Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cancer Res Clin Oncol ; 149(14): 12993-13003, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37464150

RESUMEN

PURPOSE: To discriminate the risk stratification in gastrointestinal stromal tumors (GISTs) by preoperatively constructing a model of nonenhanced computed tomography (NECT). METHODS: A total of 111 GISTs patients (77 in the training group and 34 in the validation Group) from two hospitals between 2015 and 2022 were collected retrospectively. One thousand and thirty-seven radiomics features were extracted from non-contract CT images, and the optimal radiomics signature was determined by univariate analysis and LASSO regression. The radiomics model was developed and validated from the ten optimal radiomics features by three methods. Covariates (clinical features, CT findings, and immunohistochemical characteristics) were collected to establish the clinical model, and both the radiomics features and the covariates were used to build the combined model. The effectiveness of the three models was evaluated by the Delong test. RESULTS: The experimental results showed that the clinical models (75.3%, 70.6%), the radiomics models (79.2%, 79.4%) and the combined models (81.8%, 82.4%) all had high accuracy in predicting the pathological risk of GIST in both training and validation groups. The AUC values of the combined models were significantly higher in both the training groups (0.921 vs 0.822, p= 0.032) and the validation groups (0.913 vs 0.792, p= 0.019) than that of the clinical models. According to the calibration curve, the combined model nomogram is clinically useful. CONCLUSIONS: The clinical-radiomics combined model and based on NECT performed well in discriminating the risk stratification in GISTs. As a quantitative technique, radiomics is capable of predicting the malignant potential and guiding treatment preoperatively.

2.
bioRxiv ; 2023 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-37131696

RESUMEN

Understanding how the atrial and ventricular chambers of the heart maintain their distinct identity is a prerequisite for treating chamber-specific diseases. Here, we selectively inactivated the transcription factor Tbx5 in the atrial working myocardium of the neonatal mouse heart to show that it is required to maintain atrial identity. Atrial Tbx5 inactivation downregulated highly chamber specific genes such as Myl7 and Nppa , and conversely, increased the expression of ventricular identity genes including Myl2 . Using combined single nucleus transcriptome and open chromatin profiling, we assessed genomic accessibility changes underlying the altered atrial identity expression program, identifying 1846 genomic loci with greater accessibility in control atrial cardiomyocytes compared to KO aCMs. 69% of the control-enriched ATAC regions were bound by TBX5, demonstrating a role for TBX5 in maintaining atrial genomic accessibility. These regions were associated with genes that had higher expression in control aCMs compared to KO aCMs, suggesting they act as TBX5-dependent enhancers. We tested this hypothesis by analyzing enhancer chromatin looping using HiChIP and found 510 chromatin loops that were sensitive to TBX5 dosage. Of the loops enriched in control aCMs, 73.7% contained anchors in control-enriched ATAC regions. Together, these data demonstrate a genomic role for TBX5 in maintaining the atrial gene expression program by binding to atrial enhancers and preserving tissue-specific chromatin architecture of atrial enhancers.

3.
Circulation ; 147(11): 881-896, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36705030

RESUMEN

BACKGROUND: Cardiac chamber-selective transcriptional programs underpin the structural and functional differences between atrial and ventricular cardiomyocytes (aCMs and vCMs). The mechanisms responsible for these chamber-selective transcriptional programs remain largely undefined. METHODS: We nominated candidate chamber-selective enhancers (CSEs) by determining the genome-wide occupancy of 7 key cardiac transcription factors (GATA4, MEF2A, MEF2C, NKX2-5, SRF, TBX5, TEAD1) and transcriptional coactivator P300 in atria and ventricles. Candidate enhancers were tested using an adeno-associated virus-mediated massively parallel reporter assay. Chromatin features of CSEs were evaluated by performing assay of transposase accessible chromatin sequencing and acetylation of histone H3 at lysine 27-HiChIP on aCMs and vCMs. CSE sequence requirements were determined by systematic tiling mutagenesis of 29 CSEs at 5 bp resolution. Estrogen-related receptor (ERR) function in cardiomyocytes was evaluated by Cre-loxP-mediated inactivation of ERRα and ERRγ in cardiomyocytes. RESULTS: We identified 134 066 and 97 506 regions reproducibly occupied by at least 1 transcription factor or P300, in atria or ventricles, respectively. Enhancer activities of 2639 regions bound by transcription factors or P300 were tested in aCMs and vCMs by adeno-associated virus-mediated massively parallel reporter assay. This identified 1092 active enhancers in aCMs or vCMs. Several overlapped loci associated with cardiovascular disease through genome-wide association studies, and 229 exhibited chamber-selective activity in aCMs or vCMs. Many CSEs exhibited differential chromatin accessibility between aCMs and vCMs, and CSEs were enriched for aCM- or vCM-selective acetylation of histone H3 at lysine 27-anchored loops. Tiling mutagenesis of 29 CSEs identified the binding motif of ERRα/γ as important for ventricular enhancer activity. The requirement of ERRα/γ to activate ventricular CSEs and promote vCM identity was confirmed by loss of the vCM gene profile in ERRα/γ knockout vCMs. CONCLUSIONS: We identified 229 CSEs that could be useful research tools or direct therapeutic gene expression. We showed that chamber-selective multi-transcription factor, P300 occupancy, open chromatin, and chromatin looping are predictive features of CSEs. We found that ERRα/γ are essential for maintenance of ventricular identity. Finally, our gene expression, epigenetic, 3-dimensional genome, and enhancer activity atlas provide key resources for future studies of chamber-selective gene regulation.


Asunto(s)
Histonas , Miocitos Cardíacos , Humanos , Miocitos Cardíacos/metabolismo , Histonas/genética , Histonas/metabolismo , Estudio de Asociación del Genoma Completo , Lisina/genética , Lisina/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Estrógenos
4.
Nat Cardiovasc Res ; 2(10): 881-898, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38344303

RESUMEN

Understanding how the atrial and ventricular heart chambers maintain distinct identities is a prerequisite for treating chamber-specific diseases. Here, we selectively knocked out (KO) the transcription factor Tbx5 in the atrial working myocardium to evaluate its requirement for atrial identity. Atrial Tbx5 inactivation downregulated atrial cardiomyocyte (aCM) selective gene expression. Using concurrent single nucleus transcriptome and open chromatin profiling, genomic accessibility differences were identified between control and Tbx5 KO aCMs, revealing that 69% of the control-enriched ATAC regions were bound by TBX5. Genes associated with these regions were downregulated in KO aCMs, suggesting they function as TBX5-dependent enhancers. Comparing enhancer chromatin looping using H3K27ac HiChIP identified 510 chromatin loops sensitive to TBX5 dosage, and 74.8% of control-enriched loops contained anchors in control-enriched ATAC regions. Together, these data demonstrate TBX5 maintains the atrial gene expression program by binding to and preserving the tissue-specific chromatin architecture of atrial enhancers.

5.
Stem Cell Reports ; 16(5): 1331-1346, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33891867

RESUMEN

Stem cell-based embryo models by cultured pluripotent and extra-embryonic lineage stem cells are novel platforms to model early postimplantation development. We showed that induced pluripotent stem cells (iPSCs) could form ITS (iPSCs and trophectoderm stem cells) and ITX (iPSCs, trophectoderm stem cells, and XEN cells) embryos, resembling the early gastrula embryo developed in vivo. To facilitate the efficient and unbiased analysis of the stem cell-based embryo model, we set up a machine learning workflow to extract multi-dimensional features and perform quantification of ITS embryos using 3D images collected from a high-content screening system. We found that different PSC lines differ in their ability to form embryo-like structures. Through high-content screening of small molecules and cytokines, we identified that BMP4 best promoted the morphogenesis of the ITS embryo. Our study established an innovative strategy to analyze stem cell-based embryo models and uncovered new roles of BMP4 in stem cell-based embryo models.


Asunto(s)
Embrión de Mamíferos/citología , Células Madre Pluripotentes Inducidas/citología , Aprendizaje Automático , Animales , Proteína Morfogenética Ósea 4/metabolismo , Polaridad Celular/efectos de los fármacos , Citocinas/metabolismo , Ectodermo/citología , Implantación del Embrión/efectos de los fármacos , Endodermo/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Transcriptoma/genética , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo
6.
Cell Res ; 31(6): 613-630, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33514913

RESUMEN

Organization of the genome into euchromatin and heterochromatin appears to be evolutionarily conserved and relatively stable during lineage differentiation. In an effort to unravel the basic principle underlying genome folding, here we focus on the genome itself and report a fundamental role for L1 (LINE1 or LINE-1) and B1/Alu retrotransposons, the most abundant subclasses of repetitive sequences, in chromatin compartmentalization. We find that homotypic clustering of L1 and B1/Alu demarcates the genome into grossly exclusive domains, and characterizes and predicts Hi-C compartments. Spatial segregation of L1-rich sequences in the nuclear and nucleolar peripheries and B1/Alu-rich sequences in the nuclear interior is conserved in mouse and human cells and occurs dynamically during the cell cycle. In addition, de novo establishment of L1 and B1 nuclear segregation is coincident with the formation of higher-order chromatin structures during early embryogenesis and appears to be critically regulated by L1 and B1 transcripts. Importantly, depletion of L1 transcripts in embryonic stem cells drastically weakens homotypic repeat contacts and compartmental strength, and disrupts the nuclear segregation of L1- or B1-rich chromosomal sequences at genome-wide and individual sites. Mechanistically, nuclear co-localization and liquid droplet formation of L1 repeat DNA and RNA with heterochromatin protein HP1α suggest a phase-separation mechanism by which L1 promotes heterochromatin compartmentalization. Taken together, we propose a genetically encoded model in which L1 and B1/Alu repeats blueprint chromatin macrostructure. Our model explains the robustness of genome folding into a common conserved core, on which dynamic gene regulation is overlaid across cells.


Asunto(s)
Elementos de Nucleótido Esparcido Largo , Secuencias Repetitivas de Ácidos Nucleicos , Animales , Análisis por Conglomerados , Elementos de Nucleótido Esparcido Largo/genética , Ratones , ARN , Secuencias Repetitivas de Ácidos Nucleicos/genética , Retroelementos
7.
Science ; 370(6522)2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33303584

RESUMEN

Embryo polarization is critical for mouse development; however, neither the regulatory clock nor the molecular trigger that it activates is known. Here, we show that the embryo polarization clock reflects the onset of zygotic genome activation, and we identify three factors required to trigger polarization. Advancing the timing of transcription factor AP-2 gamma (Tfap2c) and TEA domain transcription factor 4 (Tead4) expression in the presence of activated Ras homolog family member A (RhoA) induces precocious polarization as well as subsequent cell fate specification and morphogenesis. Tfap2c and Tead4 induce expression of actin regulators that control the recruitment of apical proteins on the membrane, whereas RhoA regulates their lateral mobility, allowing the emergence of the apical domain. Thus, Tfap2c, Tead4, and RhoA are regulators for the onset of polarization and cell fate segregation in the mouse.


Asunto(s)
Relojes Biológicos/fisiología , Blastocisto/fisiología , Polaridad Celular/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Musculares/fisiología , Factor de Transcripción AP-2/fisiología , Factores de Transcripción/fisiología , Proteína de Unión al GTP rhoA/fisiología , Actinas/metabolismo , Animales , Relojes Biológicos/genética , Blastocisto/citología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Polaridad Celular/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Interferencia de ARN , Factores de Transcripción de Dominio TEA , Factor de Transcripción AP-2/genética , Factores de Transcripción/genética , Proteína de Unión al GTP rhoA/genética
8.
Nature ; 587(7832): 139-144, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33116310

RESUMEN

Zygotic genome activation (ZGA) is the first transcription event in life1. However, it is unclear how RNA polymerase is engaged in initiating ZGA in mammals. Here, by developing small-scale Tn5-assisted chromatin cleavage with sequencing (Stacc-seq), we investigated the landscapes of RNA polymerase II (Pol II) binding in mouse embryos. We found that Pol II undergoes 'loading', 'pre-configuration', and 'production' during the transition from minor ZGA to major ZGA. After fertilization, Pol II is preferentially loaded to CG-rich promoters and accessible distal regions in one-cell embryos (loading), in part shaped by the inherited parental epigenome. Pol II then initiates relocation to future gene targets before genome activation (pre-configuration), where it later engages in full transcription elongation upon major ZGA (production). Pol II also maintains low poising at inactive promoters after major ZGA until the blastocyst stage, coinciding with the loss of promoter epigenetic silencing factors. Notably, inhibition of minor ZGA impairs the Pol II pre-configuration and embryonic development, accompanied by aberrant retention of Pol II and ectopic expression of one-cell targets upon major ZGA. Hence, stepwise transition of Pol II occurs when mammalian life begins, and minor ZGA has a key role in the pre-configuration of transcription machinery and chromatin for genome activation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Genoma/genética , ARN Polimerasa II/metabolismo , Cigoto/metabolismo , Alelos , Animales , Cromatina/genética , Cromatina/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/metabolismo , Epigenoma/genética , Femenino , Masculino , Herencia Materna/genética , Ratones , Ratones Endogámicos C57BL , Oocitos/enzimología , Oocitos/metabolismo , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/genética , Cigoto/citología , Cigoto/enzimología
9.
Mol Cell ; 77(4): 825-839.e7, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31837995

RESUMEN

In mammals, chromatin organization undergoes drastic reorganization during oocyte development. However, the dynamics of three-dimensional chromatin structure in this process is poorly characterized. Using low-input Hi-C (genome-wide chromatin conformation capture), we found that a unique chromatin organization gradually appears during mouse oocyte growth. Oocytes at late stages show self-interacting, cohesin-independent compartmental domains marked by H3K27me3, therefore termed Polycomb-associating domains (PADs). PADs and inter-PAD (iPAD) regions form compartment-like structures with strong inter-domain interactions among nearby PADs. PADs disassemble upon meiotic resumption from diplotene arrest but briefly reappear on the maternal genome after fertilization. Upon maternal depletion of Eed, PADs are largely intact in oocytes, but their reestablishment after fertilization is compromised. By contrast, depletion of Polycomb repressive complex 1 (PRC1) proteins attenuates PADs in oocytes, which is associated with substantial gene de-repression in PADs. These data reveal a critical role of Polycomb in regulating chromatin architecture during mammalian oocyte growth and early development.


Asunto(s)
Cromatina/química , Oocitos/crecimiento & desarrollo , Oogénesis/genética , Proteínas del Grupo Polycomb/fisiología , Animales , Blastocisto/química , Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Embrión de Mamíferos/química , Silenciador del Gen , Código de Histonas , Ratones , Oocitos/química , Transcripción Genética , Cohesinas
10.
Cell Prolif ; 52(3): e12574, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30724402

RESUMEN

Induced pluripotent stem cells (iPSCs) are reprogrammed somatic cells that gained self-renewal and differentiation capacity similar to embryonic stem cells. Taking the precious opportunity of the TianZhou-1 spacecraft mission, we studied the effect of space microgravity (µg) on the self-renewal capacity of iPSCs. Murine iPSCs carrying pluripotency reporter Oct4-GFP were used. The Oct4-EGFP-iPSCs clones were loaded into the bioreactor and exposed to µg in outer space for 14 days. The control experiment was performed in identical device but on the ground in earth gravity (1 g). iPSCs clones were compact and highly expressed Oct4 before launch. In µg condition, cells in iPSC clones spread out more rapidly than those in ground 1 g condition during the first 3 days after launch. However, in 1 g condition, as the cell density increases, the Oct4-GFP signal dropped significantly during the following 3 days. Interestingly, in µg condition, iPSCs originated from the spread-out clones during the first 3 days appeared to cluster together and reform colonies that activated strong Oct4 expression. On the other hand, iPSC clones in 1 g condition were not able to recover Oct4 expression after overgrown. Our study for the first time performed real-time imaging on the proliferation process of iPSCs in space and found that in µg condition, cell behaviour appeared to be more dynamic than on the ground.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Vuelo Espacial , Ingravidez , Animales , Reactores Biológicos , Proliferación Celular , Autorrenovación de las Células , Células Clonales , Sistemas de Computación , Ratones , Ratones Transgénicos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Regeneración
11.
Stem Cell Reports ; 12(3): 557-571, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30773485

RESUMEN

Human pluripotent stem cells (hPSCs) are susceptible to numerical and structural chromosomal alterations during long-term culture. We show that mitotic errors occur frequently in hPSCs and that prometaphase arrest leads to very rapid apoptosis in undifferentiated but not in differentiated cells. hPSCs express high levels of proapoptotic protein NOXA in undifferentiated state. Knocking out NOXA by CRISPR or upregulation of the anti-apoptosis gene BCL-XL significantly reduced mitotic cell death, allowing the survival of aneuploid cells and the formation of teratomas significantly larger than their wild-type parental hPSCs. These results indicate that the normally low threshold of apoptosis in hPSCs can safeguard their genome integrity by clearing cells undergoing abnormal division. The amplification of BCL2L1 on chromosome 20q11.21, a frequent mutation in hPSCs, although not directly oncogenic, reduces the sensitivity of hPSCs to damage caused by erroneous mitosis and increases the risk of gaining aneuploidy.


Asunto(s)
Apoptosis/genética , Supervivencia Celular/genética , Mitosis/genética , Mutación/genética , Células Madre Pluripotentes/fisiología , Aneuploidia , Proteínas Reguladoras de la Apoptosis , Muerte Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Humanos , Proteína bcl-X/genética
12.
Stem Cells Dev ; 28(6): 357-360, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30654722

RESUMEN

During space travel, exposure to microgravity may have profound influence on the physiological function of mammalian cells. In this study, we took opportunity of the Tianzhou-1 (TZ-1) mission to investigate how spaceflight may affect cardiac differentiation of mouse induced pluripotent stem cells (iPSCs). A bioreactor was engineered to perform cell culturing and the time-lapse imaging experiments on-orbit. Transgenic iPSC lines with either Oct4 or α-myosin heavy chain (αMHC) promoter driving green fluorescent protein (GFP) expression were used to study cardiomyocyte (CM) differentiation in real microgravity. The differentiation status was monitored by GFP fluorescence intensity. Interestingly, compared with cells cultured in identical environment at ground gravity, embryoid bodies (EBs) derived from Oct4 reporter iPSC downregulated GFP significantly quicker in space. Meanwhile, EBs derived from αMHC reporter iPSC activated GFP strongly 4 days after launch (P < 0.05) and lasted for 10 days afterward, indicating robust CM formation. This is the first real-time imaging study of iPSC myocardial differentiation in space. Under our experimental condition, real microgravity enhanced the CM differentiation process of iPSCs. Our study provided rare information about iPSC cardiac differentiation in space. In the future, similar automated stem cell experiments may help to realize personalized cardiac tissue biomanufacturing and drug test during space travel.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Vuelo Espacial , Ingravidez , Animales , Células Madre Pluripotentes Inducidas/citología , Ratones , Miocardio/citología , Miocitos Cardíacos/citología
13.
Protein Cell ; 10(5): 327-346, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30128894

RESUMEN

Primitive mammalian heart transforms from a single tube to a four-chambered muscular organ during a short developmental window. We found that knocking out global microRNA by deleting Dgcr8 microprocessor in Mesp1 cardiovascular progenitor cells lead to the formation of extremely dilated and enlarged heart due to defective cardiomyocyte (CM) differentiation. Transcriptome analysis revealed unusual upregulation of vascular gene expression in Dgcr8 cKO hearts. Single cell RNA sequencing study further confirmed the increase of angiogenesis genes in single Dgcr8 cKO CM. We also performed global microRNA profiling of E9.5 heart for the first time, and identified that miR-541 was transiently highly expressed in E9.5 hearts. Interestingly, introducing miR-541 back into microRNA-free CMs partially rescued their defects, downregulated angiogenesis genes and significantly upregulated cardiac genes. Moreover, miR-541 can target Ctgf and inhibit endothelial function. Our results suggest that microRNAs are required to suppress abnormal angiogenesis gene program to maintain CM differentiation.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , MicroARNs/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Células Madre Embrionarias Humanas/citología , Humanos , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/citología , Proteínas de Unión al ARN/genética , Análisis de la Célula Individual/métodos
14.
Nature ; 560(7718): E27, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29925957

RESUMEN

In this Letter, the 'Open chromatin' label in Fig. 4a should have been centred above the first three columns, and the black horizontal line underneath the label should have been removed. In addition, there should have been a vertical black line between the last two sets of panels for consistency. Minor changes have also been made to Fig. 1 and to the legend of Fig. 3. These errrors have been corrected online, and see Supplementary Information to the accompanying Amendment for the original Fig. 4.

15.
Nature ; 557(7704): 256-260, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29720659

RESUMEN

Upon fertilization, drastic chromatin reorganization occurs during preimplantation development 1 . However, the global chromatin landscape and its molecular dynamics in this period remain largely unexplored in humans. Here we investigate chromatin states in human preimplantation development using an improved assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) 2 . We find widespread accessible chromatin regions in early human embryos that overlap extensively with putative cis-regulatory sequences and transposable elements. Integrative analyses show both conservation and divergence in regulatory circuitry between human and mouse early development, and between human pluripotency in vivo and human embryonic stem cells. In addition, we find widespread open chromatin regions before zygotic genome activation (ZGA). The accessible chromatin loci are readily found at CpG-rich promoters. Unexpectedly, many others reside in distal regions that overlap with DNA hypomethylated domains in human oocytes and are enriched for transcription factor-binding sites. A large portion of these regions then become inaccessible after ZGA in a transcription-dependent manner. Notably, such extensive chromatin reorganization during ZGA is conserved in mice and correlates with the reprogramming of the non-canonical histone mark H3K4me3, which is uniquely linked to genome silencing3-5. Taken together, these data not only reveal a conserved principle that underlies the chromatin transition during mammalian ZGA, but also help to advance our understanding of epigenetic reprogramming during human early development and in vitro fertilization.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Epigénesis Genética , Genoma/genética , Cigoto/metabolismo , Animales , Sitios de Unión , Islas de CpG/genética , Metilación de ADN , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Células Madre Embrionarias/citología , Femenino , Silenciador del Gen , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Oocitos/citología , Oocitos/metabolismo , Células Madre Pluripotentes/citología , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Transposasas/metabolismo
16.
Protein Cell ; 8(9): 662-674, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28434146

RESUMEN

Coordination of cell division and cell fate is crucial for the successful development of mammalian early embryos. Aurora kinases are evolutionarily conserved serine/threonine kinases and key regulators of mitosis. Aurora kinase B (AurkB) is ubiquitously expressed while Aurora kinase C (AurkC) is specifically expressed in gametes and preimplantation embryos. We found that increasing AurkC level in one blastomere of the 2-cell embryo accelerated cell division and decreasing AurkC level slowed down mitosis. Changing AurkB level had the opposite effect. The kinase domains of AurkB and AurkC were responsible for their different ability to phosphorylate Histone H3 Serine 10 (H3S10P) and regulate metaphase timing. Using an Oct4-photoactivatable GFP fusion protein (Oct4-paGFP) and fluorescence decay after photoactivation assay, we found that AurkB overexpression reduced Oct4 retention in the nucleus. Finally, we show that blastomeres with higher AurkC level elevated pluripotency gene expression, which were inclined to enter the inner cell mass lineage and subsequently contributed to the embryo proper. Collectively, our results are the first demonstration that the activity of mitotic kinases can influence cell fate decisions in mammalian preimplantation embryos and have important implications to assisted reproduction.


Asunto(s)
Aurora Quinasa B/metabolismo , Aurora Quinasa C/metabolismo , Blastocisto/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Histonas/metabolismo , Animales , Ratones , Fosforilación/fisiología
17.
Anal Biochem ; 471: 29-37, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25447493

RESUMEN

Surface plasmon resonance imaging (SPRi) has emerged as a versatile biosensor to detect a wide range of biomolecular interactions with divergent potential applications. However, the use of this advanced-level technology for stem cell lysate study is still not much explored. Cell lysates are significant biological analytes used for disease diagnostics and proteomic studies, but their complex nature limits their use as an analyte for SPRi biosensors. Here, we review the problems associated with the use of SPRi for stem cell lysate study and examine the role of surface chemistry, running buffer, and blocking solution in order to minimize nonspecific adsorption (NSA). We detect the expression of Oct4, Sox2, Nanog, Rex1, and Lin28 biomarkers present in mouse embryonic stem cell (mESC) lysate against their corresponding antibodies immobilized on the sensor surface with reduced NSA. The current study shows that the conjunction of SPRi and microarray can be used as a label-free, high-throughput, and rapid technique for detection of biomarkers and their relative abundance in stem cell lysate study.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Resonancia por Plasmón de Superficie/métodos , Adsorción , Animales , Biomarcadores/metabolismo , Tampones (Química) , Muerte Celular , Regulación de la Expresión Génica , Ratones , Factores de Tiempo , Análisis de Matrices Tisulares
18.
Anal Biochem ; 465: 114-20, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25108231

RESUMEN

Lectin microarrays have emerged as a novel platform for glycan analysis during recent years. Here, we have combined surface plasmon resonance imaging (SPRi) with the lectin microarray for rapid and label-free profiling of stem cells. In this direction, 40 lectins from seven different glyco-binding motifs and three different cell lines-mouse embryonic stem cells (mESCs), mouse-induced pluripotent stem cells (miPSCs), and mouse embryonic fibroblast stem cells (MEFs)-were used. Pluripotent mouse stem cells were clearly distinguished from non-pluripotent stem cells. Eight lectins-DBA, MAL, PHA_E, PHA_L, EEL, AAL, PNA, and SNA-generated maximal value to define pluripotency of mouse stem cells in our experiments. The discriminant function based on lectin reactivities was highly accurate for the determination of stem cell pluripotency. These results suggested that glycomic analysis of stem cells leads to a novel comprehensive approach for quality control in cell-based therapy and regenerative medicine.


Asunto(s)
Lectinas/química , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Análisis por Matrices de Proteínas/métodos , Resonancia por Plasmón de Superficie/métodos , Animales , Línea Celular , Ratones
19.
J Mater Chem B ; 2(5): 471-476, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-32261527

RESUMEN

A material that can simultaneously support mammalian cell growth and preserve their physiological function is highly desirable in biomedical research. To meet this need, we fabricated superaligned carbon nanotube (SACNT) thin films and modified their surface using a convenient oxidization method. Our analysis demonstrated that the physical properties of oxidized SACNT films became more biocompatible. It supported the attachment and growth of primary mouse fibroblast cells as well as neonatal rat cardiomyocytes. Moreover, when cultured on oxidized SACNT films, neonatal rat cardiomyocytes spread normally and displayed calcium influx. Finally, we showed that, as oxidized SACNT films retained their electrical conductivity, attached cells can be electrotransfected in situ on them. Strong and prolonged expression of green fluorescence proteins (GFPs) or red fluorescence proteins (RFPs) was observed upon cell electroporation on oxidized SACNT films. In summary, our results provide evidence that simple oxidation greatly improved the biocompatibility of carbon nanotube films, which becomes more suitable for future applications in cell and genetic engineering.

20.
Methods Mol Biol ; 969: 221-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23296937

RESUMEN

Changing cell fate without altering its genome is very desirable in many experimental systems and for cell therapy. Compared to DNA plasmid or viral-based approach, mRNA has the advantage of high transfection efficiency, no danger of changing the genome or creating mutational insertions. Here, we describe a straightforward protocol to synthesize mRNA of genes of interest, and use them to induce pluripotency and direct cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Ingeniería Celular/métodos , Células Madre Pluripotentes Inducidas , ARN Mensajero , Transfección/métodos , Animales , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , ARN Mensajero/síntesis química , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...