Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33500348

RESUMEN

ZFP57 is a master regulator of genomic imprinting. It has both maternal and zygotic functions that are partially redundant in maintaining DNA methylation at some imprinting control regions (ICRs). In this study, we found that DNA methylation was lost at most known ICRs in Zfp57 mutant embryos. Furthermore, loss of ZFP57 caused loss of parent-of-origin-dependent monoallelic expression of the target imprinted genes. The allelic expression switch occurred in the ZFP57 target imprinted genes upon loss of differential DNA methylation at the ICRs in Zfp57 mutant embryos. Specifically, upon loss of ZFP57, the alleles of the imprinted genes located on the same chromosome with the originally methylated ICR switched their expression to mimic their counterparts on the other chromosome with unmethylated ICR. Consistent with our previous study, ZFP57 could regulate the NOTCH signaling pathway in mouse embryos by impacting allelic expression of a few regulators in the NOTCH pathway. In addition, the imprinted Dlk1 gene that has been implicated in the NOTCH pathway was significantly down-regulated in Zfp57 mutant embryos. Our allelic expression switch models apply to the examined target imprinted genes controlled by either maternally or paternally methylated ICRs. Our results support the view that ZFP57 controls imprinted expression of its target imprinted genes primarily through maintaining differential DNA methylation at the ICRs.


Asunto(s)
Alelos , Impresión Genómica , Proteínas Represoras/genética , Animales , Metilación de ADN/genética , Embrión de Mamíferos/metabolismo , Femenino , Ratones , RNA-Seq , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/genética
2.
Hepatology ; 66(6): 2002-2015, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28653763

RESUMEN

Liver regeneration (LR) happens after various types of injuries. Unlike the well-studied LR caused by partial hepatectomy (PHx), there is accumulating evidence suggesting that LR during other injuries may result from unknown mechanisms. In this study, we found that insulin-like growth factor 2 (IGF-2) was drastically induced following the liver injuries caused by tyrosinemia or long-term treatments of CCl4 . However, this was not observed during the early phase of acute liver injuries after PHx or single treatment of CCl4 . Remarkably, most IGF-2-expressing hepatocytes were located at the histological area around the central vein of the liver lobule after the liver injuries caused either in fumarylacetoacetate hydrolase-deficient mice or in CCl4 chronically treated mice. Hepatocyte proliferation in vivo was significantly promoted by induced IGF-2 overexpression, which could be inhibited by adeno-associated virus-delivered IGF-2 short hairpin RNAs or linsitinib, an inhibitor of IGF-2 signaling. Proliferating hepatocytes in vivo responded to IGF-2 through both insulin receptor and IGF-1 receptor. IGF-2 also significantly promoted DNA synthesis of primary hepatocytes in vitro. More interestingly, the significantly induced IGF-2 was also found to colocalize with glutamine synthetase in the region enriched with proliferating hepatocytes for the liver samples from patients with liver fibrosis. CONCLUSION: IGF-2 is produced by pericentral hepatocytes to promote hepatocyte proliferation and repair tissue damage in the setting of chronic liver injury, which is distinct from the signaling that occurs post-PHx. (Hepatology 2017;66:2002-2015).


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/metabolismo , Regeneración Hepática , Animales , Intoxicación por Tetracloruro de Carbono , Proliferación Celular , Hepatectomía , Hepatocitos/metabolismo , Humanos , Hidrolasas/genética , Masculino , Ratones
3.
Proc Natl Acad Sci U S A ; 112(16): E2020-9, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25848000

RESUMEN

Zfp57 is a maternal-zygotic effect gene that maintains genomic imprinting. Here we report that Zfp57 mutants exhibited a variety of cardiac defects including atrial septal defect (ASD), ventricular septal defect (VSD), thin myocardium, and reduced trabeculation. Zfp57 maternal-zygotic mutant embryos displayed more severe phenotypes with higher penetrance than the zygotic ones. Cardiac progenitor cells exhibited proliferation and differentiation defects in Zfp57 mutants. ZFP57 is a master regulator of genomic imprinting, so the DNA methylation imprint was lost in embryonic heart without ZFP57. Interestingly, the presence of imprinted DLK1, a target of ZFP57, correlated with NOTCH1 activation in cardiac cells. These results suggest that ZFP57 may modulate NOTCH signaling during cardiac development. Indeed, loss of ZFP57 caused loss of NOTCH1 activation in embryonic heart with more severe loss observed in the maternal-zygotic mutant. Maternal and zygotic functions of Zfp57 appear to play redundant roles in NOTCH1 activation and cardiomyocyte differentiation. This serves as an example of a maternal effect that can influence mammalian organ development. It also links genomic imprinting to NOTCH signaling and particular developmental functions.


Asunto(s)
Corazón/embriología , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Cigoto/metabolismo , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio , Diferenciación Celular , Proliferación Celular , Regulación hacia Abajo , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/metabolismo , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Modelos Biológicos , Mutación , Miocitos Cardíacos/patología , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Células Madre/citología , Factores de Transcripción/metabolismo
4.
J Toxicol Environ Health A ; 79(9-10): 407-18, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27267823

RESUMEN

Esophageal squamous-cell carcinoma (ESCC) is one of the most common tumors worldwide. Recent studies suggested that long noncoding RNAs (lncRNAs) might play a key role in regulating cellular processes and cancer progression. One of the lncRNAs, urothelial carcinoma associated 1 (UCA1), is known to be dysregulated in several cancers, including bladder carcinoma, colorectal, melanoma, breast, gastric, and ESCC. However, contributions of UCA1 to ESCC remain largely undiscovered. In order to understand the role and mechanisms underlying UCA1 in ESCC, the association of UCA1 expression with risk of esophageal cancer development was determined in 106 esophageal cancer tissues of ESCC patients and adjacent normal tissues using real-time reverse-transcription polymerase chain reaction (PCR). The relative expression of UCA1 was significantly reduced in cancer versus adjacent normal tissues suggesting an enhanced risk of esophageal cancer. To investigate the biological functions of UCA1 in ESCC, it was of interest to examine whether overexpression of UCA1 might influence cell proliferation, apoptosis, cell cycle distribution, migration, and invasion in vitro using EC109 cells. Our results demonstrated that UCA1 decreased cell proliferation, migration, invasion, and cell cycle progression of EC109 cells. Further, mRNA microarray analysis of overexpressed UCA1 in EC109 cells revealed that abnormal expression of UCA1 also inhibited the Wnt signaling pathway. Gene levels of DKK1 were elevated while C-myc fell significantly in overexpressed UCA1 EC109 cells. Interestingly, Western blot demonstrated no significant differences in relative expression of CTNNB1 (ß-catenin) but marked reduction in ß-catenin (active form) levels in both total and nuclear proteins. These results suggest that UCA1 may inhibit ESCC growth by regulating the Wnt signaling pathway. In conclusion, UCA1 may be a novel biomarker involved in ESCC development that may provide a potential therapeutic target for ESCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Vía de Señalización Wnt , Adulto , Apoptosis , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago , Femenino , Humanos , Masculino , Persona de Mediana Edad , Análisis por Matrices de Proteínas , ARN Largo no Codificante/metabolismo
5.
Int J Mol Sci ; 16(11): 27781-95, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26610476

RESUMEN

miR-218, consisting of miR-218-1 at 4p15.31 and miR-218-2 at 5q35.1, was significantly decreased in esophageal squamous cell carcinoma (ESCC) in our previous study. The aim of this study was to determine whether aberrant methylation is associated with miR-218 repression. Bisulfite sequencing analysis (BSP), methylation specific PCR (MSP), and 5-aza-2'-deoxycytidine treatment assay were applied to determine the methyaltion status of miR-218 in cells and clinical samples. In vitro assays were performed to explore the role of miR-218. Results showed that miR-218-1 was significantly CpG hypermethylated in tumor tissues (81%, 34/42) compared with paired non-tumor tissues (33%, 14/42) (p < 0.05). However, no statistical difference was found in miR-218-2. Accordingly, expression of miR-218 was negatively correlated with miR-218-1 methylation status (p < 0.05). After demethylation treatment by 5-aza-2'-deoxycytidine, there was a 2.53- and 2.40-fold increase of miR-218 expression in EC109 and EC9706, respectively. miR-218 suppressed cell proliferation and arrested cells at G1 phase by targeting 3' untranslated region (3'UTR) of roundabout guidance receptor 1 (ROBO1). A negative correlation was found between miR-218 and ROBO1 mRNA expression in clinical samples. In conclusion, our results support that aberrant CpG hypermethylation at least partly accounts for miR-218 silencing in ESCC, which impairs its tumor-suppressive function.


Asunto(s)
Transformación Celular Neoplásica/genética , Represión Epigenética , Neoplasias Esofágicas/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Interferencia de ARN , Receptores Inmunológicos/genética , Regiones no Traducidas 3' , Azacitidina/farmacología , Secuencia de Bases , Sitios de Unión , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Islas de CpG , Metilación de ADN , Carcinoma de Células Escamosas de Esófago , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Orden Génico , Sitios Genéticos , Humanos , MicroARNs/química , ARN Mensajero/química , ARN Mensajero/genética , Proteínas Roundabout
6.
Front Cell Dev Biol ; 12: 1331074, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38357000

RESUMEN

Neural stem cells (NSCs) exhibit self-renewing and multipotential properties. Adult NSCs are located in two neurogenic regions of adult brain: the ventricular-subventricular zone (V-SVZ) of the lateral ventricle and the subgranular zone of the dentate gyrus in the hippocampus. Maintenance and differentiation of adult NSCs are regulated by both intrinsic and extrinsic signals that may be integrated through expression of some key factors in the adult NSCs. A number of transcription factors have been shown to play essential roles in transcriptional regulation of NSC cell fate transitions in the adult brain. Epigenetic regulators have also emerged as key players in regulation of NSCs, neural progenitor cells and their differentiated progeny via epigenetic modifications including DNA methylation, histone modifications, chromatin remodeling and RNA-mediated transcriptional regulation. This minireview is primarily focused on epigenetic regulations of adult NSCs during adult neurogenesis, in conjunction with transcriptional regulation in these processes.

7.
J Biol Chem ; 287(3): 2107-18, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22144682

RESUMEN

Previously, we discovered that ZFP57 is a maternal-zygotic effect gene, and it maintains DNA methylation genomic imprint at multiple imprinted regions in mouse embryos. Despite these findings, it remains elusive how DNA methyltransferases are targeted to the imprinting control regions to initiate and maintain DNA methylation imprint. To gain insights into these essential processes in genomic imprinting, we examined how ZFP57 maintains genomic DNA methylation imprint in mouse embryonic stem (ES) cells. Here we demonstrate that the loss of ZFP57 in mouse ES cells led to a complete loss of genomic DNA methylation imprint at multiple imprinted regions, similar to its role in mouse embryos. However, reintroduction of ZFP57 into Zfp57-null ES cells did not result in reacquisition of DNA methylation imprint, suggesting that the memory for genomic imprinting had been lost or altered in Zfp57-null ES cells in culture. Interestingly, ZFP57 and DNA methyltransferases could form complexes in the presence of KAP1/TRIM28/TIF1ß when co-expressed in COS cells. We also found that the wild-type exogenous ZFP57 but not the mutant ZFP57 lacking the KRAB box that interacts with its co-factor KAP1/TRIM28/TIF1ß could substitute for the endogenous ZFP57 in maintaining the DNA methylation imprint in ES cells. These results suggest that ZFP57 may recruit DNA methyltransferases to its target regions to maintain DNA methylation imprint, and this interaction is likely facilitated by KAP1/TRIM28/TIF1ß.


Asunto(s)
Metilación de ADN/fisiología , Metilasas de Modificación del ADN/metabolismo , Células Madre Embrionarias/metabolismo , Impresión Genómica/fisiología , Proteínas Represoras/metabolismo , Dedos de Zinc , Animales , Células COS , Chlorocebus aethiops , Metilasas de Modificación del ADN/genética , Células Madre Embrionarias/citología , Ratones , Ratones Mutantes , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Proteína 28 que Contiene Motivos Tripartito
8.
STAR Protoc ; 3(2): 101436, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35693210

RESUMEN

Gene functions can be assessed in mouse embryonic stem (ES) cells and in mutant mice derived from mutant ES cells. Here, we describe an approach for efficient isolation of the ES clones carrying deletion mutations at the target genes by CRISPR-Cas9. Two sgRNAs against a target gene are co-expressed with puromycin-resistant gene in ES cells through co-transfection followed by transient puromycin selection. Deletion mutations are identified by PCR from individual ES clones that are picked from puromycin-selected ES cells.


Asunto(s)
Sistemas CRISPR-Cas , Células Madre Embrionarias de Ratones , Animales , Sistemas CRISPR-Cas/genética , Células Madre Embrionarias , Ratones , Puromicina/farmacología , Transfección
9.
Front Cell Dev Biol ; 10: 784128, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35252168

RESUMEN

Zfp57 has both maternal and zygotic functions in mouse. It maintains genomic imprinting at most known imprinted regions and controls allelic expression of the target imprinted genes in mouse embryos. The DNA methylation imprint at many imprinting control regions (ICRs) is lost when both maternal and zygotic Zfp57 are absent in Zfp57 maternal-zygotic mutant mouse embryos. Interestingly, we found that DNA methylation at a few ICRs was partially lost without maternal Zfp57 in Zfp57 heterozygous mouse embryos derived from Zfp57 homozygous female mice. This suggests that maternal Zfp57 is essential for the maintenance of DNA methylation at a small subset of imprinted regions in mouse embryos. This maternal effect of Zfp57 was applied to allelic expression switch as well as expression levels of the corresponding imprinted genes. It is rather surprising that DNA methylation imprint was affected differently at Rasgrf1 and AK008011 imprinted regions in the female or male Zfp57 maternal-zygotic mutant embryos, with more significant loss of DNA methylation observed in the male mutant embryos. Loss of ZFP57 resulted in gender-specific differences in allelic expression switch and expression level changes of some imprinted genes in female or male mutant embryos. These results indicate maternal and sexually dimorphic effects of ZFP57 on genomic imprinting in mouse.

10.
iScience ; 25(9): 105003, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-36117996

RESUMEN

ZFP57 and ZFP445 maintain genomic imprinting in mouse embryos. We found DNA methylation was lost at most examined imprinting control regions (ICRs) in mouse Zfp57 mutant ES cells, which could not be prevented by the elimination of three TET proteins. To elucidate methylation maintenance mechanisms, we generated mutant ES clones lacking three major DNA methyltransferases (DNMTs). Intriguingly, DNMT3A and DNMT3B were essential for DNA methylation at a subset of ICRs in mouse ES cells although DNMT1 maintained DNA methylation at most known ICRs. These were similarly observed after extended culture. Germline-derived DNA methylation was lost at the examined ICRs lacking DNMTs according to allelic analysis. Similar to DNMT1, DNMT3A and DNMT3B were required for maintaining DNA methylation at repeats, genic regions, and other genomic sequences. Therefore, three DNA methyltransferases play complementary roles in maintaining DNA methylation in mouse ES cells including DNA methylation at the ICRs primarily mediated through the ZFP57-dependent pathway.

11.
Aging (Albany NY) ; 13(22): 24580-24604, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34793329

RESUMEN

Valvular heart disease (VHD) is a common heart disease that affects blood flow. It usually requires heart surgery. Valvular heart disease complicated with pulmonary artery hypertension (VHD-PAH) may be lethal due to heart failure that results from increased heart burden. It is important for these patients to seek early treatment in order to minimize the heart damage. However, there is no reliable diagnosis method in VHD. In this study, we found DNA methylation was increased at the promoter of BMPR2 gene in the VHD patients compared with the healthy controls. This finding was confirmed by an independent cohort study of VHD patients and healthy controls. In addition, BMPR2 mRNA levels were reduced in the plasma of the VHD patients. There is strong correlation between BMPR2 promoter DNA methylation and the severity of VHD. Indeed, we found that both BMPR2 promoter DNA methylation and BMPR2 mRNA levels in the plasma are good biomarkers of VHD by themselves, with the respective AUC value of 0.879 and 0.725, respectively. When they were used in combination, the diagnostic value was even better, with the AUC value of 0.93. Consistent with the results in the VHD patients, we observed decreased BMPR2 and increased fibrosis in the lung of a PAH model mouse. BMPR2 was also decreased in the hearts of the PAH mice, whereas BMP4 was increased. Furthermore, BMPR2 was reduced in the heart valve tissue samples of human VHD patients after valve replacement with moderate/severe PAH compared with those with mild PAH. There was also increased apoptosis in the hearts of the PAH mice. BMPR2 promoter DNA methylation and its expression appear to be good biomarkers for VHD. Our results also suggest that DNA methylation may cause PAH through deregulation of BMP signaling and increased apoptosis.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Metilación de ADN/genética , Enfermedades de las Válvulas Cardíacas , Regiones Promotoras Genéticas/genética , Hipertensión Arterial Pulmonar , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
12.
Mol Hum Reprod ; 16(9): 695-703, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20382782

RESUMEN

Maternal effect refers to the genetic phenomenon in which a phenotype in the progeny is caused by a genetic mutation in the maternal genome rather than a mutation of its own. Maternal effect genes are usually involved in the maternal-to-zygotic transition during embryonic development before zygotic genes are turned on. Although it is widely observed in invertebrate organisms, it is not common in vertebrate animals, especially in mammals. Genomic imprinting is an epigenetic phenomenon that is unique to eutherian mammals, marsupials and plants. One characteristic of genomic imprinting is parental origin-specific expression of imprinted genes. The molecular mechanisms underlying genomic imprinting are poorly understood. Mouse Zfp57 is the first example of a mammalian maternal-zygotic effect gene and it exhibits maternal-zygotic embryonic lethality around midgestation when both maternal and zygotic functions of Zfp57 are absent. Loss of Zfp57 also results in loss of differential DNA methylation at multiple imprinted regions. Interestingly, the midgestational embryonic lethality due to loss of both maternal and zygotic functions of Zfp57 occurs much later than the typically observed maternal-zygotic embryonic lethality during the maternal-to-zygotic transition period in early preimplantation embryos. I hypothesize that gradual loss of heritable genomic DNA methylation imprints over many cell divisions could account for this spatial and temporal discrepancy between the causative molecular defect and the observed phenotype in the Zfp57 mutant.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Cigoto/fisiología , Animales , Pérdida del Embrión/genética , Desarrollo Embrionario/genética , Femenino , Genotipo , Humanos , Ratones , Mutación , Fenotipo , Proteínas Represoras/genética
13.
Stem Cell Res ; 50: 102116, 2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-33352531

RESUMEN

COVID-19 has spread to all countries around the world after it was first discovered in Wuhan of China at the end of 2019. It is caused by a novel coronavirus called SARS-CoV-2 with much semblance to SARS-CoV including the sequence homology and disease symptoms. It is reported to be more infectious than SARS-CoV due to higher binding affinities between its spike protein and the ACE2 receptor on cell surface. Despite this, its case fatality rate is much lower compared with that of SARS-CoV although it varies in different countries. However, the case fatality rate increases steadily with age and it is reported to be the highest in aged COVID-19 male patients in almost all countries. Consistent with these, females have higher antiviral immune responses. Males and females are different in inflammatory response and aberrantly hyperactive cytokines are the main lethal causes of COVID-19. Interestingly, the gene encoding the ACE2 receptor protein and some genes encoding the immune regulatory proteins such as TLR7 are located on X chromosome which is subject to X chromosome inactivation and sex hormone regulation. These may account for some sex-dependent immune responses and lethality observed in COVID-19 patients. In general, children are less likely to be infected with SARS-CoV-2 and only less than 1% of pediatric COVID-19 patients may die of COVID-19. However, the most severe pediatric cases become multisystem inflammatory syndrome that is similar to Kawasiki disease with features of viral infection. Since most infected kids were boys in China, there may be sex-dependent immune response in pediatric COVID-19 cases as well.

14.
Stem Cell Res ; 41: 101655, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31734645

RESUMEN

Pluripotent stem cells are considered to be the ideal candidates for cell-based therapies in humans. In this regard, both nuclear transfer embryonic stem (ntES) cells and induced pluripotent stem (iPS) cells are particularly advantageous because patient-specific autologous ntES and iPS cells can avoid immunorejection and other side effects that may be present in the allogenic pluripotent stem cells derived from unrelated sources. However, they have been found to contain deleterious genetic and epigenetic changes that may hinder their therapeutic applications. Indeed, deregulation of genomic imprinting has been frequently observed in reprogrammed ntES and iPS cells. We will survey the recent studies on genomic imprinting in pluripotent stem cells, particularly in iPS cells. In a previous study published about six years ago, genomic imprinting was found to be variably lost in mouse iPS clones. Intriguingly, de novo DNA methylation also occurred at the previously unmethylated imprinting control regions (ICRs) in a high percentage of iPS clones. These unexpected results were confirmed by a recent independent study with a similar approach. Since dysregulation of genomic imprinting can cause many human diseases including cancer and neurological disorders, these recent findings on genomic imprinting in reprogramming may have some implications for therapeutic applications of pluripotent stem cells.


Asunto(s)
Técnicas de Reprogramación Celular , Reprogramación Celular , Impresión Genómica , Células Madre Embrionarias Humanas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Técnicas de Transferencia Nuclear , Animales , Metilación de ADN , Células Madre Embrionarias Humanas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología
15.
Cell Rep ; 26(2): 302-312.e4, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30625312

RESUMEN

The coactivator-associated arginine methyltransferase CARM1 catalyzes the methylation of histone H3 arginine 17/26 (H3R17/26me) and non-histone proteins at arginine residues to regulate gene transactivation through profiling or Carm1 overexpression assays. However, the direct relationship between H3R17/26me and its causal role in mouse embryo development remains largely unclear. Here, we use rAPOBEC1-XTEN-Cas9n-UGI (BE3) to efficiently introduce a point mutation (R17H) at multiple Hist1/2H3 loci and a premature-stop codon into the catalytic domain of CARM1 in mouse embryos, resulting in remarkable downregulation of H3R17me levels and developmental defects in pre-implantation and fetal embryos. Transcriptomic analysis reveals that Yap1 and cell cycle signaling pathways are dysregulated in Carm1 truncation and H3R17H substitution embryos, and Yap1 overexpression could rescue the base-editing-elicited defects. Our data establish the direct regulatory relationship between CARM1-mediated H3R17me and early mouse embryo development and demonstrate that Yap1 acts downstream of CARM1-mediated H3R17me to regulate the mouse embryo development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Transducción de Señal , Animales , Dominio Catalítico , Ciclo Celular , Línea Celular Tumoral , Código de Histonas , Histonas/química , Histonas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Mutación Missense , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Transcriptoma , Proteínas Señalizadoras YAP
16.
iScience ; 15: 640-648, 2019 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-31130518

RESUMEN

RNA splicing is related to many human diseases; however, lack of efficient genetic approaches to modulate splicing has prevented us from dissecting their functions in human diseases. Recently developed base editors (BEs) offer a new strategy to modulate RNA splicing by converting conservative splice sites, but it is limited by the editing precision and scope. To overcome the limitations of currently available BE-based tools, we combined SpCas9-NG with ABEmax to generate a new BE, ABEmax-NG. We demonstrated that ABEmax-NG performed precise A⋅T to G⋅C conversion with an expanded scope, thus covering many more splicing sites. Taking advantage of this tool, we precisely achieved A⋅T to G⋅C conversion exactly at the splice sites. We further modeled pathogenic RNA splicing in vitro and in vivo. Taken together, we successfully generated a versatile tool suitable for precise and broad editing at the splice sites.

17.
BMC Cell Biol ; 8: 37, 2007 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-17725840

RESUMEN

BACKGROUND: The mechanism involved in the maintenance and differentiation of embryonic stem (ES) cells is incompletely understood. RESULTS: To address this issue, we have developed a retroviral gene trap vector that can target genes expressed in undifferentiated ES cells. This gene trap vector harbors both GFP and Neo reporter genes. G-418 drug resistance was used to select ES clones in which the vector was integrated into transcriptionally active loci. This was then followed by GFP FACS profiling to identify ES clones with reduced GFP fluorescence and, hence, reduced transcriptional activity when ES cells differentiate. Reduced expression of the GFP reporter in six of three hundred ES clones in our pilot screening was confirmed to be down-regulated by Northern blot analysis during ES cell differentiation. These six ES clones represent four different genes. Among the six integration sites, one was at Zfp-57 whose gene product is known to be enriched in undifferentiated ES cells. Three were located in an intron of a novel isoform of CSL/RBP-J kappa which encodes the key transcription factor of the LIN-12/Notch pathway. Another was inside a gene that may encode noncoding RNA transcripts. The last integration event occurred at a locus that may harbor a novel gene. CONCLUSION: Taken together, we demonstrate the use of a novel retroviral gene trap vector in identifying genes preferentially expressed in undifferentiated ES cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Expresión Génica , Vectores Genéticos , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones
18.
Stem Cell Res ; 16(2): 282-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27345984

RESUMEN

Zfp57 is a master regulator of genomic imprinting in mouse embryos. To further test its functions, we have derived multiple Zfp57 mutant ES clones directly from mouse blastocysts. Indeed, we found DNA methylation imprint was lost at most examined imprinting control regions in these Zfp57 mutant ES clones, similar to what was observed in Zfp57 mutant embryos in the previous studies. This result indicates that these blastocyst-derived Zfp57 mutant ES clones can be employed for functional analyses of Zfp57 in genomic imprinting.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Embrionarias de Ratones/citología , Animales , Blastocisto/citología , Línea Celular , Cromosomas/genética , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Femenino , Impresión Genómica , Genotipo , Heterocigoto , Masculino , Metafase , Ratones , Microscopía Fluorescente , Células Madre Embrionarias de Ratones/metabolismo , Mutagénesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo
19.
Stem Cell Res ; 55: 102468, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298434

Asunto(s)
COVID-19 , Furina , Humanos , SARS-CoV-2
20.
Stem Cell Res ; 16(2): 252-5, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27280270

RESUMEN

Parental origin-dependent expression of the imprinted genes is essential for mammalian development. Zfp57 maintains genomic imprinting in mouse embryos and ES cells. To examine the allelic expression patterns of the imprinted genes in ES cells, we obtained multiple hybrid ES clones that were directly derived from the blastocysts generated from the cross between mice on two different genetic backgrounds. The blastocyst-derived ES clones displayed largely intact DNA methylation imprint at the tested imprinted regions. These hybrid ES clones will be useful for future studies to examine the allelic expression of the imprinted genes in ES cells and their differentiated progeny.


Asunto(s)
Células Madre Embrionarias/citología , Animales , Blastocisto/citología , Línea Celular , Metilación de ADN , Cuerpos Embrioides/citología , Células Madre Embrionarias/metabolismo , Genotipo , Heterocigoto , Ratones , Ratones Endogámicos DBA , Microscopía Fluorescente , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA