Mechanistically, we demonstrated that RUNX1 interacted with Nucleophosmin 1 (NPM1) to maintain the chromatin accessibility and facilitate FOS Like 2, AP-1 Transcription Factor Subunit (FOSL2)-mediated transcriptional activation of ECM-related genes, which was independent of RUNX1's transcriptional function. ECM remodeling driven by RUNX1 promoted immunosuppressive microenvironment in GBM. In conclusion, this study provides a novel mechanism of RUNX1 binding to NPM1 in driving the ECM remodeling and GBM progression.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Activación Transcripcional , Histonas/metabolismo , Matriz Extracelular/metabolismo , Microambiente Tumoral/genética , Antígeno 2 Relacionado con Fos/genética
4.
Cell Regen ; 12(1): 31, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37656237

RESUMEN

Human pluripotent stem cells (hPSCs) have been suggested as a potential source for the production of blood cells for clinical application. In two decades, almost all types of blood cells can be successfully generated from hPSCs through various differentiated strategies. Meanwhile, with a deeper understanding of hematopoiesis, higher efficiency of generating progenitors and precursors of blood cells from hPSCs is achieved. However, how to generate large-scale mature functional cells from hPSCs for clinical use is still difficult. In this review, we summarized recent approaches that generated both hematopoietic stem cells and mature lineage cells from hPSCs, and remarked their efficiency and mechanisms in producing mature functional cells. We also discussed the major challenges in hPSC-derived products of blood cells and provided some potential solutions. Our review summarized efficient, simple, and defined methodologies for developing good manufacturing practice standards for hPSC-derived blood cells, which will facilitate the translation of these products into the clinic.

5.
Cell Prolif ; 56(10): e13457, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36959757

RESUMEN

Polycomb group (PcG) proteins are critical chromatin regulators for cell fate control. The mono-ubiquitylation on histone H2AK119 (H2AK119ub1) is one of the well-recognized mechanisms for Polycomb repressive complex 1 (PRC1)-mediated transcription repression. Unexpectedly, the specific H2AK119 deubiquitylation complex composed by additional sex comb-like proteins and BAP1 has also been genetically characterized as Polycomb repressive deubiquitnase (PR-DUB) for unclear reasons. However, it remains a mystery whether and how PR-DUB deficiency affects chromatin states and cell fates through impaired PcG silencing. Here through a careful epigenomic analysis, we demonstrate that a bulk of H2AK119ub1 is diffusely distributed away from promoter regions and their enrichment is positively correlated with PRC1 occupancy. Upon deletion of Asxl2 in mouse embryonic stem cells (ESCs), a pervasive gain of H2AK119ub1 is coincident with increased PRC1 sampling at chromatin. Accordingly, PRC1 is significantly lost from a subset of highly occupied promoters, leading to impaired silencing of associated genes before and after lineage differentiation of Asxl2-null ESCs. Therefore, our study highlights the importance of genome-wide H2AK119ub1 restriction by PR-DUB in safeguarding robust PRC1 deposition and its roles in developmental regulation.


Asunto(s)
Proteínas de Drosophila , Histonas , Animales , Ratones , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Histonas/metabolismo , Cromatina , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Ubiquitinación , Proteínas de Drosophila/metabolismo
6.
Theranostics ; 12(17): 7491-7508, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36438487

RESUMEN

Rationale: Cancer associated fibroblasts (CAFs) are a subpopulation of cells within the tumor microenvironment that usually promote cancer progression and metastasis. Hence it is critical to find out the driving factors and mechanisms for the development of CAFs from normal fibroblasts (NFs) in response to sustained stimulation of cancer cells. Here we perform transcriptomic and epigenomic analyses in paired NFs and CAFs associated with breast cancer metastasis to investigate the molecular mechanisms for stromal fibroblasts reprogramming. Methods: We conducted transcriptomic analyses in paired NFs and CAFs isolated from clinical specimens of breast cancer patients with metastasis. Meanwhile, genome-wide mapping of histone marks H3K4me1 and H3K27ac was also performed to characterize CAF-specific enhancer landscape. The function and mechanisms of activated JUN in stromal fibroblasts were studied using in vitro and in vivo models. Results: We have identified CAF-specific signature genes and activated enhancers, which are significantly associated with pro-metastatic programs. Among the CAF activated enhancers, FOS and JUN family of transcription factors are enriched. In line with this, we find that JUN protein is highly activated in the stroma of metastatic breast cancers. And through gain and loss-of-function studies, we demonstrate that activated JUN is necessary and sufficient to remodel enhancers and maintain the activation of CAF-specific enhancers, and thereby promotes breast cancer invasiveness in a non-cell-autonomous manner. Conclusions: Our study gets an insight into the transcriptomic features of invasive breast stroma and transcription regulatory mechanisms for stroma cell transformation, providing a proof-of-concept of stroma-targeting strategy in cancer treatment.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Neoplasias Primarias Secundarias , Humanos , Femenino , Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias de la Mama/patología , Microambiente Tumoral/genética , Neoplasias Primarias Secundarias/patología , Melanoma Cutáneo Maligno
7.
Mol Cell ; 82(6): 1169-1185.e7, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35202573

RESUMEN

Polycomb group (PcG) proteins are essential for post-implantation development by depositing repressive histone modifications at promoters, mainly CpG islands (CGIs), of developmental regulator genes. However, promoter PcG marks are erased after fertilization and de novo established in peri-implantation embryos, coinciding with the transition from naive to primed pluripotency. Nevertheless, the molecular basis for this establishment remains unknown. In this study, we show that the expression of the long KDM2B isoform (KDM2BLF), which contains the demethylase domain, is specifically induced at peri-implantation and that its H3K36me2 demethylase activity is required for PcG enrichment at CGIs. Moreover, KDM2BLF interacts with BRG1/BRM-associated factor (BAF) and stabilizes BAF occupancy at CGIs for subsequent gain of accessibility, which precedes PcG enrichment. Consistently, KDM2BLF inactivation results in significantly delayed post-implantation development. In summary, our data unveil dynamic chromatin configuration of CGIs during exit from naive pluripotency and provide a conceptual framework for the spatiotemporal establishment of PcG functions.


Asunto(s)
Cromatina , Proteínas de Drosophila , Islas de CpG , Proteínas de Drosophila/metabolismo , Código de Histonas , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas
8.
J Cell Mol Med ; 25(14): 6695-6708, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34114337

RESUMEN

Surface topography acts as an irreplaceable role in the long-term success of intraosseous implants. In this study, we prepared the hierarchical micro/nano topography using selective laser melting combined with alkali heat treatment (SLM-AHT) and explored the underlying mechanism of SLM-AHT surface-elicited osteogenesis. Our results show that cells cultured on SLM-AHT surface possess the largest number of mature FAs and exhibit a cytoskeleton reorganization compared with control groups. SLM-AHT surface could also significantly upregulate the expression of the cell adhesion-related molecule p-FAK, the osteogenic differentiation-related molecules RUNX2 and OCN as well as the mTORC2 signalling pathway key molecule Rictor. Notably, after the knocked-down of Rictor, there were no longer significant differences in the gene expression levels of the cell adhesion-related molecules and osteogenic differentiation-related molecules among the three titanium surfaces, and the cells on SLM-AHT surface failed to trigger cytoskeleton reorganization. In conclusion, the results suggest that mTORC2 can regulate the hierarchical micro/nano topography-mediated osteogenesis via cell adhesion and cytoskeletal reorganization.


Asunto(s)
Adhesión Celular/genética , Diferenciación Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Osteocalcina/genética , Osteogénesis/genética , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citoesqueleto/genética , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Polimerizacion/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Propiedades de Superficie/efectos de los fármacos , Titanio/farmacología
9.
J Exp Med ; 218(5)2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33755722

RESUMEN

The oncogene DEK is found fused with the NUP214 gene creating oncoprotein DEK-NUP214 that induces acute myeloid leukemia (AML) in patients, and secreted DEK protein functions as a hematopoietic cytokine to regulate hematopoiesis; however, the intrinsic role of nuclear DEK in hematopoietic stem cells (HSCs) remains largely unknown. Here, we show that HSCs lacking DEK display defects in long-term self-renew capacity, eventually resulting in impaired hematopoiesis. DEK deficiency reduces quiescence and accelerates mitochondrial metabolism in HSCs, in part, dependent upon activating mTOR signaling. At the molecular level, DEK recruits the corepressor NCoR1 to repress acetylation of histone 3 at lysine 27 (H3K27ac) and restricts the chromatin accessibility of HSCs, governing the expression of quiescence-associated genes (e.g., Akt1/2, Ccnb2, and p21). Inhibition of mTOR activity largely restores the maintenance and potential of Dek-cKO HSCs. These findings highlight the crucial role of nuclear DEK in preserving HSC potential, uncovering a new link between chromatin remodelers and HSC homeostasis, and have clinical implications.


Asunto(s)
Proteínas de Unión al ADN/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas/metabolismo , Animales , Autorrenovación de las Células/genética , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/citología , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
10.
Sci Adv ; 7(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523829

RESUMEN

Proper formation of area identities of the cerebral cortex is crucial for cognitive functions and social behaviors of the brain. It remains largely unknown whether epigenetic mechanisms, including histone methylation, regulate cortical arealization. Here, we removed SETD2, the methyltransferase for histone 3 lysine-36 trimethylation (H3K36me3), in the developing dorsal forebrain in mice and showed that Setd2 is required for proper cortical arealization and the formation of cortico-thalamo-cortical circuits. Moreover, Setd2 conditional knockout mice exhibit defects in social interaction, motor learning, and spatial memory, reminiscent of patients with the Sotos-like syndrome bearing SETD2 mutations. SETD2 maintains the expression of clustered protocadherin (cPcdh) genes in an H3K36me3 methyltransferase-dependent manner. Aberrant cortical arealization was recapitulated in cPcdh heterozygous mice. Together, our study emphasizes epigenetic mechanisms underlying cortical arealization and pathogenesis of the Sotos-like syndrome.

11.
Development ; 148(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-35020876

RESUMEN

The production and expansion of intermediate progenitors (IPs) are essential for neocortical neurogenesis during development and over evolution. Here, we have characterized an epigenetic circuit that precisely controls neurogenic programs, particularly properties of IPs, during neocortical development. The circuit comprises a long non-coding RNA (LncBAR) and the BAF (SWI/SNF) chromatin-remodeling complex, which transcriptionally maintains the expression of Zbtb20. LncBAR knockout neocortex contains more deep-layer but fewer upper-layer projection neurons. Intriguingly, loss of LncBAR promotes IP production, but paradoxically prolongs the duration of the cell cycle of IPs during mid-later neocortical neurogenesis. Moreover, in LncBAR knockout mice, depletion of the neural progenitor pool at embryonic stage results in fewer adult neural progenitor cells in the subventricular zone of lateral ventricles, leading to a failure in adult neurogenesis to replenish the olfactory bulb. LncBAR binds to BRG1, the core enzymatic component of the BAF chromatin-remodeling complex. LncBAR depletion enhances association of BRG1 with the genomic locus of, and suppresses the expression of, Zbtb20, a transcription factor gene known to regulate both embryonic and adult neurogenesis. ZBTB20 overexpression in LncBAR-knockout neural precursors reverses compromised cell cycle progressions of IPs.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Neurogénesis/genética , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Animales , Ciclo Celular/genética , Desarrollo Embrionario/genética , Epigénesis Genética/genética , Ratones , Ratones Noqueados , Neocórtex/crecimiento & desarrollo , Neocórtex/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo
12.
PLoS Biol ; 18(11): e3000749, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33253175

RESUMEN

Clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated (Cas) 9 has been widely used far beyond genome editing. Fusions of deactivated Cas9 (dCas9) to transcription effectors enable interrogation of the epigenome and controlling of gene expression. However, the large transgene size of dCas9-fusion hinders its applications especially in somatic tissues. Here, we develop a robust CRISPR interference (CRISPRi) system by transgenic expression of doxycycline (Dox) inducible dCas9-KRAB in mouse embryonic stem cells (iKRAB ESC). After introduction of specific single-guide RNAs (sgRNAs), the induced dCas9-KRAB efficiently maintains gene inactivation, although it modestly down-regulates the expression of active genes. The proper timing of Dox addition during cell differentiation or reprogramming allows us to study or screen spatiotemporally activated promoters or enhancers and thereby the gene functions. Furthermore, taking the ESC for blastocyst injection, we generate an iKRAB knock-in (KI) mouse model that enables the shutdown of gene expression and loss-of-function (LOF) studies ex vivo and in vivo by a simple transduction of gRNAs. Thus, our inducible CRISPRi ESC line and KI mouse provide versatile and convenient platforms for functional interrogation and high-throughput screens of specific genes and potential regulatory elements in the setting of development or diseases.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Factores de Transcripción de Tipo Kruppel/genética , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Doxiciclina/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Expresión Génica/genética , Silenciador del Gen/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Mutación con Pérdida de Función/genética , Ratones , Ratones Noqueados , Modelos Biológicos , ARN Guía de Kinetoplastida/genética , Reproducibilidad de los Resultados , Transgenes/genética
13.
Genome Biol ; 21(1): 48, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32093739

RESUMEN

Recently, several non-classical functions of histone modification regulators (HMRs), independent of their known histone modification substrates and products, have been reported to be essential for specific cellular processes. However, there is no framework designed for identifying such functions systematically. Here, we develop ncHMR detector, the first computational framework to predict non-classical functions and cofactors of a given HMR, based on ChIP-seq data mining. We apply ncHMR detector in ChIP-seq data-rich cell types and predict non-classical functions of HMRs. Finally, we experimentally reveal that the predicted non-classical function of CBX7 is biologically significant for the maintenance of pluripotency.


Asunto(s)
Genómica/métodos , Código de Histonas , Programas Informáticos , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Histonas/química , Histonas/metabolismo , Humanos , Análisis de Secuencia de Proteína/métodos
14.
Cell Death Differ ; 27(3): 1052-1066, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31358914

RESUMEN

The initiation and transduction of DNA damage response (DDR) occur in the context of chromatin, and modifications as well as the structure of chromatin are crucial for DDR signaling. How the profound chromatin alterations are confined to DNA lesions by epigenetic factors remains largely unclear. Here, we discover that JMJD6, a Jumonji C domain-containing protein, is recruited to DNA double-strand breaks (DSBs) after microirradiation. JMJD6 controls the spreading of histone ubiquitination, as well as the subsequent accumulation of repair proteins and transcriptional silencing around DSBs, but does not regulate the initial DNA damage sensing. Furthermore, JMJD6 deficiency results in promotion of the efficiency of nonhomologous end joining (NHEJ) and homologous recombination (HR), rapid cell-cycle checkpoint recovery, and enhanced survival after irradiation. Regarding the mechanism involved, we demonstrate that JMJD6, independently of its catalytic activity, interacts with SIRT1 and recruits it to chromatin to downregulate H4K16ac around DSBs. Our study reveals JMJD6 as a modulator of the epigenome around DNA lesions, and adds to the understanding of the role of epigenetic factors in DNA damage response.


Asunto(s)
Daño del ADN , Regulación hacia Abajo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Lisina/metabolismo , Acetilación , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Silenciador del Gen , Recombinación Homóloga/genética , Humanos , Unión Proteica , Transducción de Señal , Sirtuina 1/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitinación
15.
Nat Commun ; 9(1): 4552, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30382083

RESUMEN

Transcriptional deregulation has a vital role in glioblastoma multiforme (GBM). Thus, identification of epigenetic modifiers essential for oncogenic transcriptional programs is a key to designing effective therapeutics for this deadly disease. Here we report that Protein Arginine Methyltransferase 2 (PRMT2) is highly expressed in GBM and correlated with poor prognosis. The silencing or inactivation of PRMT2 inhibits GBM cell growth and glioblastoma stem cell self-renewal in vitro, and suppresses orthotopic tumor growth, accompanied with significant deregulation of genes mainly associated with cell cycle progression and pathways in cancer. Mechanistically PRMT2 is responsible for H3R8 asymmetric methylation (H3R8me2a), whose enrichment at promoters and enhancers is closely correlated with known active histone marks and is required for the maintenance of target gene expression. Together, this study demonstrates that PRMT2 acts as a transcriptional co-activator for oncogenic gene expression programs in GBM pathogenesis and provides a rationale for PRMT2 targeting in aggressive gliomas.


Asunto(s)
Arginina/metabolismo , Neoplasias Encefálicas/genética , Carcinogénesis/genética , Carcinogénesis/patología , Glioblastoma/genética , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oncogenes , Proteína-Arginina N-Metiltransferasas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Autorrenovación de las Células , Elementos de Facilitación Genéticos/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Metilación , Regiones Promotoras Genéticas/genética , Proteína-Arginina N-Metiltransferasas/genética , Transcripción Genética
17.
Oncotarget ; 7(43): 69674-69687, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27626484

RESUMEN

Histone methyltransferase DOT1L is implicated in various biological processes including cell proliferation, differentiation and embryogenesis. Gene ablation of Dot1l results in embryonic lethality and cardiovascular defects including decreased vasculature. However, how DOT1L might contribute to the development of vasculature is not clear. Here, we report that DOT1L is required for angiogenesis. We demonstrated that silencing of DOT1L in human umbilical vein endothelial cells (HUVECs) leads to decreased cell viability, migration, tube formation, and capillary sprout formation in vitro, as well as reduced formation of functional vascular networks in matrigel plugs in vivo. Genome-wide analysis of DOT1L targets via H3K79me2 ChIP-seq annotation in HUVECs identified a number of genes including VEGFR2 that are critically involved in angiogenesis. We showed that DOT1L cooperates with transcription factor ETS-1 to stimulate the expression of VEGFR2, thereby activating ERK1/2 and AKT signaling pathways and promoting angiogenesis. Our study revealed a mechanistic role for DOT1L in the promotion of angiogenesis, adding to the understanding of the biological function of this histone methyltransferase.


Asunto(s)
Metiltransferasas/fisiología , Neovascularización Fisiológica/fisiología , Proteína Proto-Oncogénica c-ets-1/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Movimiento Celular , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/fisiología , N-Metiltransferasa de Histona-Lisina , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Activación Transcripcional , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
18.
Nat Commun ; 7: 12648, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27557628

RESUMEN

Whether transcriptional regulators are functionally involved in mitosis is a fundamental question in cell biology. Here we report that the RNF20/40 complex, a major ubiquitin ligase catalysing histone H2B monoubiquitination, interacts with the motor protein Eg5 during mitosis and participates in spindle assembly. We show that the RNF20/40 complex monoubiquitinates and stabilizes Eg5. Loss of RNF20/40 results in spindle assembly defects, cell cycle arrest and apoptosis. Consistently, depletion of either RNF20/40 or Eg5 suppresses breast cancer in vivo. Significantly, RNF20/40 and Eg5 are concurrently upregulated in human breast carcinomas and high Eg5 expression is associated with poorer overall survival of patients with luminal A, or B, breast cancer. Our study uncovers an important spindle assembly role of the RNF20/40 complex, and implicates the RNF20/40-Eg5 axis in breast carcinogenesis, supporting the pursuit of these proteins as potential targets for breast cancer therapeutic interventions.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinogénesis/metabolismo , Cinesinas/metabolismo , Huso Acromático/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis/genética , Biocatálisis , Neoplasias de la Mama/genética , Carcinogénesis/patología , Puntos de Control del Ciclo Celular/genética , Proliferación Celular , Supervivencia Celular , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Cinesinas/química , Mutación con Pérdida de Función , Lisina/metabolismo , Células MCF-7 , Ratones Desnudos , Mitosis , Unión Proteica , Estabilidad Proteica , Transducción de Señal/genética , Polos del Huso/metabolismo , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...