Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 43(10): 1900-1920, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37589142

RESUMEN

BACKGROUND: Thoracic aortic dissection (TAD) is a life-threatening aortic disease without effective medical treatment. Increasing evidence has suggested a role for NE (neutrophil elastase) in vascular diseases. In this study, we aimed at investigating a causal role for NE in TAD and exploring the molecular mechanisms involved. METHODS: ß-aminopropionitrile monofumarate was administrated in mice to induce TAD. NE deficiency mice, pharmacological inhibitor GW311616A, and adeno-associated virus-2-mediated in vivo gene transfer were applied to explore a causal role for NE and associated target gene in TAD formation. Multiple functional assays and biochemical analyses were conducted to unravel the underlying cellular and molecular mechanisms of NE in TAD. RESULTS: NE aortic gene expression and plasma activity was significantly increased during ß-aminopropionitrile monofumarate-induced TAD and in patients with acute TAD. NE deficiency prevents ß-aminopropionitrile monofumarate-induced TAD onset/development, and GW311616A administration ameliorated TAD formation/progression. Decreased levels of neutrophil extracellular traps, inflammatory cells, and MMP (matrix metalloproteinase)-2/9 were observed in NE-deficient mice. TBL1x (F-box-like/WD repeat-containing protein TBL1x) has been identified as a novel substrate and functional downstream target of NE in TAD. Loss-of-function studies revealed that NE mediated inflammatory cell transendothelial migration by modulating TBL1x-LTA4H (leukotriene A4 hydrolase) signaling and that NE regulated smooth muscle cell phenotype modulation under TAD pathological condition by regulating TBL1x-MECP2 (methyl CpG-binding protein 2) signal axis. Further mechanistic studies showed that TBL1x inhibition decreased the binding of TBL1x and HDAC3 (histone deacetylase 3) to MECP2 and LTA4H gene promoters, respectively. Finally, adeno-associated virus-2-mediated Tbl1x gene knockdown in aortic smooth muscle cells confirmed a regulatory role for TBL1x in NE-mediated TAD formation. CONCLUSIONS: We unravel a critical role of NE and its target TBL1x in regulating inflammatory cell migration and smooth muscle cell phenotype modulation in the context of TAD. Our findings suggest that the NE-TBL1x signal axis represents a valuable therapeutic for treating high-risk TAD patients.


Asunto(s)
Aneurisma de la Aorta Torácica , Disección Aórtica , Disección de la Aorta Torácica , Animales , Humanos , Ratones , Aminopropionitrilo/toxicidad , Aneurisma de la Aorta Torácica/inducido químicamente , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/metabolismo , Disección Aórtica/inducido químicamente , Disección Aórtica/genética , Elastasa de Leucocito/genética , Elastasa de Leucocito/efectos adversos
3.
Artículo en Inglés | MEDLINE | ID: mdl-37386312

RESUMEN

Sluggish cognitive tempo (SCT) was initially studied in the context of attention deficit hyperactivity disorder (ADHD), but is now recognized as a distinct disorder. Despite the growing recognition of SCT, its impact on academic achievement among adolescents remains controversial, even when controlling for the level of ADHD. This may be due to the influence of other factors such as learning engagement and emotional distress. To address this gap, we conducted a longitudinal study with a sample of 782 Chinese senior high school students, measuring their SCT, learning engagement, and emotional distress at Grade 10 (Time1, T1) to predict their academic achievement evaluated based on final exams scores five months later (Time2, T2). Results showed that learning engagement mediated the negative relationship between SCT and later academic achievement. Additionally, individuals with high SCT showed less impact by emotional distress on learning engagement. These findings may shed light on the complex interplay between SCT, emotional distress and learning engagement in shaping academic achievement, underscoring the potential adaptive function of SCT as a coping strategy for managing emotional challenges.

4.
Hepatology ; 74(3): 1251-1270, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33713358

RESUMEN

BACKGROUND AND AIMS: Characterized by hepatocyte steatosis, inflammation, and fibrosis, NASH is a complicated process that contributes to end-stage liver disease and, eventually, HCC. TNF-α-induced protein 8-like 1 (TIPE1), a new member of the TNF-α-induced protein 8 family, has been explored in immunology and oncology research; but little is known about its role in metabolic diseases. APPROACH AND RESULTS: Here, we show that hepatocyte-specific deletion of TIPE1 exacerbated diet-induced hepatic steatosis, inflammation, and fibrosis as well as systemic metabolic disorders during NASH pathogenesis. Conversely, hepatocyte-specific overexpression of TIPE1 dramatically prevented the progression of these abnormalities. Mechanically, TIPE1 directly interacted with apoptosis signal-regulating kinase 1 (ASK1) to suppress its TNF receptor-associated factor 6 (TRAF6)-catalyzed polyubiquitination activation upon metabolic challenge, thereby inhibiting the downstream c-Jun N-terminal kinase and p38 signaling pathway. Importantly, dramatically reduced TIPE1 expression was observed in the livers of patients with NAFLD, suggesting that TIPE1 might be a promising therapeutic target for NAFLD and related metabolic diseases. CONCLUSIONS: TIPE1 protects against hepatic steatosis, inflammation, and fibrosis through directly binding ASK1 and restraining its TRAF6-catalyzed polyubiquitination during the development of NASH. Therefore, targeting TIPE1 could be a promising therapeutic approach for NAFLD treatment.


Asunto(s)
Hígado Graso/genética , Péptidos y Proteínas de Señalización Intracelular/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Adulto , Anciano , Animales , Dieta Alta en Grasa , Regulación hacia Abajo , Hígado Graso/metabolismo , Hígado Graso/patología , Femenino , Humanos , Inflamación , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Poliubiquitina/metabolismo
5.
J Psycholinguist Res ; 51(3): 485-500, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35084648

RESUMEN

This study aimed to investigate the effects of judgment frames, cues, and test criteria on the accuracy of metacomprehension monitoring. The design was a 2 (rating comprehension vs. predicting performance) × 2 (memory cues vs. comprehension cues) × 2 (detailed questions test vs. inferential questions test) mixed design with judgment frames and cues as the between-subjects factors and test criteria as the within-subjects factor. The results showed that the influence of judgment frames on accuracy was moderated by the test criteria. The readers' monitoring was more accurate in rating comprehension than predicting performance when inferential questions were used as the criteria; when detailed questions were used as the criteria, this situation was reversed. The interaction effect of judgment cues and criteria on metacomprehension monitoring accuracy was significant. When readers predicted their performances on a test, those who received memory cues were more accurate than those who received comprehension cues. However, when readers rated their comprehension, those who received comprehension cues were more accurate than those who received memory cues.


Asunto(s)
Señales (Psicología) , Juicio , Comprensión , Humanos , Lectura
6.
Circ Res ; 125(2): 223-241, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31079549

RESUMEN

RATIONALE: Transplantation-accelerated arteriosclerosis is one of the major challenges for long-term survival of patients with solid organ transplantation. Although stem/progenitor cells have been implicated to participate in this process, the cells of origin and underlying mechanisms have not been fully defined. OBJECTIVE: The objective of our study was to investigate the role of c-Kit lineage cells in allograft-induced neointima formation and to explore the mechanisms underlying this process. METHODS AND RESULTS: Using an inducible lineage tracing Kit-CreER;Rosa26-tdTomato mouse model, we observed that c-Kit is expressed in multiple cell types in the blood vessels, rather than a specific stem/progenitor cell marker. We performed allograft transplantation between different donor and recipient mice, as well as bone marrow transplantation experiments, demonstrating that recipient c-Kit+ cells repopulate neointimal smooth muscle cells (SMCs) and leukocytes, and contribute to neointima formation in an allograft transplantation model. c-Kit-derived SMCs originate from nonbone marrow tissues, whereas bone marrow-derived c-Kit+ cells mainly generate CD45+ leukocytes. However, the exact identity of c-Kit lineage cells contributing to neointimal SMCs remains unclear. ACK2 (anti-c-Kit antibody), which specifically binds and blocks c-Kit function, ameliorates allograft-induced arteriosclerosis. Stem cell factor and TGF (transforming growth factor)-ß1 levels were significantly increased in blood and neointimal lesions after allograft transplantation, by which stem cell factor facilitated c-Kit+ cell migration through the stem cell factor/c-Kit axis and downstream activation of small GTPases, MEK (mitogen-activated protein kinase kinase)/ERK (extracellular signal-regulated kinase)/MLC (myosin light chain), and JNK (c-Jun N-terminal kinase)/c-Jun signaling pathways, whereas TGF-ß1 induces c-Kit+ cell differentiation into SMCs via HK (hexokinase)-1-dependent metabolic reprogramming and a possible downstream O-GlcNAcylation of myocardin and serum response factor. CONCLUSIONS: Our findings provide evidence that recipient c-Kit lineage cells contribute to vascular remodeling in an allograft transplantation model, in which the stem cell factor/c-Kit axis is responsible for cell migration and HK-1-dependent metabolic reprogramming for SMC differentiation.


Asunto(s)
Arteriosclerosis/terapia , Movimiento Celular , Miocitos del Músculo Liso/fisiología , Animales , Aorta/fisiología , Aorta/trasplante , Células Cultivadas , Reprogramación Celular , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regeneración , Factor de Células Madre/metabolismo , Túnica Íntima/citología , Túnica Íntima/fisiología
7.
Cell Mol Life Sci ; 77(24): 5299-5320, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32166394

RESUMEN

Stem/progenitor cells (SPCs) have been implicated to participate in vascular repair. However, the exact role of SPCs in endothelial repair of large vessels still remains controversial. This study aimed to delineate the cellular heterogeneity and possible functional role of endogenous vascular SPCs in large vessels. Using single-cell RNA-sequencing (scRNA-seq) and genetic lineage tracing mouse models, we uncovered the cellular heterogeneity of SPCs, i.e., c-Kit+ cells in the mouse aorta, and found that endogenous c-Kit+ cells acquire endothelial cell fate in the aorta under both physiological and pathological conditions. While c-Kit+ cells contribute to aortic endothelial turnover in the atheroprone regions during homeostasis, recipient c-Kit+ cells of nonbone marrow source replace both luminal and microvessel endothelial cells in transplant arteriosclerosis. Single-cell pseudotime analysis of scRNA-seq data and in vitro cell experiments suggest that vascular SPCs display endothelial differentiation potential and undergo metabolic reprogramming during cell differentiation, in which AKT/mTOR-dependent glycolysis is critical for endothelial gene expression. These findings demonstrate a critical role for c-Kit lineage cells in aortic endothelial turnover and replacement, and may provide insights into therapeutic strategies for vascular diseases.


Asunto(s)
Linaje de la Célula/genética , Endotelio Vascular/crecimiento & desarrollo , Análisis de la Célula Individual/métodos , Células Madre/metabolismo , Animales , Aorta/crecimiento & desarrollo , Aorta/metabolismo , Diferenciación Celular/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Ratones , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-kit/genética , RNA-Seq , Células Madre/citología , Serina-Treonina Quinasas TOR/genética
8.
Proc Natl Acad Sci U S A ; 115(41): 10351-10356, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30249667

RESUMEN

VEGF-B was discovered a long time ago. However, unlike VEGF-A, whose function has been extensively studied, the function of VEGF-B and the mechanisms involved still remain poorly understood. Notwithstanding, drugs that inhibit VEGF-B and other VEGF family members have been used to treat patients with neovascular diseases. It is therefore critical to have a better understanding of VEGF-B function and the underlying mechanisms. Here, using comprehensive methods and models, we have identified VEGF-B as a potent antioxidant. Loss of Vegf-b by gene deletion leads to retinal degeneration in mice, and treatment with VEGF-B rescues retinal cells from death in a retinitis pigmentosa model. Mechanistically, we demonstrate that VEGF-B up-regulates numerous key antioxidative genes, particularly, Gpx1 Loss of Gpx1 activity largely diminished the antioxidative effect of VEGF-B, demonstrating that Gpx1 is at least one of the critical downstream effectors of VEGF-B. In addition, we found that the antioxidant function of VEGF-B is mediated mainly by VEGFR1. Given that oxidative stress is a crucial factor in numerous human diseases, VEGF-B may have therapeutic value for the treatment of such diseases.


Asunto(s)
Antioxidantes/metabolismo , Degeneración Retiniana/genética , Factor B de Crecimiento Endotelial Vascular/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glutatión Peroxidasa/genética , Ratones Endogámicos C57BL , Ratones Mutantes , Estrés Oxidativo , Retina/efectos de los fármacos , Retina/patología , Degeneración Retiniana/tratamiento farmacológico , Retinitis Pigmentosa/genética , Factor B de Crecimiento Endotelial Vascular/genética , Factor B de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Glutatión Peroxidasa GPX1
9.
Proc Natl Acad Sci U S A ; 114(40): 10737-10742, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28923916

RESUMEN

Ocular neovascularization is a devastating pathology of numerous ocular diseases and is a major cause of blindness. Caveolin-1 (Cav-1) plays important roles in the vascular system. However, little is known regarding its function and mechanisms in ocular neovascularization. Here, using comprehensive model systems and a cell permeable peptide of Cav-1, cavtratin, we show that Cav-1 is a critical player in ocular neovascularization. The genetic deletion of Cav-1 exacerbated and cavtratin administration inhibited choroidal and retinal neovascularization. Importantly, combined administration of cavtratin and anti-VEGF-A inhibited neovascularization more effectively than monotherapy, suggesting the existence of other pathways inhibited by cavtratin in addition to VEGF-A. Indeed, we found that cavtratin suppressed multiple critical components of pathological angiogenesis, including inflammation, permeability, PDGF-B and endothelial nitric oxide synthase expression (eNOS). Mechanistically, we show that cavtratin inhibits CNV and the survival and migration of microglia and macrophages via JNK. Together, our data demonstrate the unique advantages of cavtratin in antiangiogenic therapy to treat neovascular diseases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Caveolina 1/fisiología , Neovascularización Coroidal/prevención & control , MAP Quinasa Quinasa 4/metabolismo , Fragmentos de Péptidos/farmacología , Neovascularización Retiniana/prevención & control , Animales , Caveolina 1/farmacología , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Quimioterapia Combinada , Humanos , Ratones Noqueados , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
10.
J Mol Cell Cardiol ; 132: 178-188, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31100313

RESUMEN

AIMS: microRNA-124(miR-124) has recently been reported to be elevated in cardiovascular disease. In this study, we aimed to investigate the exact role of miR-124 in cardiomyocytes and myocardial infarction, identifying the functional target and its regulatory mechanisms. METHODS AND RESULTS: Cultured cardiomyocytes, myocardial-infarction mouse model, and clinical data were used to study the effects of miR-124 on myocardial ischemia. Expression of miR-124 was up-regulated in H2O2 and hypoxia induced cardiomyocyte injury. miR-124 over-expression significantly increased cardiomyocyte apoptosis, whereas miR-124 inhibition attenuated cell death. 3ß-hydroxysteroid-Delta24 reductase (Dhcr24), a multi-functional enzyme implicated in cholesterol synthesis and various diseases, was identified as a novel functional target of miR-124 in cardiac myocytes. The miR-124-Dhcr24 axis was responsible for cardiomyocyte apoptosis regulation. Furthermore, myocardial infarction induced miR-124 activation and Dhcr24 reduction in vivo. Modulation of miR-124 by intra-myocardial injection of agomiR or antagomiR was capable of manipulating cardiomyocyte apoptosis and myocardial infarction in mice. More importantly, circulating miR-124 was also observed to be elevated in acute myocardial infarction (AMI) patients and was correlated with myocardial injury and cardiac function. CONCLUSION: Our findings strongly demonstrated that miR-124 targeting Dhcr24 regulates oxidative stress and hypoxia induced cardiomyocyte apoptosis and myocardial infarction. The miR-124-Dhcr24 axis could be a potential biomarker as well as the therapeutic target for AMI.


Asunto(s)
Apoptosis/fisiología , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Animales , Hipoxia de la Célula/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Regulación hacia Arriba/fisiología
11.
Circulation ; 137(17): 1824-1841, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29246895

RESUMEN

BACKGROUND: MicroRNA-22 (miR-22) has recently been reported to play a regulatory role during vascular smooth muscle cell (VSMC) differentiation from stem cells, but little is known about its target genes and related pathways in mature VSMC phenotypic modulation or its clinical implication in neointima formation following vascular injury. METHODS: We applied a wire-injury mouse model, and local delivery of AgomiR-22 or miR-22 inhibitor, as well, to explore the therapeutic potential of miR-22 in vascular diseases. Furthermore, normal and diseased human femoral arteries were harvested, and various in vivo, ex vivo, and in vitro models of VSMC phenotype switching were conducted to examine miR-22 expression during VSMC phenotype switching. RESULTS: Expression of miR-22 was closely regulated during VSMC phenotypic modulation. miR-22 overexpression significantly increased expression of VSMC marker genes and inhibited VSMC proliferation and migration, whereas the opposite effect was observed when endogenous miR-22 was knocked down. As expected, 2 previously reported miR-22 target genes, MECP2 (methyl-CpG binding protein 2) and histone deacetylase 4, exhibited a regulatory role in VSMC phenotypic modulation. A transcriptional regulator and oncoprotein, EVI1 (ecotropic virus integration site 1 protein homolog), has been identified as a novel miR-22 target gene in VSMC phenotypic modulation. It is noteworthy that overexpression of miR-22 in the injured vessels significantly reduced the expression of its target genes, decreased VSMC proliferation, and inhibited neointima formation in wire-injured femoral arteries, whereas the opposite effect was observed with local application of a miR-22 inhibitor to injured arteries. We next examined the clinical relevance of miR-22 expression and its target genes in human femoral arteries. We found that miR-22 expression was significantly reduced, whereas MECP2 and EVI1 expression levels were dramatically increased, in diseased in comparison with healthy femoral human arteries. This inverse relationship between miR-22 and MECP2 and EVI1 was evident in both healthy and diseased human femoral arteries. CONCLUSIONS: Our data demonstrate that miR-22 and EVI1 are novel regulators of VSMC function, specifically during neointima hyperplasia, offering a novel therapeutic opportunity for treating vascular diseases.


Asunto(s)
MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima , Lesiones del Sistema Vascular/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antagomirs/genética , Antagomirs/metabolismo , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , MicroARNs/genética , Persona de Mediana Edad , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fenotipo , Transducción de Señal , Técnicas de Cultivo de Tejidos , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología
12.
Pharmacol Res ; 146: 104277, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31112749

RESUMEN

Neovascular diseases, such as many cancers and ocular disorders, are life threatening and devastating. Although anti-vascular endothelial growth factor A (VEGF-A) therapy is available, many patients are not responsive and drug resistance can develop. To try to overcome these problems, combination therapy targeting VEGF-A and platelet-derived growth factor B (PDGF-B) was tested. However, one obvious drawback was that the other VEGF and PDGF family members were not inhibited and therefore could compensate. Indeed, this was, at least to some extent, demonstrated by the disappointing outcomes. To this end, we designed novel multi-targeted inhibitors that can block most of the VEGF and PDGF family members simultaneously by making a fusion protein containing the ligand-binding domains of vascular endothelial growth factor receptor 1 (VEGFR1), vascular endothelial growth factor receptor 2 (VEGFR2) and platelet-derived growth factor receptor beta (PDGFRß), which can therefore act as a decoy blocker for most of the VEGF and PDGF family members. Indeed, in cultured cells, the novel inhibitors suppressed the migration and proliferation of both vascular endothelial cells and smooth muscle cells, and abolished VEGFR2 and PDGFRß activation. Importantly, in a choroidal neovascularization model in vivo, the novel inhibitor inhibited ocular neovascularization more efficiently than the mono-inhibitors against VEGFR or PDGFR alone respectively. Mechanistically, a genome-wide microarray analysis unveiled that the novel inhibitor regulated unique sets of genes that were not regulated by the mono-inhibitors, further demonstrating the functional uniqueness and superiority of the novel inhibitor. Together, we show that the multi-targeted inhibitors that can block VEGFR1, VEGFR2 and PDGFRß simultaneously suppress pathological angiogenesis more efficiently than monotherapy, and may therefore have promising therapeutic value for the treatment of neovascular diseases.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Ojo/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ojo/irrigación sanguínea , Ojo/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Transcriptoma/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
J Mol Cell Cardiol ; 121: 277-286, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30053526

RESUMEN

AIMS: Accumulating evidence indicates the presence of vascular stem/progenitor cells that may play a role in endothelial repair and lesion formation in the injured artery, in which c-kit+ stem/progenitor cells have been reported to differentiate into endothelial and smooth muscle cells in vitro and in ischemic tissue. In this study, we investigated whether and how endogenous c-kit+ stem/progenitor cells contribute to vascular injury and neointima formation in vivo. METHODS AND RESULTS: We created Kit-CreERxRosa26-RFP mice and performed genetic lineage tracing analysis of c-kit+ stem/progenitor cells in injury-induced neointima formation in vivo. We provide direct evidence that endogenous c-kit+ stem/progenitor cells minimally differentiate into endothelial or smooth muscle cells facilitating vascular repair, but predominantly generate monocytes/macrophages and granulocytes contributing to vascular immuno-inflammatory response to endothelial injury. Although c-kit+ cells reside in both bone marrow and vessel wall, bone marrow transplantation data indicate that bone marrow-derived c-kit+ cells are the main source for enhancing neointima formation. Furthermore, treatment of ACK2, a c-kit receptor antagonizer, attenuates neointimal hyperplasia after injury at least in part by depleting c-kit+ cells and their generated progeny. CONCLUSIONS: c-kit+ stem/progenitor cells are not a main source for endothelial regeneration and smooth muscle accumulation of the large artery injury, but a plausible interventional approach to reduce vascular immuno-inflammatory response and subsequently to ameliorate vascular lesions.


Asunto(s)
Arterias/crecimiento & desarrollo , Linaje de la Célula/genética , Proteínas Proto-Oncogénicas c-kit/genética , Células Madre/citología , Túnica Íntima/crecimiento & desarrollo , Animales , Arterias/lesiones , Diferenciación Celular/genética , Línea Celular , Movimiento Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Granulocitos/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Monocitos/metabolismo , Neointima/genética , Neointima/patología , Células Madre/metabolismo , Túnica Íntima/lesiones , Túnica Íntima/patología
14.
Arterioscler Thromb Vasc Biol ; 37(11): 2182-2194, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28912364

RESUMEN

OBJECTIVE: hnRNPA1 (heterogeneous nuclear ribonucleoprotein A1) plays a variety of roles in gene expression. However, little is known about the functional involvement of hnRNPA1 in vascular smooth muscle cell (VSMC) function and neointima hyperplasia. In this study, we have attempted to investigate the functional roles of hnRNPA1 in the contexts of VSMC function, injury-induced vessel remodeling, and human atherosclerotic lesions, as well as discern the molecular mechanisms involved. APPROACH AND RESULTS: hnRNPA1 expression levels were consistently modulated during VSMC phenotype switching and neointimal lesion formation induced by wire injury. Functional studies showed that VSMC-specific gene expression, proliferation, and migration were regulated by hnRNPA1. Our data show that hnRNPA1 exerts its effects on VSMC functions through modulation of IQGAP1 (IQ motif containing GTPase activating protein 1). Mechanistically, hnRNPA1 regulates IQGAP1 mRNA degradation through 2 mechanisms: upregulating microRNA-124 (miR-124) and binding to AU-rich element of IQGAP1 gene. Further evidence suggests that hnRNPA1 upregulates miR-124 by modulating miR-124 biogenesis and that IQGAP1 is the authentic target gene of miR-124. Importantly, ectopic overexpression of hnRNPA1 greatly reduced VSMC proliferation and inhibited neointima formation in wire-injured carotid arteries. Finally, lower expression levels of hnRNPA1 and miR-124, while higher expression levels of IQGAP1, were observed in human atherosclerotic lesions. CONCLUSIONS: Our data show that hnRNPA1 is a critical regulator of VSMC function and behavior in the context of neointima hyperplasia, and the hnRNPA1/miR-124/IQGAP1 regulatory axis represents a novel therapeutic target for the prevention of cardiovascular diseases.


Asunto(s)
Traumatismos de las Arterias Carótidas/metabolismo , Proliferación Celular , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima , Regiones no Traducidas 3' , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Hiperplasia , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
15.
Chemistry ; 23(7): 1490-1493, 2017 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-27935140

RESUMEN

A new super heat-resistant explosive, potassium 4-(5-amino-3-nitro-1H-1,2,4-triazol-1-yl)-3,5-dinitropyrazole (KCPT, 1), featuring a three-dimensional (3D) energetic metal-organic framework (MOF) was synthesized and fully characterized. The new 3D MOF was found to be extremely heat-resistant, having a high decomposition temperature of 323 °C. In addition, KCPT exhibits the best calculated detonation performance (vD =8457 m s-1 , p=32.5 GPa) among the reported super heat-resistant explosives or energetic potassium salts while retaining a suitable impact sensitivity of 7.5 J, which makes it one of the most promising heat-resistant explosives.

16.
J Biol Chem ; 290(31): 19158-72, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26092731

RESUMEN

Matrix metalloproteinase-8 (MMP8) has been shown to influence various cellular functions. As monocytes and macrophages (Mφ) express MMP8, we investigated if MMP8 played a role in macrophage differentiation and polarization. MMP8 expression was significantly increased during monocyte differentiation into Mφ. Monocyte-derived Mφ from MMP8-deficient mice expressed higher levels of M1-Mφ markers but lower levels of M2-Mφ markers than monocyte-derived Mφ from wild-type mice. Although Mφ from either MMP8-deficient or wild-type mice were inducible by interferon-γ into M1-Mφ, only wild-type Mφ but not MMP8-deficient Mφ could be induced into M2-Mφ by interleukin-4. However, MMP8-deficient Mφ exposed to conditioned culture media of wild-type Mφ developed a M2-Mφ phenotype. Compared with conditioned culture media of wild-type Mφ, conditioned culture media of MMP8-deficient Mφ contained a lower concentration of active transforming growth factor-ß (TGF-ß), an M2-Mφ inducer. Moreover, evidence also showed that the degradation of the TGF-ß sequester, fibromodulin, was modulated by MMP8. The data indicate a previously unknown role of MMP8 in M2-Mφ polarization by cleaving fibromodulin and therefore increasing the bioavailability of the M2-Mφ inducer TGF-ß.


Asunto(s)
Diferenciación Celular , Polaridad Celular , Macrófagos/fisiología , Metaloproteinasa 8 de la Matriz/genética , Animales , Línea Celular , Proteínas de la Matriz Extracelular/metabolismo , Fibromodulina , Regulación Enzimológica de la Expresión Génica , Interleucina-4/fisiología , Metaloproteinasa 8 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteoglicanos/metabolismo , Proteolisis , Transducción de Señal , Factor de Crecimiento Transformador beta1/fisiología , Regulación hacia Arriba
17.
Arterioscler Thromb Vasc Biol ; 35(4): 918-29, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25722434

RESUMEN

OBJECTIVE: In this study, we attempted to uncover the functional impact of microRNA-22 (miR-22) and its target gene in smooth muscle cell (SMC) differentiation and delineate the molecular mechanism involved. APPROACH AND RESULTS: miR-22 was found to be significantly upregulated during SMC differentiation from embryonic stem cells and adventitia stem/progenitor cells. Enforced expression of miR-22 by its mimic, while knockdown of miR-22 by its antagomiR, promotes or inhibits SMC differentiation from embryonic stem cells and adventitia stem/progenitor cells, respectively. Expectedly, miR-22 overexpression in stem cells promoted SMC differentiation in vivo. Methyl CpG-binding protein 2 (MECP2) was predicted as one of the top targets of miR-22. Interestingly, the gene expression levels of MECP2 were significantly decreased during SMC differentiation, and MECP2 was dramatically decreased in miR-22 overexpressing cells but significantly increased when miR-22 was knockdown in the differentiating stem cells. Importantly, luciferase assay showed that miR-22 substantially inhibited wild-type, but not mutant MECP2-3' untranslated region-luciferase activity. In addition, modulation of MECP2 expression levels affects multiple SMC-specific gene expression in differentiated embryonic stem cells. Mechanistically, our data showed that MECP2 could transcriptionally repress SMC gene expression through modulating various SMC transcription factors, as well as several proven SMC differentiation regulators. Evidence also revealed that enrichment of H3K9 trimethylation around the promoter regions of the SMC differentiation regulators genes were significantly increased by MECP2 overexpression. Finally, miR-22 was upregulated by platelet-derived growth factor-BB and transforming growth factor-ß through a transcriptional mechanism during SMC differentiation. CONCLUSIONS: miR-22 plays an important role in SMC differentiation, and epigenetic regulation through MECP2 is required for miR-22 mediated SMC differentiation.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , MicroARNs/metabolismo , Miocitos del Músculo Liso/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Regiones no Traducidas 3' , Animales , Becaplermina , Sitios de Unión , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/efectos de los fármacos , Epigénesis Genética , Regulación de la Expresión Génica , Histonas/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Metilación , Ratones , MicroARNs/genética , Mutación , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oligonucleótidos/metabolismo , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Interferencia de ARN , Elemento de Respuesta al Suero , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Factores de Tiempo , Transactivadores/genética , Transactivadores/metabolismo , Transcripción Genética , Transfección , Factor de Crecimiento Transformador beta/farmacología
19.
J Mol Cell Cardiol ; 89(Pt A): 75-86, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26493107

RESUMEN

AIMS: We have recently reported that microRNA-34a (miR-34a) regulates vascular smooth muscle cell (VSMC) differentiation from stem cells in vitro and in vivo. However, little is known about the functional involvements of miR-34a in VSMC functions and vessel injury-induced neointima formation. In the current study, we aimed to establish the causal role of miR-34a and its target genes in VSMC proliferation, migration and neointima lesion formation. METHODS AND RESULTS: Various pathological stimuli regulate miR-34a expression in VSMCs through a transcriptional mechanism, and the P53 binding site is required for miR-34a gene regulation by these stimuli. miR-34a over-expression in serum-starved VSMCs significantly inhibited VSMC proliferation and migration, while knockdown of miR-34a dramatically promoted VSMC proliferation and migration, respectively. Notch homolog 1 (Notch1), a well-reported regulator in VSMC functions and arterial remodeling, was predicted as one of the top targets of miR-34a by using several computational miRNA target prediction tools, and was negatively regulated by miR-34a in VSMCs. Luciferase assay showed miR-34a substantially repressed wild type Notch1-3'-UTR-luciferase activity in VSMCs, but not mutant Notch1-3'-UTR-luciferease reporter, confirming the Notch1 is the functional target of miR-34a in VSMCs. Data from co-transfection experiments also revealed that miR-34a inhibited VSMC proliferation and migration through modulating Notch gene expression levels. Importantly, the expression level of miR-34a was significantly down-regulated in injured arteries, and miR-34a perivascular over-expression significantly reduced Notch1 expression levels, decreased VSMC proliferation, and inhibited neointima formation in wire-injured femoral arteries. CONCLUSION: Our data have demonstrated that miR-34a is an important regulator in VSMC functions and neointima hyperplasia, suggesting its potential therapeutic application for vascular diseases.


Asunto(s)
Movimiento Celular , MicroARNs/metabolismo , Miocitos del Músculo Liso/patología , Neointima/genética , Neointima/patología , Animales , Apoptosis , Secuencia de Bases , Movimiento Celular/genética , Proliferación Celular , Arteria Femoral/lesiones , Arteria Femoral/patología , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , MicroARNs/genética , Datos de Secuencia Molecular , Músculo Liso Vascular/patología , Fenotipo , Receptores Notch/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Differentiation ; 85(1-2): 41-54, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23337518

RESUMEN

Embryonic stem cells (ES cells), the pluripotent derivatives of the inner cell mass from blastocysts, have the capacity for unlimited growth, self-renewal and differentiation toward all types of somatic cells. Angiotensin II (Ang II), the most important effector peptide of the renin-angiotensin system, is also an angiogenesis factor. However, the potential impact of Ang II on ES cell differentiation is still unknown. In the present study, we have successfully induced the differentiation of ES cells into smooth muscle cells (SMCs) on collagen IV. Interestingly, incubation of ES cells with Ang II further promoted SMC differentiation from ES cells, which was abolished by prior treatment with Ang II type 1 (AT1) receptor antagonist losartan, but not Ang II type 2 (AT2) receptor antagonist PD123319. Moreover, we found that, in parallel with SMC specific-marker induction, the expression levels of phosphoAkt and NF-Kappa B (NF-κB) p50 were up-regulated by Ang II. Importantly, addition of phosphoinositide-3 kinase (PI3K) inhibitor LY294002 led to a marked inhibition of Ang II induced SMC specific markers, phosphoAkt and NF-κB p50 expression. Furthermore, NF-κB inhibitor BAY11-7082 can inhibit Ang II induced expression of SMC specific markers. Thus, we demonstrate for the first time that Ang II plays a promotive role in the stage of ES cell differentiation to SMCs through AT1 receptor. We further confirmed that PI3K/Akt signaling pathway and NF-κB play key roles in this process.


Asunto(s)
Angiotensina II/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Miocitos del Músculo Liso/citología , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Línea Celular , Colágeno/farmacología , Células Madre Embrionarias/metabolismo , Imidazoles/farmacología , Losartán/farmacología , Ratones , Miocitos del Músculo Liso/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Nitrilos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Sulfonas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA