Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Aquat Toxicol ; 267: 106830, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38198987

RESUMEN

Environmental pollution is considered to lead to Ulva sp. green tides. Nevertheless, nutrients with high concentrations inhibit algae which may be damaged by antibiotics, such as OTC (oxytetracycline). Thus, Ulva sp. algae might pay a physiological cost under nutrient-OTC combined pressures. If this hypothesis is confirmed, Ulva sp. algae cannot easily form green tides, or green tides are difficult to maintain. To test this hypothesis, an uniform design experiment during which OTC, ammonia (NH4-N) and phosphate (PO4-P) were factors was set to simulate nutrient-OTC combined pressures, and Ulva lactuca was exposed to the pressures for 96 h. The TN (total nitrogen, CTN) or TP (total phosphorus, CTP) content in U. lactuca increased with increasing nutrient concentrations, as CTN = 21.206±1.000+ 1.227±0.418NH4-N × PO4-P (R2 = 0.282, p < 0.05) and CTP = 1.886±0.266+ 0.877±0.126PO4-P (R2 = 0.689, p < 0.05), respectively. The increase in dry weight of U. lactuca (Wdry) had a relationship with combined pressures, Wdry = 0.011±0.029 - 0.036±0.014PO4-P (R2 = 0.243, p < 0.05), i.e., the algal growth was inhibited by increasing PO4-P concentration. The SOD (Superoxide dismutase) activity (ASOD) was stimulated by OTC, as ASOD = 127.868±8.741+9.587±4.179 OTC (R2 = 0.193, p < 0.05). The contents of Chl a and b (Ca and Cb) were negatively affected by OTC or PO4-P with high concentration, as Ca = 0.566±0.042 - 0.024±0.022 OTC × PO4-P (R2 = 0.179, p < 0.05) and Cb = 0.512±0.043-0.044±0.020PO4-P (R2 = 0.180, p < 0.05). Thus, too high concentrations of PO4-P or OTC may hinder the formation and maintenance of Ulva sp. green tides.


Asunto(s)
Algas Comestibles , Oxitetraciclina , Ulva , Contaminantes Químicos del Agua , Contaminantes Químicos del Agua/toxicidad , Nutrientes , Superóxido Dismutasa
3.
Cell Mol Life Sci ; 80(6): 157, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37208522

RESUMEN

Virilizer-like m6A methyltransferase-associated protein (VIRMA) maintains the stability of the m6A writer complex. Although VIRMA is critical for RNA m6A deposition, the impact of aberrant VIRMA expression in human diseases remains unclear. We show that VIRMA is amplified and overexpressed in 15-20% of breast cancers. Of the two known VIRMA isoforms, the nuclear-enriched full-length but not the cytoplasmic-localised N-terminal VIRMA promotes m6A-dependent breast tumourigenesis in vitro and in vivo. Mechanistically, we reveal that VIRMA overexpression upregulates the m6A-modified long non-coding RNA, NEAT1, which contributes to breast cancer cell growth. We also show that VIRMA overexpression enriches m6A on transcripts that regulate the unfolded protein response (UPR) pathway but does not promote their translation to activate the UPR under optimal growth conditions. Under stressful conditions that are often present in tumour microenvironments, VIRMA-overexpressing cells display enhanced UPR and increased susceptibility to death. Our study identifies oncogenic VIRMA overexpression as a vulnerability that may be exploited for cancer therapy.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Respuesta de Proteína Desplegada/genética , ARN/metabolismo , Interferencia de ARN , Microambiente Tumoral
4.
Nat Commun ; 13(1): 2756, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589713

RESUMEN

Multiple pluripotent states have been described in mouse and human stem cells. Here, we apply single-cell RNA-seq to a newly established BMP4 induced mouse primed to naïve transition (BiPNT) system and show that the reset is not a direct reversal of cell fate but goes through a primordial germ cell-like cells (PGCLCs) state. We first show that epiblast stem cells bifurcate into c-Kit+ naïve and c-Kit- trophoblast-like cells, among which, the naïve branch undergoes further transition through a PGCLCs intermediate capable of spermatogenesis in vivo. Mechanistically, we show that DOT1L inhibition permits the transition from primed pluripotency to PGCLCs in part by facilitating the loss of H3K79me2 from Gata3/6. In addition, Prdm1/Blimp1 is required for PGCLCs and naïve cells, while Gata2 inhibits PGC-like state by promoting trophoblast-like fate. Our work not only reveals an alternative route for primed to naïve transition, but also gains insight into germ cell development.


Asunto(s)
Células Germinativas , Estratos Germinativos , Animales , Proteína Morfogenética Ósea 4 , Diferenciación Celular , Masculino , Ratones , Células Madre , Trofoblastos
5.
Blood Adv ; 5(17): 3241-3253, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34448811

RESUMEN

Epigenetic abnormalities are frequently involved in the initiation and progression of cancers, including acute myeloid leukemia (AML). A subtype of AML, acute promyelocytic leukemia (APL), is mainly driven by a specific oncogenic fusion event of promyelocytic leukemia-RA receptor fusion oncoprotein (PML-RARα). PML-RARα was reported as a transcription repressor through the interaction with nuclear receptor corepressor and histone deacetylase complexes leading to the mis-suppression of its target genes and differentiation blockage. Although previous studies were mainly focused on the connection of histone acetylation, it is still largely unknown whether alternative epigenetics mechanisms are involved in APL progression. KDM5A is a demethylase of histone H3 lysine 4 di- and tri-methylations (H3K4me2/3) and a transcription corepressor. Here, we found that the loss of KDM5A led to APL NB4 cell differentiation and retarded growth. Mechanistically, through epigenomics and transcriptomics analyses, KDM5A binding was detected in 1889 genes, with the majority of the binding events at promoter regions. KDM5A suppressed the expression of 621 genes, including 42 PML-RARα target genes, primarily by controlling the H3K4me2 in the promoters and 5' end intragenic regions. In addition, a recently reported pan-KDM5 inhibitor, CPI-455, on its own could phenocopy the differentiation effects as KDM5A loss in NB4 cells. CPI-455 treatment or KDM5A knockout could greatly sensitize NB4 cells to all-trans retinoic acid-induced differentiation. Our findings indicate that KDM5A contributed to the differentiation blockage in the APL cell line NB4, and inhibition of KDM5A could greatly potentiate NB4 differentiation.


Asunto(s)
Leucemia Promielocítica Aguda , Diferenciación Celular , Expresión Génica , Humanos , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína 2 de Unión a Retinoblastoma
6.
Nature ; 591(7849): 317-321, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33505026

RESUMEN

METTL3 (methyltransferase-like 3) mediates the N6-methyladenosine (m6A) methylation of mRNA, which affects the stability of mRNA and its translation into protein1. METTL3 also binds chromatin2-4, but the role of METTL3 and m6A methylation in chromatin is not fully understood. Here we show that METTL3 regulates mouse embryonic stem-cell heterochromatin, the integrity of which is critical for silencing retroviral elements and for mammalian development5. METTL3 predominantly localizes to the intracisternal A particle (IAP)-type family of endogenous retroviruses. Knockout of Mettl3 impairs the deposition of multiple heterochromatin marks onto METTL3-targeted IAPs, and upregulates IAP transcription, suggesting that METTL3 is important for the integrity of IAP heterochromatin. We provide further evidence that RNA transcripts derived from METTL3-bound IAPs are associated with chromatin and are m6A-methylated. These m6A-marked transcripts are bound by the m6A reader YTHDC1, which interacts with METTL3 and in turn promotes the association of METTL3 with chromatin. METTL3 also interacts physically with the histone 3 lysine 9 (H3K9) tri-methyltransferase SETDB1 and its cofactor TRIM28, and is important for their localization to IAPs. Our findings demonstrate that METTL3-catalysed m6A modification of RNA is important for the integrity of IAP heterochromatin in mouse embryonic stem cells, revealing a mechanism of heterochromatin regulation in mammals.


Asunto(s)
Ensamble y Desensamble de Cromatina , Heterocromatina/genética , Heterocromatina/metabolismo , Metiltransferasas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Animales , Retrovirus Endógenos/genética , Regulación de la Expresión Génica , Genes de Partícula A Intracisternal/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Histonas/metabolismo , Ratones , Proteína 28 que Contiene Motivos Tripartito/metabolismo
7.
Cell Rep ; 33(12): 108544, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33357433

RESUMEN

N6 methylation at adenosine 1832 (m6A1832) of mammalian 18S rRNA, occupying a critical position within the decoding center, is modified by a conserved methyltransferase, METTL5. Here, we find that METTL5 shows strong substrate preference toward the 18S A1832 motif but not the other reported m6A motifs. Comparison with a yeast ribosome structural model unmodified at this site indicates that the modification may facilitate mRNA binding by inducing conformation changes in the mammalian ribosomal decoding center. METTL5 promotes p70-S6K activation and proper translation initiation, and the loss of METTL5 significantly reduces the abundance of polysome. METTL5 expression is elevated in breast cancer patient samples and is required for growth of several breast cancer cell lines. We further find that Caenorhabditis elegans lacking the homolog metl-5 develop phenotypes known to be associated with impaired translation. Altogether, our findings uncover critical and conserved roles of METTL5 in the regulation of translation.


Asunto(s)
Neoplasias de la Mama/enzimología , Metiltransferasas/metabolismo , ARN Ribosómico 18S/metabolismo , Adenosina/metabolismo , Animales , Neoplasias de la Mama/patología , Caenorhabditis elegans , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Femenino , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Metilación
8.
Cell Rep ; 32(3): 107918, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32668215

RESUMEN

Coronavirus disease 2019 (COVID-19) has become a worldwide threat to humans, and neutralizing antibodies have therapeutic potential. We have purified more than 1,000 memory B cells specific to SARS-CoV-2 S1 or its RBD (receptor binding domain) and obtain 729 paired heavy- and light-chain fragments. Among these, 178 antibodies test positive for antigen binding, and the majority of the top 17 binders with EC50 below 1 nM are RBD binders. Furthermore, we identify 11 neutralizing antibodies, eight of which show IC50 within 10 nM, and the best one, 414-1, with IC50 of 1.75 nM. Through epitope mapping, we find three main epitopes in RBD recognized by these antibodies, and epitope-B antibody 553-15 could substantially enhance the neutralizing abilities of most of the other antibodies. We also find that 515-5 could cross neutralize the SARS-CoV pseudovirus. Altogether, our study provides 11 potent human neutralizing antibodies for COVID-19 as therapeutic candidates.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betacoronavirus/inmunología , Receptores Virales/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/inmunología , COVID-19 , Infecciones por Coronavirus/terapia , Mapeo Epitopo , Epítopos/inmunología , Humanos , Memoria Inmunológica/inmunología , Pruebas de Neutralización , Pandemias , Neumonía Viral/terapia , Dominios Proteicos/inmunología , SARS-CoV-2
9.
Cell Res ; 30(3): 256-268, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32047271

RESUMEN

Meiotic recombination is initiated by the formation of double-strand breaks (DSBs), which are repaired as either crossovers (COs) or noncrossovers (NCOs). In most mammals, PRDM9-mediated H3K4me3 controls the nonrandom distribution of DSBs; however, both the timing and mechanism of DSB fate control remain largely undetermined. Here, we generated comprehensive epigenomic profiles of synchronized mouse spermatogenic cells during meiotic prophase I, revealing spatiotemporal and functional relationships between epigenetic factors and meiotic recombination. We find that PRDM9-mediated H3K4me3 at DSB hotspots, coinciding with H3K27ac and H3K36me3, is intimately connected with the fate of the DSB. Our data suggest that the fate decision is likely made at the time of DSB formation: earlier formed DSBs occupy more open chromatins and are much more competent to proceed to a CO fate. Our work highlights an intrinsic connection between PRDM9-mediated H3K4me3 and the fate decision of DSBs, and provides new insight into the control of CO homeostasis.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Espermatogonias , Animales , Epigénesis Genética , Masculino , Profase Meiótica I , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espermatogonias/citología , Espermatogonias/metabolismo
10.
Protein Cell ; 11(3): 230, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31814083

RESUMEN

The author would like to add the below information in this correction. A similar study from Chao Lu group was published online on 5 September 2019 in Nature, entitled "The histone mark H3K36me2 recruits DNMT3A and shapes the intergenic DNA methylation landscape" (Weinberg et al., 2019). Although both the studies reported the preferential recognition of H3K36me2 by DNMT3A PWWP, ours in addition uncovered a stimulation function by such interaction on the activity of DNMT3A. On the disease connections, we used a NSD2 gain-of-function model which led to the discovery of potential therapeutic implication of DNA inhibitors in the related cancers, while the other study only used NSD1 and DNMT3A loss-of-function models.

12.
Genome Res ; 29(10): 1622-1634, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31537639

RESUMEN

How individual genes are regulated from a mitochondrial polycistronic transcript to have variable expression remains an enigma. Here, through bisulfite sequencing and strand-specific mapping, we show mitochondrial genomes in humans and other animals are strongly biased to light (L)-strand non-CpG methylation with conserved peak loci preferentially located at gene-gene boundaries, which was also independently validated by MeDIP and FspEI digestion. Such mtDNA methylation patterns are conserved across different species and developmental stages but display dynamic local or global changes during development and aging. Knockout of DNMT3A alone perturbed mtDNA regional methylation patterns, but not global levels, and altered mitochondrial gene expression, copy number, and oxygen respiration. Overexpression of DNMT3A strongly increased mtDNA methylation and strand bias. Overall, methylation at gene bodies and boundaries was negatively associated with mitochondrial transcript abundance and also polycistronic transcript processing. Furthermore, HPLC-MS confirmed the methylation signals on mitochondria DNA. Together, these data provide high-resolution mtDNA methylation maps that revealed a strand-specific non-CpG methylation, its dynamic regulation, and its impact on the polycistronic mitochondrial transcript processing.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , ADN Mitocondrial/genética , Epigenoma/genética , Animales , Islas de CpG/genética , ADN Metiltransferasa 3A , Regulación de la Expresión Génica/genética , Humanos , Mitocondrias/genética
13.
Leukemia ; 33(7): 1822-1827, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30755707

Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Regulación Neoplásica de la Expresión Génica , Leucemia/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/química , Proteína 1 Compañera de Translocación de RUNX1/química , Translocación Genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Leucemia/genética , Leucemia/metabolismo , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/genética , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Res ; 29(1): 90, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30559438

RESUMEN

We apologize for an error introduced during format conversion in the paper published online on 18 October 2018. The resolution of the MALDI-TOF result in Fig. 1e underwent an unexpected reduction when transformed from Powerpoint format. The corrected Fig. 1e is provided below on the left.

15.
Proc Natl Acad Sci U S A ; 116(3): 890-899, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30593567

RESUMEN

The AML1-ETO fusion protein, generated by the t(8;21) chromosomal translocation, is causally involved in nearly 20% of acute myeloid leukemia (AML) cases. In leukemic cells, AML1-ETO resides in and functions through a stable protein complex, AML1-ETO-containing transcription factor complex (AETFC), that contains multiple transcription (co)factors. Among these AETFC components, HEB and E2A, two members of the ubiquitously expressed E proteins, directly interact with AML1-ETO, confer new DNA-binding capacity to AETFC, and are essential for leukemogenesis. However, the third E protein, E2-2, is specifically silenced in AML1-ETO-expressing leukemic cells, suggesting E2-2 as a negative factor of leukemogenesis. Indeed, ectopic expression of E2-2 selectively inhibits the growth of AML1-ETO-expressing leukemic cells, and this inhibition requires the bHLH DNA-binding domain. RNA-seq and ChIP-seq analyses reveal that, despite some overlap, the three E proteins differentially regulate many target genes. In particular, studies show that E2-2 both redistributes AETFC to, and activates, some genes associated with dendritic cell differentiation and represses MYC target genes. In AML patients, the expression of E2-2 is relatively lower in the t(8;21) subtype, and an E2-2 target gene, THPO, is identified as a potential predictor of relapse. In a mouse model of human t(8;21) leukemia, E2-2 suppression accelerates leukemogenesis. Taken together, these results reveal that, in contrast to HEB and E2A, which facilitate AML1-ETO-mediated leukemogenesis, E2-2 compromises the function of AETFC and negatively regulates leukemogenesis. The three E proteins thus define a heterogeneity of AETFC, which improves our understanding of the precise mechanism of leukemogenesis and assists development of diagnostic/therapeutic strategies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/etiología , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/metabolismo , Recurrencia
17.
J Biol Chem ; 293(24): 9188-9197, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29716999

RESUMEN

Spermatogenesis is precisely controlled by complex gene expression programs and involves epigenetic reprogramming, including histone modification and DNA methylation. SET domain-containing 2 (SETD2) is the predominant histone methyltransferase catalyzing the trimethylation of histone H3 lysine 36 (H3K36me3) and plays key roles in embryonic stem cell differentiation and somatic cell development. However, its role in male germ cell development remains elusive. Here, we demonstrate an essential role of Setd2 for spermiogenesis, the final stage of spermatogenesis. Using RNA-seq, we found that, in postnatal mouse testes, Setd2 mRNA levels dramatically increase in 14-day-old mice. Using a germ cell-specific Setd2 knockout mouse model, we also found that targeted Setd2 knockout in germ cells causes aberrant spermiogenesis with acrosomal malformation before step 8 of the round-spermatid stage, resulting in complete infertility. Furthermore, we noted that the Setd2 deficiency results in complete loss of H3K36me3 and significantly decreases expression of thousands of genes, including those encoding acrosin-binding protein 1 (Acrbp1) and protamines, required for spermatogenesis. Our findings thus reveal a previously unappreciated role of the SETD2-dependent H3K36me3 modification in spermiogenesis and provide clues to the molecular mechanisms in epigenetic disorders underlying male infertility.


Asunto(s)
Proteínas Portadoras/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Protaminas/genética , Espermatogénesis , Acrosoma/metabolismo , Acrosoma/patología , Animales , Células Cultivadas , Código de Histonas , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Espermátides/citología , Espermátides/metabolismo , Espermátides/patología
18.
Cell ; 165(2): 331-42, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058665

RESUMEN

Regulation of enhancer activity is important for controlling gene expression programs. Here, we report that a biochemical complex containing a potential chromatin reader, RACK7, and the histone lysine 4 tri-methyl (H3K4me3)-specific demethylase KDM5C occupies many active enhancers, including almost all super-enhancers. Loss of RACK7 or KDM5C results in overactivation of enhancers, characterized by the deposition of H3K4me3 and H3K27Ac, together with increased transcription of eRNAs and nearby genes. Furthermore, loss of RACK7 or KDM5C leads to de-repression of S100A oncogenes and various cancer-related phenotypes. Our findings reveal a RACK7/KDM5C-regulated, dynamic interchange between histone H3K4me1 and H3K4me3 at active enhancers, representing an additional layer of regulation of enhancer activity. We propose that RACK7/KDM5C functions as an enhancer "brake" to ensure appropriate enhancer activity, which, when compromised, could contribute to tumorigenesis.


Asunto(s)
Carcinogénesis , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Histona Demetilasas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Técnicas de Inactivación de Genes , Xenoinjertos , Humanos , Ratones , Trasplante de Neoplasias , Receptores de Cinasa C Activada , Proteínas S100/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA