Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Cell ; 187(10): 2428-2445.e20, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38579712

RESUMEN

Alveolar type 2 (AT2) cells are stem cells of the alveolar epithelia. Previous genetic lineage tracing studies reported multiple cellular origins for AT2 cells after injury. However, conventional lineage tracing based on Cre-loxP has the limitation of non-specific labeling. Here, we introduced a dual recombinase-mediated intersectional genetic lineage tracing approach, enabling precise investigation of AT2 cellular origins during lung homeostasis, injury, and repair. We found AT1 cells, being terminally differentiated, did not contribute to AT2 cells after lung injury and repair. Distinctive yet simultaneous labeling of club cells, bronchioalveolar stem cells (BASCs), and existing AT2 cells revealed the exact contribution of each to AT2 cells post-injury. Mechanistically, Notch signaling inhibition promotes BASCs but impairs club cells' ability to generate AT2 cells during lung repair. This intersectional genetic lineage tracing strategy with enhanced precision allowed us to elucidate the physiological role of various epithelial cell types in alveolar regeneration following injury.


Asunto(s)
Células Epiteliales Alveolares , Pulmón , Células Madre , Animales , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/citología , Diferenciación Celular , Linaje de la Célula , Pulmón/citología , Pulmón/metabolismo , Pulmón/fisiología , Lesión Pulmonar/patología , Ratones Endogámicos C57BL , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Receptores Notch/metabolismo , Regeneración , Transducción de Señal , Células Madre/metabolismo , Células Madre/citología
2.
EMBO J ; 39(4): e102675, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31943281

RESUMEN

Site-specific recombinase-mediated genetic technology, such as inducible Cre-loxP recombination (CreER), is widely used for in vivo genetic manipulation with temporal control. The Cre-loxP technology improves our understanding on the in vivo function of specific genes in organ development, tissue regeneration, and disease progression. However, inducible CreER often remains inefficient in gene deletion. In order to improve the efficiency of gene manipulation, we generated a self-cleaved inducible CreER (sCreER) that switches inducible CreER into a constitutively active Cre by itself. We generated endocardial driver Npr3-sCreER and fibroblast driver Col1a2-sCreER, and compared them with conventional Npr3-CreER and Col1a2-CreER, respectively. For easy-to-recombine alleles such as R26-tdTomato, there was no significant difference in recombination efficiency between sCreER and the conventional CreER. However, for alleles that were relatively inert for recombination such as R26-Confetti, R26-LZLT, R26-GFP, or VEGFR2flox/flox alleles, sCreER showed a significantly higher efficiency in recombination compared with conventional CreER in endocardial cells or fibroblasts. Compared with conventional CreER, sCreER significantly enhances the efficiency of recombination to induce gene expression or gene deletion, allowing temporal yet effective in vivo genomic modification for studying gene function in specific cell lineages.


Asunto(s)
Integrasas/genética , Recombinación Genética , Alelos , Animales , Linaje de la Célula , Femenino , Fibroblastos , Eliminación de Gen , Expresión Génica , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
3.
J Biol Chem ; 298(6): 101965, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35461809

RESUMEN

Genetic technology using site-specific recombinases, such as the Cre-loxP system, has been widely employed for labeling specific cell populations and for studying their functions in vivo. To enhance the precision of cell lineage tracing and functional study, a similar site-specific recombinase system termed Dre-rox has been recently used in combination with Cre-loxP. To enable more specific cell lineage tracing and ablation through dual recombinase activity, we generated two mouse lines that render Dre- or Dre+Cre-mediated recombination to excise a stop codon sequence that prevents the expression of diphtheria toxin receptor (DTR) knocked into the ubiquitously expressed and safe Rosa26 locus. Using different Dre- and Cre-expressing mouse lines, we showed that the surrogate gene reporters tdTomato and DTR were simultaneously expressed in target cells and in their descendants, and we observed efficient ablation of tdTomato+ cells after diphtheria toxin administration. These mouse lines were used to simultaneously trace and deplete the target cells of interest through the inducible expression of a reporter and DTR using dual Cre and Dre recombinases, allowing a more precise and efficient study of the role of specific cell subsets within a heterogeneous population in pathophysiological conditions in vivo.


Asunto(s)
Linaje de la Célula , Integrasas , Recombinasas , Animales , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Recombinasas/genética , Recombinasas/metabolismo , Recombinación Genética
4.
Development ; 147(4)2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31988189

RESUMEN

Cellular proliferation is a basic process during organ development, tissue homeostasis and disease progression. Likewise, after injury typically multiple cell lineages respond to various cues and proliferate to initiate repair and/or remodeling of the injured tissue. Unravelling the specific role of proliferation of one cell type and its lineage in the context of the whole organism during tissue regeneration and/or disease progression would provide valuable information on these processes. Here, we report a new genetic system that allows cell proliferation to be inhibited in a tissue-specific manner. We generated Cre- or Dre-inducible p21-GFP (ip21-GFP) transgenic mice that enable experimentally induced permanent cell cycle arrest of specific cell lineages of interest, while genetically marking these cells. This system allows for the inhibition of pathogenic cell proliferation. We found that cardiac fibroblast proliferation inhibition significantly reduced scar formation, and promoted neovascularization and cardiomyocyte survival. Additionally, we found that inhibition of one type of cell proliferation (namely, hepatocytes) induces the lineage conversion of another type cells (i.e. ductal cells) during tissue regeneration. These results validate the use of ip21-GFP mice as a new genetic tool for cell lineage-specific inhibition of cell proliferation in vivo.


Asunto(s)
Proliferación Celular , Regulación de la Expresión Génica , Técnicas Genéticas , Alelos , Animales , Linaje de la Célula , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Femenino , Fibroblastos/fisiología , Proteínas Fluorescentes Verdes , Corazón/crecimiento & desarrollo , Corazón/fisiología , Hepatocitos/citología , Hepatocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/patología , Miocitos Cardíacos/citología
5.
Sensors (Basel) ; 23(13)2023 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-37447659

RESUMEN

Erasure-code-based storage systems suffer from problems such as long repair time and low I/O performance, resulting in high repair costs. For many years, researchers have focused on reducing the cost of repairing erasure-code-based storage systems. In this study, we discuss the demerits of node selecting, data transferring and data repair in erasure-code-based storage systems. Based on the network topology and node structure, we propose SDNC-Repair, a cooperative data repair strategy based on erasure code for SDS (Software Defined Storage), and describe its framework. Then, we propose a data source selection algorithm that senses the available network bandwidth between nodes and a data flow scheduling algorithm in SDNC-Repair. Additionally, we propose a data repair method based on node collaboration and data aggregation. Experiments illustrate that the proposed method has better repair performance under different data granularities. Compared to the conventional repair method, although the SDNC-Repair is more constrained by the cross-rack bandwidth, it improves system throughput effectively and significantly reduces data repair time in scenarios where multiple nodes fail and bandwidth is limited.


Asunto(s)
Algoritmos , Programas Informáticos
6.
J Biol Chem ; 295(3): 690-700, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31771978

RESUMEN

Genetic lineage tracing is widely used to study organ development and tissue regeneration. Multicolor reporters are a powerful platform for simultaneously tracking discrete cell populations. Here, combining Dre-rox and Cre-loxP systems, we generated a new dual-recombinase reporter system, called Rosa26 traffic light reporter (R26-TLR), to monitor red, green, and yellow fluorescence. Using this new reporter system with the three distinct fluorescent reporters combined on one allele, we found that the readouts of the two recombinases Cre and Dre simultaneously reflect Cre+Dre-, Cre-Dre+, and Cre+Dre+ cell lineages. As proof of principle, we show specific labeling in three distinct progenitor/stem cell populations, including club cells, AT2 cells, and bronchoalveolar stem cells, in Sftpc-DreER;Scgb1a1-CreER;R26-TLR mice. By using this new dual-recombinase reporter system, we simultaneously traced the cell fate of these three distinct cell populations during lung repair and regeneration, providing a more comprehensive picture of stem cell function in distal airway repair and regeneration. We propose that this new reporter system will advance developmental and regenerative research by facilitating a more sophisticated genetic approach to studying in vivo cell fate plasticity.


Asunto(s)
Linaje de la Célula/genética , Integrasas/genética , Recombinasas/genética , Células Madre/citología , Alelos , Animales , Diferenciación Celular/genética , Fluorescencia , Marcación de Gen , Genes Reporteros/genética , Integrasas/química , Ratones , Ratones Transgénicos/genética , Células Madre/química
7.
Circulation ; 141(1): 67-79, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31779484

RESUMEN

BACKGROUND: Mutations in low-density lipoprotein (LDL) receptor (LDLR) are one of the main causes of familial hypercholesterolemia, which induces atherosclerosis and has a high lifetime risk of cardiovascular disease. The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system is an effective tool for gene editing to correct gene mutations and thus to ameliorate disease. METHODS: The goal of this work was to determine whether in vivo somatic cell gene editing through the CRISPR/Cas9 system delivered by adeno-associated virus (AAV) could treat familial hypercholesterolemia caused by the Ldlr mutant in a mouse model. We generated a nonsense point mutation mouse line, LdlrE208X, based on a relevant familial hypercholesterolemia-related gene mutation. The AAV-CRISPR/Cas9 was designed to correct the point mutation in the Ldlr gene in hepatocytes and was delivered subcutaneously into LdlrE208X mice. RESULTS: We found that homogeneous LdlrE208X mice (n=6) exhibited severe atherosclerotic phenotypes after a high-fat diet regimen and that the Ldlr mutation was corrected in a subset of hepatocytes after AAV-CRISPR/Cas9 treatment, with LDLR protein expression partially restored (n=6). Compared with the control groups (n=6 each group), the AAV-CRISPR/Cas9 with targeted single guide RNA group (n=6) had significant reductions in total cholesterol, total triglycerides, and LDL cholesterol in the serum, whereas the aorta had smaller atherosclerotic plaques and a lower degree of macrophage infiltration. CONCLUSIONS: Our work shows that in vivo AAV-CRISPR/Cas9-mediated Ldlr gene correction can partially rescue LDLR expression and effectively ameliorate atherosclerosis phenotypes in Ldlr mutants, providing a potential therapeutic approach for the treatment of patients with familial hypercholesterolemia.


Asunto(s)
Aterosclerosis , Sistemas CRISPR-Cas , Dependovirus , Edición Génica , Hiperlipoproteinemia Tipo II , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/terapia , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/metabolismo , Hiperlipoproteinemia Tipo II/patología , Hiperlipoproteinemia Tipo II/terapia , Ratones , Ratones Transgénicos , Mutación Missense , Receptores de LDL/genética , Receptores de LDL/metabolismo
8.
Circ Res ; 125(3): 343-355, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31185811

RESUMEN

RATIONALE: The developing heart is composed of cardiomyocytes and noncardiomyocytes since the early stage. It is generally believed that noncardiomyocytes including the cardiac progenitors contribute to new cardiomyocytes of the looping heart. However, it remains unclear what the cellular dynamics of nonmyocyte to cardiomyocyte conversion are and when the lineage segregation occurs during development. It also remains unknown whether nonmyocyte to cardiomyocyte conversion contributes to neonatal heart regeneration. OBJECTIVE: We quantify the lineage conversion of noncardiomyocytes to cardiomyocytes in the embryonic and neonatal hearts and determine when the 2 cell lineages segregate during heart development. Moreover, we directly test if nonmyocyte to cardiomyocyte conversion contributes to neonatal heart regeneration. METHODS AND RESULTS: We generated a dual genetic lineage tracing strategy in which cardiomyocytes and noncardiomyocytes of the developing heart could be simultaneously labeled by 2 orthogonal recombination systems. Genetic fate mapping showed that nonmyocyte to cardiomyocyte conversion peaks at E8.0 (embryonic day) to E8.5 and gradually declines at E9.5 and E10.5. Noncardiomyocytes do not generate any cardiomyocyte at and beyond E11.5 to E12.5. In the neonatal heart, noncardiomyocytes also do not contribute to any new cardiomyocyte in homeostasis or after injury. CONCLUSIONS: Noncardiomyocytes contribute to new cardiomyocytes of the developing heart at early embryonic stage before E11.5. The noncardiomyocyte and cardiomyocyte lineage segregation occurs between E10.5 and E11.5, which is maintained afterward even during neonatal heart regeneration.


Asunto(s)
Linaje de la Célula , Corazón Fetal/citología , Genes Reporteros , Miocitos Cardíacos/citología , Animales , Animales Recién Nacidos , Rastreo Celular , Regulación del Desarrollo de la Expresión Génica , Marcadores Genéticos , Edad Gestacional , Corazón/embriología , Corazón/fisiología , Ratones , Ratones Transgénicos , Regeneración , Células Madre/clasificación , Células Madre/citología
9.
Circ Res ; 122(7): 984-993, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29374073

RESUMEN

RATIONALE: Endocardium is the major source of coronary endothelial cells (ECs) in the fetal and neonatal hearts. It remains unclear whether endocardium in the adult stage is also the main origin of neovascularization after cardiac injury. OBJECTIVE: To define the vascular potential of adult endocardium in homeostasis and after cardiac injuries by fate-mapping studies. METHODS AND RESULTS: We generate an inducible adult endocardial Cre line (Npr3 [natriuretic peptide receptor C]-CreER) and show that Npr3-CreER efficiently and specifically labels endocardial cells but not coronary blood vessels in the adult heart. The adult endocardial cells do not contribute to any vascular ECs during cardiac homeostasis. To examine the formation of blood vessels from endocardium after injury, we generate 4 cardiac injury models with Npr3-CreER mice: myocardial infarction, myocardial ischemia-reperfusion, cryoinjury, and transverse aortic constriction. Lineage tracing experiments show that adult endocardium minimally contributes to coronary ECs after myocardial infarction. In the myocardial ischemia-reperfusion, cryoinjury, or transverse aortic constriction models, adult endocardial cells do not give rise to any vascular ECs, and they remain on the inner surface of myocardium that connects with lumen circulation. In the myocardial infarction model, very few endocardial cells are trapped in the infarct zone of myocardium shortly after ligation of coronary artery, indicating the involvement of endocardial entrapment during blood vessels formation. When these adult endocardial cells are relocated and trapped in the infarcted myocardium by transplantation or myocardial constriction model, very few endocardial cells survive and gain vascular EC properties, and their contribution to neovascularization in the injured myocardium remains minimal. CONCLUSIONS: Unlike its fetal or neonatal counterpart, adult endocardium naturally generates minimal, if any, coronary arteries or vascular ECs during cardiac homeostasis or after injuries.


Asunto(s)
Linaje de la Célula/genética , Endocardio/citología , Endotelio Vascular/citología , Neovascularización Fisiológica , Trasplante de Células Madre/métodos , Animales , Estenosis de la Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/terapia , Línea Celular , Transdiferenciación Celular , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Endocardio/metabolismo , Endotelio Vascular/metabolismo , Humanos , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/terapia , Receptores del Factor Natriurético Atrial/genética , Receptores del Factor Natriurético Atrial/metabolismo
10.
Circ Res ; 123(1): 86-99, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29764841

RESUMEN

RATIONALE: Organs of the body require vascular networks to supply oxygen and nutrients and maintain physiological function. The blood vessels of different organs are structurally and functionally heterogeneous in nature. To more precisely dissect their distinct in vivo function in individual organs, without potential interference from off-site targets, it is necessary to genetically target them in an organ-specific manner. OBJECTIVE: The objective of this study was to generate a genetic system that targets vascular endothelial cells in an organ- or tissue-specific manner and to exemplify the potential application of intersectional genetics for precise, target-specific gene manipulation in vivo. METHODS AND RESULTS: We took advantage of 2 orthogonal recombination systems, Dre-rox and Cre-loxP, to create a genetic targeting system based on intersectional genetics. Using this approach, Cre activity was only detectable in cells that had expressed both Dre and Cre. Applying this new system, we generated a coronary endothelial cell-specific Cre (CoEC-Cre) and a brain endothelial cell-specific Cre (BEC-Cre). Through lineage tracing, gene knockout and overexpression experiments, we demonstrated that CoEC-Cre and BEC-Cre efficiently and specifically target blood vessels in the heart and brain, respectively. By deletion of vascular endothelial growth factor receptor 2 using BEC-Cre, we showed that vascular endothelial growth factor signaling regulates angiogenesis in the central nervous system and also controls the integrity of the blood-brain barrier. CONCLUSIONS: We provide 2 examples to illustrate the use of intersectional genetics for more precise gene targeting in vivo, namely manipulation of genes in blood vessels of the heart and brain. More broadly, this system provides a valuable strategy for tissue-specific gene manipulation that can be widely applied to other fields of biomedical research.


Asunto(s)
Vasos Sanguíneos , Encéfalo/irrigación sanguínea , Vasos Coronarios , Marcación de Gen/métodos , Animales , Barrera Hematoencefálica , Hipoxia de la Célula , Células Endoteliales , Técnicas de Inactivación de Genes , Hibridación in Situ/métodos , Ratones , Neovascularización Fisiológica , Especificidad de Órganos , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
11.
Circulation ; 138(8): 793-805, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-29700121

RESUMEN

BACKGROUND: Whether the adult mammalian heart harbors cardiac stem cells for regeneration of cardiomyocytes is an important yet contentious topic in the field of cardiovascular regeneration. The putative myocyte stem cell populations recognized without specific cell markers, such as the cardiosphere-derived cells, or with markers such as Sca1+, Bmi1+, Isl1+, or Abcg2+ cardiac stem cells have been reported. Moreover, it remains unclear whether putative cardiac stem cells with unknown or unidentified markers exist and give rise to de novo cardiomyocytes in the adult heart. METHODS: To address this question without relying on a particular stem cell marker, we developed a new genetic lineage tracing system to label all nonmyocyte populations that contain putative cardiac stem cells. Using dual lineage tracing system, we assessed whether nonmyocytes generated any new myocytes during embryonic development, during adult homeostasis, and after myocardial infarction. Skeletal muscle was also examined after injury for internal control of new myocyte generation from nonmyocytes. RESULTS: By this stem cell marker-free and dual recombinases-mediated cell tracking approach, our fate mapping data show that new myocytes arise from nonmyocytes in the embryonic heart, but not in the adult heart during homeostasis or after myocardial infarction. As positive control, our lineage tracing system detected new myocytes derived from nonmyocytes in the skeletal muscle after injury. CONCLUSIONS: This study provides in vivo genetic evidence for nonmyocyte to myocyte conversion in embryonic but not adult heart, arguing again the myogenic potential of putative stem cell populations for cardiac regeneration in the adult stage. This study also provides a new genetic strategy to identify endogenous stem cells, if any, in other organ systems for tissue repair and regeneration.


Asunto(s)
Células Madre Adultas/fisiología , Diferenciación Celular , Linaje de la Célula , Rastreo Celular/métodos , Corazón/embriología , Integrasas/genética , Células Madre Embrionarias de Ratones/fisiología , Miocitos Cardíacos/fisiología , Células Madre Adultas/metabolismo , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Proteínas de Escherichia coli/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Fenotipo , Recombinasas/genética , Regeneración , Transducción de Señal
12.
J Biol Chem ; 292(21): 8594-8604, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28377509

RESUMEN

The liver possesses a remarkable capacity to regenerate after damage. There is a heated debate on the origin of new hepatocytes after injuries in adult liver. Hepatic stem/progenitor cells have been proposed to produce functional hepatocytes after injury. Recent studies have argued against this model and suggested that pre-existing hepatocytes, rather than stem cells, contribute new hepatocytes. This hepatocyte-to-hepatocyte model is mainly based on labeling of hepatocytes with Cre-recombinase delivered by the adeno-associated virus. However, the impact of virus infection on cell fate determination, consistency of infection efficiency, and duration of Cre-virus in hepatocytes remain confounding factors that interfere with the data interpretation. Here, we generated a new genetic tool Alb-DreER to label almost all hepatocytes (>99.5%) and track their contribution to different cell lineages in the liver. By "pulse-and-chase" strategy, we found that pre-existing hepatocytes labeled by Alb-DreER contribute to almost all hepatocytes during normal homeostasis and after liver injury. Virtually all hepatocytes in the injured liver are descendants of pre-existing hepatocytes through self-expansion. We concluded that stem cell differentiation is unlikely to be responsible for the generation of a substantial number of new hepatocytes in adult liver. Our study also provides a new mouse tool for more precise in vivo genetic study of hepatocytes in the field.


Asunto(s)
Diferenciación Celular , Hepatocitos , Regeneración Hepática , Hígado , Células Madre , Animales , Rastreo Celular/métodos , Hepatocitos/metabolismo , Hepatocitos/patología , Integrasas/biosíntesis , Integrasas/genética , Hígado/lesiones , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Transgénicos , Células Madre/metabolismo , Células Madre/patología
14.
Circ Res ; 118(12): 1880-93, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27056912

RESUMEN

RATIONALE: There is persistent uncertainty regarding the developmental origins of coronary vessels, with 2 principal sources suggested as ventricular endocardium or sinus venosus (SV). These 2 proposed origins implicate fundamentally distinct mechanisms of vessel formation. Resolution of this controversy is critical for deciphering the programs that result in the formation of coronary vessels and has implications for research on therapeutic angiogenesis. OBJECTIVE: To resolve the controversy over the developmental origin of coronary vessels. METHODS AND RESULTS: We first generated nuclear factor of activated T cells (Nfatc1)-Cre and Nfatc1-Dre lineage tracers for endocardium labeling. We found that Nfatc1 recombinases also label a significant portion of SV endothelial cells in addition to endocardium. Therefore, restricted endocardial lineage tracing requires a specific marker that distinguishes endocardium from SV. By single-cell gene expression analysis, we identified a novel endocardial gene natriuretic peptide receptor 3 (Npr3). Npr3 is expressed in the entirety of the endocardium but not in the SV. Genetic lineage tracing based on Npr3-CreER showed that endocardium contributes to a minority of coronary vessels in the free walls of embryonic heart. Intersectional genetic lineage tracing experiments demonstrated that endocardium minimally contributes to coronary endothelium in the embryonic ventricular free walls. CONCLUSIONS: Our study suggested that SV, but not endocardium, is the major origin for coronary endothelium in the embryonic ventricular free walls. This work thus resolves the recent controversy over the developmental origin of coronary endothelium, providing the basis for studying coronary vessel formation and regeneration after injury.


Asunto(s)
Linaje de la Célula , Vasos Coronarios/embriología , Endocardio/embriología , Endotelio Vascular/metabolismo , Ventrículos Cardíacos/embriología , Animales , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Endocardio/citología , Endocardio/metabolismo , Endotelio Vascular/citología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Ratones , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Receptores del Factor Natriurético Atrial/genética , Receptores del Factor Natriurético Atrial/metabolismo
15.
Circ Res ; 118(2): 254-65, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26659641

RESUMEN

RATIONALE: Unraveling the developmental origin of cardiac fat could offer important implications for the treatment of cardiovascular disease. The recent identification of the mesothelial source of epicardial fat tissues reveals a heterogeneous origin of adipocytes in the adult heart. However, the developmental origin of adipocytes inside the heart, namely intramyocardial adipocytes, remains largely unknown. OBJECTIVE: To trace the developmental origin of intramyocardial adipocytes. METHODS AND RESULTS: In this study, we identified that the majority of intramyocardial adipocytes were restricted to myocardial regions in close proximity to the endocardium. Using a genetic lineage tracing model of endocardial cells, we found that Nfatc1(+) endocardial cells contributed to a substantial number of intramyocardial adipocytes. Despite the capability of the endocardium to generate coronary vascular endothelial cells surrounding the intramyocardial adipocytes, results from our lineage tracing analyses showed that intramyocardial adipocytes were not derived from coronary vessels. Nevertheless, the endocardium of the postnatal heart did not contribute to intramyocardial adipocytes during homeostasis or after myocardial infarction. CONCLUSIONS: Our in vivo fate-mapping studies demonstrated that the developing endocardium, but not the vascular endothelial cells, gives rise to intramyocardial adipocytes in the adult heart.


Asunto(s)
Adipocitos/citología , Adipogénesis , Linaje de la Célula , Endocardio/citología , Corazón Fetal/citología , Adipocitos/metabolismo , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Endocardio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Homeostasis , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Fenotipo
16.
J Biol Chem ; 289(27): 18681-92, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24831012

RESUMEN

Cardiac malformations due to aberrant development of the atrioventricular (AV) valves are among the most common forms of congenital heart diseases. Normally, heart valve mesenchyme is formed from an endothelial to mesenchymal transition (EMT) of endothelial cells of the endocardial cushions. Yes-associated protein 1 (YAP1) has been reported to regulate EMT in vitro, in addition to its known role as a major regulator of organ size and cell proliferation in vertebrates, leading us to hypothesize that YAP1 is required for heart valve development. We tested this hypothesis by conditional inactivation of YAP1 in endothelial cells and their derivatives. This resulted in markedly hypocellular endocardial cushions due to impaired formation of heart valve mesenchyme by EMT and to reduced endocardial cell proliferation. In endothelial cells, TGFß induces nuclear localization of Smad2/3/4 complex, which activates expression of Snail, Twist1, and Slug, key transcription factors required for EMT. YAP1 interacts with this complex, and loss of YAP1 disrupts TGFß-induced up-regulation of Snail, Twist1, and Slug. Together, our results identify a role of YAP1 in regulating EMT through modulation of TGFß-Smad signaling and through proliferative activity during cardiac cushion development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transdiferenciación Celular , Cojinetes Endocárdicos/citología , Cojinetes Endocárdicos/embriología , Células Endoteliales/citología , Mesodermo/citología , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Linaje de la Célula , Endocardio/citología , Endocardio/embriología , Endocardio/metabolismo , Femenino , Eliminación de Gen , Masculino , Ratones , Mutación , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Transducción de Señal , Proteínas Smad/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Proteínas Señalizadoras YAP
19.
Biophys Rep ; 9(6): 309-324, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38524699

RESUMEN

The liver consists predominantly of hepatocytes and biliary epithelial cells (BECs), which serve distinct physiological functions. Although hepatocytes primarily replenish their own population during homeostasis and injury repair, recent findings have suggested that BECs can transdifferentiate into hepatocytes when hepatocyte-mediated liver regeneration is impaired. However, the cellular and molecular mechanisms governing this BEC-to-hepatocyte conversion remain poorly understood largely because of the inefficiency of existing methods for inducing lineage conversion. Therefore, this study introduces a novel mouse model engineered by the Zhou's lab, where hepatocyte senescence is induced by the deletion of the fumarylacetoacetate (Fah) gene. This model facilitates the efficient conversion of BECs to hepatocytes and allows for the simultaneous lineage tracing of BECs; consequently, a transitional liver progenitor cell population can be identified during lineage conversion. This study also outlines the technical procedures for utilizing this model to determine the underlying cellular and molecular mechanisms of BEC-to-hepatocyte conversion and provides new insights into liver regeneration and its underlying molecular mechanism.

20.
Dev Cell ; 58(16): 1502-1512.e3, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37348503

RESUMEN

Cardiac resident macrophages play vital roles in heart development, homeostasis, repair, and regeneration. Recent studies documented the hematopoietic potential of cardiac endothelium that supports the generation of cardiac macrophages and peripheral blood cells in mice. However, the conclusion was not strongly supported by previous genetic tracing studies, given the non-specific nature of conventional Cre-loxP tracing tools. Here, we develop an intercellular genetic labeling system that can permanently trace heart-specific endothelial cells based on cell-cell interaction in mice. Results from cell-cell contact-mediated genetic fate mapping demonstrate that cardiac endothelial cells do not exhibit hemogenic potential and do not contribute to cardiac macrophages or other circulating blood cells. This Matters Arising paper is in response to Shigeta et al. (2019), published in Developmental Cell. See also the response by Liu and Nakano (2023), published in this issue.


Asunto(s)
Células Endoteliales , Corazón , Ratones , Animales , Linaje de la Célula/genética , Diferenciación Celular , Endotelio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA