Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(16): 3008-3024.e16, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35870449

RESUMEN

Here, we report inducible mosaic animal for perturbation (iMAP), a transgenic platform enabling in situ CRISPR targeting of at least 100 genes in parallel throughout the mouse body. iMAP combines Cre-loxP and CRISPR-Cas9 technologies and utilizes a germline-transmitted transgene carrying a large array of individually floxed, tandemly linked gRNA-coding units. Cre-mediated recombination triggers expression of all the gRNAs in the array but only one of them per cell, converting the mice to mosaic organisms suitable for phenotypic characterization and also for high-throughput derivation of conventional single-gene perturbation lines via breeding. Using gRNA representation as a readout, we mapped a miniature Perturb-Atlas cataloging the perturbations of 90 genes across 39 tissues, which yields rich insights into context-dependent gene functions and provides a glimpse of the potential of iMAP in genome decoding.


Asunto(s)
Sistemas CRISPR-Cas , ARN Guía de Kinetoplastida , Animales , Sistemas CRISPR-Cas/genética , Edición Génica , Genoma , Ratones , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Transgenes
2.
Nucleic Acids Res ; 50(19): e109, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-35929067

RESUMEN

Genomes can be edited by homologous recombination stimulated by CRISPR/Cas9 [clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated peptide 9]-induced DNA double-strand breaks. However, this approach is inefficient for inserting or deleting long fragments in mammalian cells. Here, we describe a simple genome-editing method, termed transcription-coupled Cas9-mediated editing (TEd), that can achieve higher efficiencies than canonical Cas9-mediated editing (CEd) in deleting genomic fragments, inserting/replacing large DNA fragments and introducing point mutations into mammalian cell lines. We also found that the transcription on DNA templates is crucial for the promotion of homology-directed repair, and that tethering transcripts from TEd donors to targeted sites further improves editing efficiency. The superior efficiency of TEd for the insertion and deletion of long DNA fragments expands the applications of CRISPR for editing mammalian genomes.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Animales , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Recombinación Homóloga/genética , Roturas del ADN de Doble Cadena , ADN/genética , Mamíferos/genética
3.
Reprod Biol Endocrinol ; 21(1): 40, 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37101140

RESUMEN

BACKGROUND: Studies have shown that sperm-borne microRNAs (miRNAs) are involved in mammalian preimplantation embryonic development. In humans, spermatozoan miR-34c levels are correlated with in vitro fertilization outcomes, such as embryo quality and the clinical pregnancy and live birth rates. In rabbits and cows, miR-34c improves the developmental competence of embryos generated by somatic cell nuclear transfer. However, the mechanisms underlying the regulation of embryonic development by miR-34c remain unknown. METHODS: Female C57BL/6 mice (6-8 weeks old) were superovulated, and pronucleated zygotes were collected and microinjected with an miR-34c inhibitor or a negative-control RNA. The embryonic development of the microinjected zygotes was evaluated, and the messenger RNA (mRNA) expression profiles of the embryos at the two-cell, four-cell and blastocyst stages (five embryos per group) were determined by RNA sequencing analysis. Gene expression levels were verified by reverse transcription-quantitative polymerase chain reaction. Cluster analysis and heat map visualization were performed to detect differentially expressed mRNAs. Pathway and process enrichment analyses were performed using ontology resources. Differentially expressed mRNAs were systematically analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins database to determine their biological functions. RESULTS: Embryonic developmental potential was significantly reduced in zygotes microinjected with the miR-34c inhibitor compared with those microinjected with a negative-control RNA. Two-cell stage embryos microinjected with an miR-34c inhibitor presented altered transcriptomic profiles, with upregulated expression of maternal miR-34c target mRNAs and classical maternal mRNAs. Differentially expressed transcripts were mainly of genes associated with lipid metabolism and cellular membrane function at the two-cell stage, with cell-cycle phase transition and energy metabolism at the four-cell stage; and with vesicle organization, lipid biosynthetic process and endomembrane system organization at the blastocyst stage. We also showed that genes related to preimplantation embryonic development, including Alkbh4, Sp1, Mapk14, Sin3a, Sdc1 and Laptm4b, were significantly downregulated after microinjection of an miR-34c inhibitor. CONCLUSIONS: Sperm-borne miR-34c may regulate preimplantation embryonic development by affecting multiple biological processes, such as maternal mRNA degradation, cellular metabolism, cell proliferation and blastocyst implantation. Our data demonstrate the importance of sperm-derived miRNAs in the development of preimplantation embryos.


Asunto(s)
MicroARNs , ARN Mensajero Almacenado , Humanos , Embarazo , Masculino , Animales , Femenino , Ratones , Bovinos , Conejos , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Ratones Endogámicos C57BL , Semen/metabolismo , Desarrollo Embrionario/genética , Espermatozoides/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Blastocisto , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estabilidad del ARN , Mamíferos , Proteínas de la Membrana/metabolismo , Proteínas Oncogénicas/metabolismo
4.
Mol Ther ; 30(9): 2923-2932, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35799444

RESUMEN

The prime editor is a versatile tool for targeted precise editing to generate point mutations, small insertions, or small deletions in eukaryotes. However, canonical PE3 system is less efficient, notably in primary cells or pluripotent stem cells. Here, we employed RNA polymerase II promoter instead of RNA polymerase III promoter, whose application is limited by specific DNA contexts, to produce Csy4-processed intronic prime editing guide RNAs (pegRNAs) and, together with other optimizations, achieved efficient targeting with poly(T)-containing pegRNAs, as well as combinatorial and conditional genetic editing. We also found simultaneous suppression of both DNA mismatch repair and DNA damage response could achieve efficient and accurate editing in human embryonic stem cells. These findings relieve the restrictions of RNA polymerase III (RNA-Pol-III)-based base editors and broadened the applications of prime editing.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , ARN Polimerasa II , Humanos , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , ARN Polimerasa III/genética , ARN Guía de Kinetoplastida/genética
5.
Mol Ther ; 30(9): 2933-2941, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35821638

RESUMEN

Adenine base editors (ABEs) are novel genome-editing tools, and their activity has been greatly enhanced by eight additional mutations, thus named ABE8e. However, elevated catalytic activity was concomitant with frequent generation of bystander mutations. This bystander effect precludes its safe applications required in human gene therapy. To develop next-generation ABEs that are both catalytically efficient and positionally precise, we performed combinatorial engineering of NG-ABE8e. We identify a novel variant (NG-ABE9e), which harbors nine mutations. NG-ABE9e exhibits robust and precise base-editing activity in human cells, with more than 7-fold bystander editing reduction at some sites, compared with NG-ABE8e. To demonstrate its practical utility, we used NG-ABE9e to correct the frequent T17M mutation in Rhodopsin for autosomal dominant retinitis pigmentosa. It reduces bystander editing by ∼4-fold while maintaining comparable efficiency. NG-ABE9e possesses substantially higher activity than NG-ABEmax and significantly lower bystander editing than NG-ABE8e in rice. Therefore, this study provides a versatile and improved adenine base editor for genome editing.


Asunto(s)
Adenina , Edición Génica , Sistemas CRISPR-Cas , Humanos , Mutación
6.
Circ Res ; 127(10): 1221-1232, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-32791884

RESUMEN

RATIONALE: Endothelial cells are thought to emerge de novo from the mesoderm to form the entire circulatory system. Recently, erythro-myeloid progenitors (EMPs) have been proposed to be another remarkable developmental origin for blood vessels in multiple organs, including the hindbrain, liver, lung, and heart, as demonstrated by lineage tracing studies using different genetic tools. These observations challenge the current consensus that intraembryonic vessels are thought to expand solely by the proliferation of preexisting endothelial cells. Resolution of this controversy over the developmental origin of endothelial cells is crucial for developing future therapeutics for vessel-dependent organ repair and regeneration. OBJECTIVE: To examine the contribution of EMPs to intraembryonic endothelial cells. METHODS AND RESULTS: We first used a transgenic mouse expressing a tamoxifen-inducible Mer-iCre fusion protein driven by the Csf1r (colony stimulating factor 1 receptor) promoter. Genetic lineage tracing based on Csf1r-Mer-iCre-Mer showed no contribution of EMPs to brain endothelial cells identified by several markers. We also generated a knock-in mouse line by inserting an internal ribosome entry site-iCre cassette into the 3' untranslated region of Csf1r gene to further investigate the cellular fates of EMPs. Similarly, we did not find any Csf1r-ires-iCre traced endothelial cells in brain, liver, lung, or heart in development either. Additionally, we found that Kit (KIT proto-oncogene receptor tyrosine kinase) was expressed not only in EMPs but also in embryonic hindbrain endothelial cells. Therefore, Kit promoter-driven recombinase, such as Kit-CreER, is a flawed tool for lineage tracing when examining the contribution of EMPs to hindbrain endothelial cells. We also traced CD45 (protein tyrosine phosphatase receptor type C; Ptprc)+ circulating EMPs and did not find any CD45 lineage-derived endothelial cells during development. CONCLUSIONS: Our study suggested that EMPs are not the origin of intraembryonic endothelial cells.


Asunto(s)
Linaje de la Célula , Células Endoteliales/citología , Células Precursoras Eritroides/citología , Animales , Endotelio Vascular/citología , Endotelio Vascular/embriología , Corazón Fetal/citología , Hígado/citología , Hígado/embriología , Pulmón/citología , Pulmón/embriología , Macrófagos/citología , Mesodermo/citología , Ratones , Rombencéfalo/citología , Rombencéfalo/embriología
7.
Genes Dev ; 28(5): 438-50, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24532687

RESUMEN

As bona fide p53 transcriptional targets, miR-34 microRNAs (miRNAs) exhibit frequent alterations in many human tumor types and elicit multiple p53 downstream effects upon overexpression. Unexpectedly, miR-34 deletion alone fails to impair multiple p53-mediated tumor suppressor effects in mice, possibly due to the considerable redundancy in the p53 pathway. Here, we demonstrate that miR-34a represses HDM4, a potent negative regulator of p53, creating a positive feedback loop acting on p53. In a Kras-induced mouse lung cancer model, miR-34a deficiency alone does not exhibit a strong oncogenic effect. However, miR-34a deficiency strongly promotes tumorigenesis when p53 is haploinsufficient, suggesting that the defective p53-miR-34 feedback loop can enhance oncogenesis in a specific context. The importance of the p53/miR-34/HDM4 feedback loop is further confirmed by an inverse correlation between miR-34 and full-length HDM4 in human lung adenocarcinomas. In addition, human lung adenocarcinomas generate an elevated level of a short HDM4 isoform through alternative polyadenylation. This short HDM4 isoform lacks miR-34-binding sites in the 3' untranslated region (UTR), thereby evading miR-34 regulation to disable the p53-miR-34 positive feedback. Taken together, our results elucidated the intricate cross-talk between p53 and miR-34 miRNAs and revealed an important tumor suppressor effect generated by this positive feedback loop.


Asunto(s)
Adenocarcinoma/fisiopatología , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/fisiopatología , MicroARNs/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma del Pulmón , Animales , Línea Celular Tumoral , Eliminación de Gen , Haploinsuficiencia , Humanos , Ratones , MicroARNs/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteínas ras/genética , Proteínas ras/metabolismo
8.
Transgenic Res ; 30(6): 821-835, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34542814

RESUMEN

Gene deletion by the Cre-Loxp system has facilitated functional studies of many critical genes in mice, offering important insights and allowing deeper understanding on the mechanisms underlying organ development and diseases, such as heart development and diseases. In this study, we generated a Myh6-Cre knockin mouse model by inserting the IRES-Cre-wpre-polyA cassette between the translational stop codon and the 3' untranslated region of the endogenous Myh6 gene. By crossing knockin mice with the Rosa26 reporter lines, we found that Myh6-Cre targeted cardiomyocytes at the embryonic and postnatal stages. In addition, we were able to inactivate the desmosome gene Desmoplakin (Dsp) by breeding Myh6-Cre mice with a conditional Dspflox knockout mouse line, which resulted in embryonic lethality during the mid-term pregnancy. These results suggest that the new Myh6-Cre mouse line can serve as a robust tool to dissect the distinct roles of genes involved in heart development and function.


Asunto(s)
Integrasas , Miocitos Cardíacos , Animales , Eliminación de Gen , Integrasas/genética , Ratones , Ratones Noqueados , Ratones Transgénicos
9.
J Biol Chem ; 294(22): 8894-8906, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31010826

RESUMEN

Early embryonic endocardium undergoes endothelial-to-mesenchymal transition to form cardiac cushion mesenchymal cells (MCs). Embryonic endocardium also gives rise to fibroblasts, intramyocardial adipocytes, and coronary mural cells, including smooth muscle cells and pericytes, in development. Whether endocardial cells directly differentiate into fibroblasts, coronary mural cells, and adipocytes or indirectly via an intermediate stage of endocardial-derived cushion MCs remains unknown. In addition to endocardium, epicardium and neural crest also contribute to cardiac cushion MCs. Given the developmental heterogeneity of cushion MCs and the lack of specific markers for endocardial-derived cushion MCs, conventional genetic lineage tracing utilizing Cre recombinase driven by one specific regulatory element is not sufficient to examine the fates of endocardial-derived cushion MCs. Intersectional genetic targeting approaches, which combine regulatory elements from two or more genes, have been employed to increase the specificity of cell targeting. Here, we developed a dual-recombinase intersectional targeting approach using Nfatc1-Dre, Sox9-CreER, and Cre/Dre double-dependent reporter Ai66 to specifically label endocardial-derived cushion MCs. Taking advantage of intersectional lineage tracing, we found that a subset of cardiac cells including fibroblasts, coronary mural cells, and intramyocardial adipocytes in adult hearts were derived from endocardial-derived cushion MCs. Our study suggests that embryonic endocardium contributes to cushion MCs first, and then endocardial-derived cushion MCs migrate into myocardium and differentiate into fibroblasts, coronary mural cells, and adipocytes in development. Understanding developmental origins of cardiac cell lineages will provide us more insights into cardiac development, regeneration, and diseases.


Asunto(s)
Adipocitos/citología , Linaje de la Célula , Endocardio/citología , Células Endoteliales/citología , Fibroblastos/citología , Células Madre Mesenquimatosas/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Miocardio/metabolismo , Miocardio/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo
10.
Gene Ther ; 27(7-8): 406, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32778810

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Gene Ther ; 27(7-8): 329-337, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32424232

RESUMEN

Gene therapy for auditory diseases is gradually maturing. Recent progress in gene therapy treatments for genetic and acquired hearing loss has demonstrated the feasibility in animal models. However, a number of hurdles, such as lack of safe viral vector with high efficiency and specificity, robust deafness large animal models, translating animal studies to clinic etc., still remain to be solved. It is necessary to overcome these challenges in order to effectively recover auditory function in human patients. Here, we review the progress made in our group, especially our efforts to make more effective and cell type-specific viral vectors for targeting cochlea cells.


Asunto(s)
Dependovirus , Terapia Genética , Pérdida Auditiva , Animales , Cóclea , Dependovirus/genética , Vectores Genéticos/genética , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Humanos
12.
Int J Mol Sci ; 21(11)2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32486079

RESUMEN

The authors wish to make the following correction to this paper [...].

13.
Int J Mol Sci ; 20(15)2019 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-31349654

RESUMEN

Early embryonic development in mammals, from fertilization to implantation, can be viewed as a process in which stem cells alternate between self-renewal and differentiation. During this process, the fates of stem cells in embryos are gradually specified, from the totipotent state, through the segregation of embryonic and extraembryonic lineages, to the molecular and cellular defined progenitors. Most of those stem cells with different potencies in vivo can be propagated in vitro and recapitulate their differentiation abilities. Complex and coordinated regulations, such as epigenetic reprogramming, maternal RNA clearance, transcriptional and translational landscape changes, as well as the signal transduction, are required for the proper development of early embryos. Accumulated studies suggest that Dicer-dependent noncoding RNAs, including microRNAs (miRNAs) and endogenous small-interfering RNAs (endo-siRNAs), are involved in those regulations and therefore modulate biological properties of stem cells in vitro and in vivo. Elucidating roles of these noncoding RNAs will give us a more comprehensive picture of mammalian embryonic development and enable us to modulate stem cell potencies. In this review, we will discuss roles of miRNAs in regulating the maintenance and cell fate potential of stem cells in/from mouse and human early embryos.


Asunto(s)
Diferenciación Celular/genética , Autorrenovación de las Células/genética , Regulación de la Expresión Génica , MicroARNs/genética , Células Madre/citología , Células Madre/metabolismo , Animales , Embrión de Mamíferos , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética
15.
Theranostics ; 14(6): 2379-2395, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646644

RESUMEN

Background: It is poorly understood what cellular types participate in ductular reaction (DR) and whether DR facilitates recovery from injury or accelerates hepatic fibrosis. The aim of this study is to gain insights into the role of hepatic progenitor cell (HPC)-originated DR during fibrotic progression. Methods: DR in liver specimens of PBC, chronic HBV infection (CHB) or NAFLD, and four rodent fibrotic models by different pathogenic processes was evaluated. Gli1 expression was inhibited in rodent models or cell culture and organoid models by AAV-shGli1 or treating with GANT61. Results: Severity of liver fibrosis was positively correlated with DR extent in patients with PBC, CHB or NAFLD. HPCs were activated, expanded, differentiated into reactive cholangiocytes and constituted "HPC-originated DR", accompanying with exacerbated fibrosis in rodent models of HPC activation & proliferation (CCl4/2-AAF-treated), Μdr2-/- spontaneous PSC, BDL-cholestatic fibrosis or WD-fed/CCl4-treated NASH-fibrosis. Gli1 expression was significantly increased in enriched pathways in vivo and in vitro. Enhanced Gli1 expression was identified in KRT19+-reactive cholangiocytes. Suppressing Gli1 expression by administration of AAV-shGli1 or GANT61 ameliorated HPC-originated DR and fibrotic extent. KRT19 expression was reduced after GANT61 treatment in sodium butyrate-stimulated WB-F344 cells or organoids or in cells transduced with Gli1 knockdown lentiviral vectors. In contrast, KRT19 expression was elevated after transducing Gli1 overexpression lentiviral vectors in these cells. Conclusions: During various modes of chronic injury, Gli1 acted as an important mediator of HPC activation, expansion, differentiation into reactive cholangiocytes that formed DR, and subsequently provoked hepatic fibrogenesis.


Asunto(s)
Proteínas Hedgehog , Cirrosis Hepática , Transducción de Señal , Células Madre , Proteína con Dedos de Zinc GLI1 , Animales , Femenino , Humanos , Masculino , Ratones , Ratas , Diferenciación Celular , Modelos Animales de Enfermedad , Proteínas Hedgehog/metabolismo , Hepatitis B Crónica/metabolismo , Hepatitis B Crónica/patología , Hepatitis B Crónica/complicaciones , Hígado/patología , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones Endogámicos C57BL , Piridinas/farmacología , Pirimidinas/farmacología , Células Madre/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína con Dedos de Zinc GLI1/genética
16.
Biomedicines ; 11(8)2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37626665

RESUMEN

Applying programmable nucleases in gene editing has greatly shaped current research in basic biology and clinical translation. Gene editing in human pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), is highly relevant to clinical cell therapy and thus should be examined with particular caution. First, since all mutations in PSCs will be carried to all their progenies, off-target edits of editors will be amplified. Second, due to the hypersensitivity of PSCs to DNA damage, double-strand breaks (DSBs) made by gene editing could lead to low editing efficiency and the enrichment of cell populations with defective genomic safeguards. In this regard, DSB-independent gene editing tools, such as base editors and prime editors, are favored due to their nature to avoid these consequences. With more understanding of the microbial world, new systems, such as Cas-related nucleases, transposons, and recombinases, are also expanding the toolbox for gene editing. In this review, we discuss current applications of programmable nucleases in PSCs for gene editing, the efforts researchers have made to optimize these systems, as well as new tools that can be potentially employed for differentiation modeling and therapeutic applications.

17.
FEBS J ; 290(7): 1705-1718, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35114069

RESUMEN

Triglyceride droplets can be stored within cardiac adipocytes (CAs) and cardiomyocytes in the heart. Cardiac adipocytes reside in three distinct regions: pericardial, epicardial, and intramyocardial adipose tissues. In healthy individuals, cardiac adipose tissues modulate cardiovascular functions and energy partitioning, which are, thus, protective. However, ectopic deposition of cardiac adipose tissues turns them into adverse lipotoxic, prothrombotic, and pro-inflammatory tissues with local and systemic contribution to the development of cardiovascular disorders. Accumulation of triglyceride droplets in cardiomyocytes may lead to lipotoxic injury of cardiomyocytes and contribute to the development of cardiac hypertrophy and dysfunction. Here, we summarize the roles of CAs and myocardial triglyceride droplets under physiological and pathological conditions and review the cellular sources of CAs in heart development and diseases. Understanding the functions and cellular origins of cardiac fat will provide clues for future studies on pathophysiological processes and treatment of cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Obesidad , Humanos , Obesidad/patología , Tejido Adiposo , Adipocitos/patología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/patología , Triglicéridos
18.
Stem Cell Res Ther ; 14(1): 161, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37316879

RESUMEN

The epicardium is the important outermost mesothelial/epithelial layer of the heart that serves as a signaling center for cardiac development and repair. During heart development, epicardial cells undergo a process known as epithelial-to-mesenchymal transition to form diverse mesenchymal cell lineages, such as fibroblasts, coronary vascular smooth muscle cells, and pericytes. However, it is not clear whether the reverse process, mesenchymal-to-epithelial transition (MET), takes place in the mammalian heart. In this study, we performed apical resection on neonatal hearts and used Fap-CreER;Ai9 labeling to track activated fibroblasts in the injured cardiac regions. We found that these fibroblasts underwent MET to generate epicardial cells during heart regeneration. To our knowledge, this is the first report of MET occurring in vivo during heart development and regeneration. Our findings suggest that it is feasible to directly convert fibroblasts into epicardial cells, providing a novel approach to generate epicardial cells.


Asunto(s)
Transición Epitelial-Mesenquimal , Corazón , Animales , Diferenciación Celular , Linaje de la Célula/genética , Fibroblastos , Mamíferos
19.
Sci China Life Sci ; 66(1): 51-66, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36322324

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is one of the most common inherited cardiomyopathies, characterized by progressive fibrofatty replacement in the myocardium. However, the cellular origin of cardiac adipocytes in ACM remains largely unknown. Unraveling the cellular source of cardiac adipocytes in ACM would elucidate the underlying pathological process and provide a potential target for therapy. Herein, we generated an ACM mouse model by inactivating desmosomal gene desmoplakin in cardiomyocytes; and examined the adipogenic fates of several cell types in the disease model. The results showed that SOX9+, PDGFRa+, and PDGFRb+ mesenchymal cells, but not cardiomyocytes or smooth muscle cells, contribute to the intramyocardial adipocytes in the ACM model. Mechanistically, Bmp4 was highly expressed in the ACM mouse heart and functionally promoted cardiac mesenchymal-to-adipose transition in vitro.


Asunto(s)
Cardiomiopatías , Corazón , Ratones , Animales , Miocardio/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis/fisiología , Obesidad/metabolismo
20.
Nat Commun ; 14(1): 5556, 2023 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-37689690

RESUMEN

Chemicals or drugs can accumulate within biomolecular condensates formed through phase separation in cells. Here, we use super-resolution imaging to search for chemicals that induce phase transition within chromatin at the microscale. This microscopic screening approach reveals that adriamycin (doxorubicin) - a widely used anticancer drug that is known to interact with chromatin - specifically induces visible local condensation and global conformational change of chromatin in cancer and primary cells. Hi-C and ATAC-seq experiments systematically and quantitatively demonstrate that adriamycin-induced chromatin condensation is accompanied by weakened chromatin interaction within topologically associated domains, compartment A/B switching, lower chromatin accessibility, and corresponding transcriptomic changes. Mechanistically, adriamycin complexes with histone H1 and induces phase transition of H1, forming fibrous aggregates in vitro. These results reveal a phase separation-driven mechanism for a chemotherapeutic drug.


Asunto(s)
Condensados Biomoleculares , Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Doxorrubicina/farmacología , Perfilación de la Expresión Génica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA