Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Biochem ; 125(4): e30541, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38372186

RESUMEN

Epithelial cells (ECs) have been proposed to contribute to myofibroblasts or fibroblasts through epithelial-mesenchymal transition (EMT) during renal fibrosis. However, since EMT may occur dynamically, transiently, and reversibly during kidney fibrosis, conventional lineage tracing based on Cre-loxP recombination in renal ECs could hardly capture the transient EMT activity, yielding inconsistent results. Moreover, previous EMT research has primarily focused on renal proximal tubule ECs, with few reports of distal tubules and collecting ducts. Here, we generated dual recombinases-mediated genetic lineage tracing systems for continuous monitoring of transient mesenchymal gene expression in E-cadherin+ and EpCAM+ ECs of distal tubules and collecting ducts during renal fibrosis. Activation of key EMT-inducing transcription factor (EMT-TF) Zeb1 and mesenchymal markers αSMA, vimentin, and N-cadherin, were investigated following unilateral ureteral obstruction (UUO). Our data revealed that E-cadherin+ and EpCAM+ ECs did not transdifferentiate into myofibroblasts, nor transiently expressed these mesenchymal genes during renal fibrosis. In contrast, in vitro a large amount of cultured renal ECs upregulated mesenchymal genes in response to TGF-ß, a major inducer of EMT.


Asunto(s)
Transición Epitelial-Mesenquimal , Enfermedades Renales , Humanos , Molécula de Adhesión Celular Epitelial/genética , Molécula de Adhesión Celular Epitelial/metabolismo , Fibrosis , Enfermedades Renales/metabolismo , Células Epiteliales/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
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.
Circ Res ; 130(3): 352-365, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-34995101

RESUMEN

BACKGROUND: Unraveling how new coronary arteries develop may provide critical information for establishing novel therapeutic approaches to treating ischemic cardiac diseases. There are 2 distinct coronary vascular populations derived from different origins in the developing heart. Understanding the formation of coronary arteries may provide insights into new ways of promoting coronary artery formation after myocardial infarction. METHODS: To understand how intramyocardial coronary arteries are generated to connect these 2 coronary vascular populations, we combined genetic lineage tracing, light sheet microscopy, fluorescence micro-optical sectioning tomography, and tissue-specific gene knockout approaches to understand their cellular and molecular mechanisms. RESULTS: We show that a subset of intramyocardial coronary arteries form by angiogenic extension of endocardium-derived vascular tunnels in the neonatal heart. Three-dimensional whole-mount fluorescence imaging showed that these endocardium-derived vascular tunnels or tubes adopt an arterial fate in neonates. Mechanistically, we implicate Mettl3 (methyltransferase-like protein 3) and Notch signaling in regulating endocardium-derived intramyocardial coronary artery formation. Functionally, these intramyocardial arteries persist into adulthood and play a protective role after myocardial infarction. CONCLUSIONS: A subset of intramyocardial coronary arteries form by extension of endocardium-derived vascular tunnels in the neonatal heart.


Asunto(s)
Vasos Coronarios/embriología , Endocardio/embriología , Animales , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Endocardio/crecimiento & desarrollo , Endocardio/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Organogénesis
4.
J Nat Prod ; 87(4): 876-883, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38377956

RESUMEN

Cytochrome P450-modified bacterial terpenoids remain in a vast chemical space to be explored. In the present study, we conducted global genome mining of 223,829 bacterial genomes and identified 2892 bacterial terpenoid biosynthetic gene clusters (BGCs) with cytochrome P450 genes. Among these, we selected 562 with multiple P450 enzymes, which were further clustered as 355 gene cluster families by sequence similarity analysis. We then chose lev, a BGC from Streptomyces levis MCCC1A01616, for heterologous expression and discovered four new α-amorphene-type sesquiterpenoids, levinoids A-D (1-4). The structures and absolute configurations of these four new compounds were determined by employing extensive NMR analysis, NMR chemical shift calculations with DP4+, and ECD calculations. Furthermore, levinoid C (3) exhibited a moderate level of neuroprotective activity (EC50 = 21 µM) in the glutamate-induced excitotoxicity cell model. Our findings highlight the untapped chemical diversity of P450-modified bacterial terpenoids, opening new avenues for further exploration and discovery.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Sesquiterpenos , Streptomyces , Sesquiterpenos/química , Sesquiterpenos/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Streptomyces/química , Streptomyces/genética , Estructura Molecular , Genoma Bacteriano , Familia de Multigenes , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/química
5.
BMC Public Health ; 24(1): 1032, 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38615002

RESUMEN

BACKGROUND: Hemorrhagic fever with renal syndrome (HFRS) signals a recurring risk in Eurasia in recent years owing to its continued rise in case notifications and the extension of geographical distribution. This study was undertaken to investigate the spatiotemporal drivers and incidence heterogeneity of HFRS transmission in Shandong Province. METHODS: The epidemiological data for HFRS, meteorological data and socioeconomic data were obtained from China Information System for Disease Control and Prevention, China Meteorological Data Sharing Service System, and Shandong Statistical Yearbook, respectively. The spatial-temporal multicomponent model was employed to analyze the values of spatial-temporal components and the heterogeneity of HFRS transmission across distinct regions. RESULTS: The total effect values of the autoregressive, epidemic, and endemic components were 0.451, 0.187, and 0.033, respectively, exhibiting significant heterogeneity across various cities. This suggested a pivotal role of the autoregressive component in propelling HFRS transmission in Shandong Province. The epidemic component of Qingdao, Weifang, Yantai, Weihai, and Jining declined sharply at the onset of 2020. The random effect identified distinct incidence levels associated with Qingdao and Weifang, signifying regional variations in HFRS occurrence. CONCLUSIONS: The autoregressive component emerged as a significant driver in the transmission of HFRS in Shandong Province. Targeted preventive measures should be strategically implemented across various regions, taking into account the predominant component influencing the epidemic.


Asunto(s)
Epidemias , Fiebre Hemorrágica con Síndrome Renal , Humanos , Fiebre Hemorrágica con Síndrome Renal/epidemiología , Incidencia , China/epidemiología , Ciudades
6.
BMC Infect Dis ; 23(1): 891, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38124061

RESUMEN

BACKGROUND: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease discovered in China in 2009. The purpose of this study was to describe the spatiotemporal distribution of SFTS and to identify its environmental influencing factors and potential high-risk areas in Shandong Province, China. METHODS: Data on the SFTS incidence from 2010 to 2021 were collected. Spatiotemporal scan statistics were used to identify the time and area of SFTS clustering. The maximum entropy (MaxEnt) model was used to analyse environmental influences and predict high-risk areas. RESULTS: From 2010 to 2021, a total of 5705 cases of SFTS were reported in Shandong. The number of SFTS cases increased yearly, with a peak incidence from April to October each year. Spatiotemporal scan statistics showed the existence of one most likely cluster and two secondary likely clusters in Shandong. The most likely cluster was in the eastern region, from May to October 2021. The first secondary cluster was in the central region, from May to October 2021. The second secondary cluster was in the southeastern region, from May to September 2020. The MaxEnt model showed that the mean annual wind speed, NDVI, cattle density and annual cumulative precipitation were the key factors influencing the occurrence of SFTS. The predicted risk map showed that the area of high prevalence was 28,120 km2, accounting for 18.05% of the total area of the province. CONCLUSIONS: The spatiotemporal distribution of SFTS was heterogeneous and influenced by multidimensional environmental factors. This should be considered as a basis for delineating SFTS risk areas and developing SFTS prevention and control measures.


Asunto(s)
Phlebovirus , Síndrome de Trombocitopenia Febril Grave , Trombocitopenia , Animales , Bovinos , Trombocitopenia/epidemiología , Incidencia , China/epidemiología
7.
J Mol Cell Cardiol ; 167: 67-82, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35354073

RESUMEN

Neovascularization of the ischemic myocardium following infarction is vital for the survival of cardiomyocytes and prevention of heart failure. However, the intrinsic revascularization following ischemic injury in the heart is inadequate to restore blood flow to the infarcted myocardium. A comprehensive understanding of how coronary vasculature is constructed and what developmental pathways might be reactivated after infarction is beneficial to develop effective strategies for heart revascularization. The latest lineage tracing studies of coronary vasculature have revealed multiple developmental origins for coronary vascular endothelial cells. The development and growth of coronary vessels emanated from different cellular origins are governed by distinct regulatory mechanisms. Here, we highlight recent research advancements on cellular sources and molecular mechanisms of coronary vessel formation during heart development and regeneration, and also elaborate on how these mechanisms can be reactivated or recapitulated to facilitate therapeutic revascularization in ischemic heart disease.


Asunto(s)
Vasos Coronarios , Células Endoteliales , Humanos , Infarto , Miocardio , Miocitos Cardíacos , Neovascularización Fisiológica/fisiología
8.
J Biol Chem ; 296: 100509, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33676891

RESUMEN

Site-specific recombinases (SSRs) are invaluable genome engineering tools that have enormously boosted our understanding of gene functions and cell lineage relationships in developmental biology, stem cell biology, regenerative medicine, and multiple diseases. However, the ever-increasing complexity of biomedical research requires the development of novel site-specific genetic recombination technologies that can manipulate genomic DNA with high efficiency and fine spatiotemporal control. Here, we review the latest innovative strategies of the commonly used Cre-loxP recombination system and its combinatorial strategies with other site-specific recombinase systems. We also highlight recent progress with a focus on the new generation of chemical- and light-inducible genetic systems and discuss the merits and limitations of each new and established system. Finally, we provide the future perspectives of combining various recombination systems or improving well-established site-specific genetic tools to achieve more efficient and precise spatiotemporal genetic manipulation.


Asunto(s)
ADN Nucleotidiltransferasas/metabolismo , Recombinación Genética , Sistemas CRISPR-Cas , Catálisis , Activación Enzimática , Integrasas/metabolismo , Luz , Plantas/enzimología , Regiones Promotoras Genéticas
9.
Development ; 145(18)2018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-30111655

RESUMEN

In vivo genomic engineering is instrumental for studying developmental biology and regenerative medicine. Development of novel systems with more site-specific recombinases (SSRs) that complement with the commonly used Cre-loxP would be valuable for more precise lineage tracing and genome editing. Here, we introduce a new SSR system via Nigri-nox. By generating tissue-specific Nigri knock-in and its responding nox reporter mice, we show that the Nigri-nox system works efficiently in vivo by targeting specific tissues. As a new orthogonal system to Cre-loxP, Nigri-nox provides an additional control of genetic manipulation. We also demonstrate how the two orthogonal systems Nigri-nox and Cre-loxP could be used simultaneously to map the cell fate of two distinct developmental origins of cardiac valve mesenchyme in the mouse heart, providing dynamics of cellular contribution from different origins for cardiac valve mesenchyme during development. This work provides a proof-of-principle application of the Nigri-nox system for in vivo mouse genomic engineering. Coupled with other SSR systems, Nigri-nox would be valuable for more precise delineation of origins and cell fates during development, diseases and regeneration.


Asunto(s)
ADN Nucleotidiltransferasas/metabolismo , Ingeniería Genética/métodos , Válvulas Cardíacas/embriología , Mesodermo/embriología , Animales , Antígenos CD/metabolismo , Sistemas CRISPR-Cas/genética , Cadherinas/metabolismo , Células Endoteliales/citología , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL
10.
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
11.
Nature ; 525(7570): 479-85, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26375005

RESUMEN

The elucidation of factors that activate the regeneration of the adult mammalian heart is of major scientific and therapeutic importance. Here we found that epicardial cells contain a potent cardiogenic activity identified as follistatin-like 1 (Fstl1). Epicardial Fstl1 declines following myocardial infarction and is replaced by myocardial expression. Myocardial Fstl1 does not promote regeneration, either basally or upon transgenic overexpression. Application of the human Fstl1 protein (FSTL1) via an epicardial patch stimulates cell cycle entry and division of pre-existing cardiomyocytes, improving cardiac function and survival in mouse and swine models of myocardial infarction. The data suggest that the loss of epicardial FSTL1 is a maladaptive response to injury, and that its restoration would be an effective way to reverse myocardial death and remodelling following myocardial infarction in humans.


Asunto(s)
Proteínas Relacionadas con la Folistatina/metabolismo , Miocardio/metabolismo , Pericardio/crecimiento & desarrollo , Pericardio/metabolismo , Regeneración , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Proteínas Relacionadas con la Folistatina/genética , Humanos , Masculino , Ratones , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/efectos de los fármacos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pericardio/citología , Pericardio/efectos de los fármacos , Ratas , Regeneración/efectos de los fármacos , Transducción de Señal , Porcinos , Transgenes/genética
12.
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
13.
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
14.
Development ; 143(6): 936-49, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26893347

RESUMEN

Although the mammalian heart can regenerate during the neonatal stage, this endogenous regenerative capacity is lost with age. Importantly, replication of cardiomyocytes has been found to be the key mechanism responsible for neonatal cardiac regeneration. Unraveling the transcriptional regulatory network for inducing cardiomyocyte replication will, therefore, be crucial for the development of novel therapies to drive cardiac repair after injury. Here, we investigated whether the key cardiac transcription factor GATA4 is required for neonatal mouse heart regeneration. Using the neonatal mouse heart cryoinjury and apical resection models with an inducible loss of GATA4 specifically in cardiomyocytes, we found severely depressed ventricular function in the Gata4-ablated mice (mutant) after injury. This was accompanied by reduced cardiomyocyte replication. In addition, the mutant hearts displayed impaired coronary angiogenesis and increased hypertrophy and fibrosis after injury. Mechanistically, we found that the paracrine factor FGF16 was significantly reduced in the mutant hearts after injury compared with littermate controls and was directly regulated by GATA4. Cardiac-specific overexpression of FGF16 via adeno-associated virus subtype 9 (AAV9) in the mutant hearts partially rescued the cryoinjury-induced cardiac hypertrophy, promoted cardiomyocyte replication and improved heart function after injury. Altogether, our data demonstrate that GATA4 is required for neonatal heart regeneration through regulation of Fgf16, suggesting that paracrine factors could be of potential use in promoting myocardial repair.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción GATA4/metabolismo , Corazón/fisiopatología , Regeneración , Animales , Animales Recién Nacidos , Secuencia de Bases , Proliferación Celular , Dependovirus/metabolismo , Eliminación de Gen , Ratones Noqueados , Datos de Secuencia Molecular , Mutación/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Especificidad de Órganos , Fenotipo
15.
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
16.
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
17.
Mar Drugs ; 16(1)2018 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-29346329

RESUMEN

Thirty-one isolates belonging to eight genera in seven orders were identified from 141 strains that were isolated from several marine plants. Alternaria sp. and Fusarium sp. were found to be the predominant fungi. Evaluation of the anti-phytopathogenic bacterial and fungal activities, as well as the cytotoxicity of these 31 extracts, revealed that most of them displayed different levels of bioactivities. Due to their interesting bioactivities, two fungal strains-Fusarium equiseti (P18) and Alternaria sp. (P8)-were selected for chemical investigation and compounds 1-4 were obtained. The structure of 1 was elucidated by 1D and 2D NMR analysis, as well as high-resolution electrospray ionization mass spectroscopy (HRESIMS), and the absolute configuration of its stereogenic carbon (C-11) was established by comparison of the experimental and calculated electronic circular-dichroism (ECD) spectra. Moreover, alterperylenol (4) exhibited antibacterial activity against Clavibacter michiganensis with a minimum inhibitory concentration (MIC) of 1.95 µg/mL, which was 2-fold stronger than that of streptomycin sulfate. Additionally, an antibacterial mechanism study revealed that 4 caused membrane hyperpolarization without evidence of destruction of cell membrane integrity. Furthermore, stemphyperylenol (3) displayed potent antifungal activity against Pestallozzia theae and Alternaria brassicicola with MIC values equal to those of carbendazim. The cytotoxicity of 1 and 2 against human lung carcinoma (A-549), human cervical carcinoma (HeLa), and human hepatoma (HepG2) cell lines were also evaluated.


Asunto(s)
Antibacterianos/farmacología , Antifúngicos/farmacología , Organismos Acuáticos/química , Mezclas Complejas/farmacología , Citotoxinas/farmacología , Hongos/química , Hongos/metabolismo , Células A549 , Alternaria/química , Antibacterianos/química , Antifúngicos/química , Bacterias/efectos de los fármacos , Línea Celular Tumoral , Mezclas Complejas/química , Citotoxinas/química , Hongos/efectos de los fármacos , Fusarium/química , Células HeLa , Células Hep G2 , Humanos , Pruebas de Sensibilidad Microbiana/métodos
18.
Circ Res ; 116(3): 515-30, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25634974

RESUMEN

Coronary artery disease causes acute myocardial infarction and heart failure. Identifying coronary vascular progenitors and their developmental program could inspire novel regenerative treatments for cardiac diseases. The developmental origins of the coronary vessels have been shrouded in mystery and debated for several decades. Recent identification of progenitors for coronary vessels within the endocardium, epicardium, and sinus venosus provides new insights into this question. In addition, significant progress has been achieved in elucidating the cellular and molecular programs that orchestrate coronary artery development. Establishing adequate vascular supply will be an essential component of cardiac regenerative strategies, and these findings raise exciting new strategies for therapeutic cardiac revascularization.


Asunto(s)
Linaje de la Célula , Vasos Coronarios/embriología , Neovascularización Fisiológica , Organogénesis , Animales , Vasos Coronarios/citología , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Humanos
19.
Biochem Biophys Res Commun ; 471(4): 430-6, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26902114

RESUMEN

Recent identification of the neonatal 2nd coronary vascular population (2nd CVP) suggests that a subset of these vessels form de novo and mature in the inner myocardial wall of the postnatal heart. However, the origin of smooth muscle cells (SMCs) in the postnatal 2nd CVP remains undetermined. Using a tamoxifen-inducible Wt1-CreER driver and a Rosa26-RFP reporter line, we traced the lineage of epicardial cells to determine if they contribute to SMCs of the 2nd CVP. Late embryonic and postnatal induction of Wt1-CreER activity demonstrated that at these stages Wt1-labeled epicardium does not significantly migrate into the myocardium to form SMCs. However, following tamoxifen treatment at an early embryonic stage (E10.5), we detected Wt1 descendants (epicardium-derived cells, or EPDCs) in the outer myocardial wall at E17.5. When the 2nd CVP forms and remodels at postnatal stage, these early labeled EDPCs re-migrate deep into the inner myocardial wall and contribute to 2nd CVP-SMCs in the adult heart. Our findings reveal that SMCs in the postnatal 2nd CVP are pre-specified as EPDCs from the earliest wave of epicardial cell migration. Rather than the re-activation and migration of epicardial cells at later stages, these resident EPDCs mobilize and contribute to smooth muscle of the 2nd CVP during postnatal development.


Asunto(s)
Vasos Coronarios/citología , Músculo Liso Vascular/citología , Músculo Liso Vascular/embriología , Animales , Animales Recién Nacidos , Movimiento Celular , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/embriología , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/citología , Pericardio/citología , Pericardio/embriología , Tamoxifeno
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA