Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Lab Invest ; 101(10): 1318-1326, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34230646

RESUMEN

In the myocardial infarction microenvironment, the effect of macrophages on the function of bone marrow mesenchymal stem cells (BMSCs) is unclear. In this study, we investigated the role of hypoxia/serum deprivation (H/SD)-induced M1-type macrophage-derived exosomes on BMSC viability, migration, and apoptosis. We found that H/SD reduced BMSC viability and migration, increased BMSC apoptosis, and induced macrophage polarization toward the M1 phenotype. BMSCs were cultured by the supernatant of H/SD-induced THP-1 cells (M1-type macrophages) with or without exosome inhibitor treatment. The results show that BMSC apoptosis is increased in the H/SD-induced THP-1 cell supernatant group and is decreased by GM4869 treatment, indicating that M1-type macrophages induce BMSC apoptosis through exosomes. In addition, we confirm that miR-222 plays an important role in promoting BMSC apoptosis by targeting B-cell lymphoma (Bcl)-2. M1-type macrophage-derived exosomes significantly decrease BMSC viability and migration and increase BMSC apoptosis, and these effects are partly abolished by a miR-222 inhibitor. Our findings suggest that under H/SD conditions, exosomes derived from M1-type macrophages can induce BMSC apoptosis by delivering miR-222 to BMSCs.


Asunto(s)
Apoptosis/fisiología , Exosomas , Macrófagos , Células Madre Mesenquimatosas , MicroARNs/metabolismo , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
2.
J Cell Physiol ; 234(7): 10535-10546, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30456911

RESUMEN

Acute myocardial infarction (AMI), as a severe consequence of coronary atherosclerotic heart disease, always contributes to the loss of myocardial cells. Mounting evidence shows that annexin protects the myocardium from ischemic injury. In this study, we examine the inhibition of annexin A3 (ANXA3) on AMI through the phosphatidylinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. We selected rats to build an AMI model which was then assigned into different groups. The hemodynamic parameters after transfection were detected by using enzyme-linked immunosorbent assay. The effect of silencing of ANXA3 on inflammatory reaction and the PI3K/Akt signaling pathway was assessed. Rats transfected with ANXA3-short hairpin RNA had alleviated hemodynamics, inflammatory reaction, decreased infarct size, α-smooth muscle actin, Collagen I, and Collagen III as well as an increased vascular endothelial growth factor. Silencing ANAX3 would promote repair and healing of myocardial tissue by activation of the PI3K/Akt signaling pathway. Collectively, our study provides evidence that the downregulation of ANXA3 promotes the repair and healing of myocardial tissues by activating the PI3K/Akt signaling pathway.


Asunto(s)
Anexina A3/genética , Inflamación/genética , Infarto del Miocardio/genética , Miocardio/metabolismo , Animales , Anexina A3/antagonistas & inhibidores , Apoptosis/genética , Biología Computacional , Modelos Animales de Enfermedad , Silenciador del Gen , Humanos , Inflamación/patología , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Transducción de Señal/genética
3.
Mol Ther ; 26(7): 1685-1693, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29914756

RESUMEN

Treatment of myocardial infarction (MI) with bone marrow cells (BMCs) improves post-MI cardiac function in rodents. However, clinical trials of BMC therapy have been less effective. While most rodent experiments use young healthy donors, patients undergoing autologous cell therapy are older and post-MI. We previously demonstrated that BMCs from aged and post-MI donor mice are therapeutically impaired, and that donor MI induces inflammatory changes in BMC composition including reduced levels of B lymphocytes. Here, we hypothesized that B cell alterations in bone marrow account for the reduced therapeutic potential of post-MI and aged donor BMCs. Injection of BMCs from increasingly aged donor mice resulted in progressively poorer cardiac function and larger infarct size. Flow cytometry revealed fewer B cells in aged donor bone marrow. Therapeutic efficacy of young healthy donor BMCs was reduced by depletion of B cells. Implantation of intact or lysed B cells improved cardiac function, whereas intact or lysed T cells provided only minor benefit. We conclude that B cells play an important paracrine role in effective BMC therapy for MI. Reduction of bone marrow B cells because of age or MI may partially explain why clinical autologous cell therapy has not matched the success of rodent experiments.


Asunto(s)
Envejecimiento/fisiología , Linfocitos B/citología , Células de la Médula Ósea/citología , Médula Ósea/fisiología , Corazón/fisiología , Infarto del Miocardio/fisiopatología , Animales , Trasplante de Médula Ósea/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Citometría de Flujo/métodos , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Nicotine Tob Res ; 16(5): 584-90, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24302638

RESUMEN

INTRODUCTION: We sought to determine the effects of brief exposures to low concentrations of tobacco secondhand smoke (SHS) on arterial flow-mediated dilation (FMD, a nitric oxide-dependent measure of vascular endothelial function), in a controlled animal model never before exposed to smoke. In humans, SHS exposure for 30 min impairs FMD. It is important to gain a better understanding of the acute effects of exposure to SHS at low concentrations and for brief periods of time. METHODS: We measured changes in FMD in rats exposed to a range of real-world levels of SHS for durations of 30 min, 10 min, 1 min, and 4 breaths (roughly 15 s). RESULTS: We observed a dose-response relationship between SHS particle concentration over 30 min and post-exposure impairment of FMD, which was linear through the range typically encountered in smoky restaurants and then saturated at higher concentrations. One min of exposure to SHS at moderate concentrations was sufficient to impair FMD. CONCLUSIONS: Brief SHS exposure at real-world levels reversibly impairs FMD. Even 1 min of SHS exposure can cause reduction of endothelial function.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Dilatación Patológica , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Arteria Femoral/fisiopatología , Humanos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
6.
Drug Deliv ; 28(1): 285-292, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33501867

RESUMEN

In relieving local pains, lidocaine, one of ester-type local anesthetics, has been used. To develop the lidocaine membranes of enhanced local anesthetic effects, we have designed to establish the composition of wound dressings based on lidocaine chloride (LCH) (anesthetic drug)-loaded chitosan (CS)/polymyxin B sulfate (PMB). The LCH membranes (LCH-CS/PMB) was fabricated by the LCH oxide solutions within the CS/PMB matrix. The influences of different experimental limitations on CS/PMB membrane formations were examined. The double membrane particle sizes were evaluated by scanning electron microscopy (HR-SEM). Additionally, antibacterial efficacy was developed for gram-positive and negative microorganisms. Moreover, we examined in vivo healing of skin wounds formed in mouse models over 16 days. In contrast to the untreated wounds, rapid healing was perceived in the LCH-CS/PMB-treated wound with less damaging. These findings indicate that LCH-CS/PMB-based bandaging materials could be a potential innovative biomaterial for tissue repair and regeneration for wound healing applications in an animal model.


Asunto(s)
Anestésicos Locales/farmacología , Antibacterianos/farmacología , Quitosano , Regeneración Tisular Dirigida , Lidocaína/farmacología , Nanopartículas , Polimixina B/farmacología , Cicatrización de Heridas/efectos de los fármacos , Anestésicos Locales/administración & dosificación , Animales , Antibacterianos/administración & dosificación , Vendajes , Materiales Biocompatibles , Técnicas In Vitro , Lidocaína/administración & dosificación , Pruebas Mecánicas , Ratones , Polimixina B/administración & dosificación , Andamios del Tejido
7.
Exp Ther Med ; 22(5): 1272, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34594409

RESUMEN

A single injection of low-dose human umbilical cord-derived mesenchymal stem cells (UC-MSCs) has been previously demonstrated to relieve synovitis and bone erosion in animal models of arthritis, but whether frequent injections of high-dose UC-MSCs relieve arthritis and inhibit loss of muscle mass has remained elusive. In the present study, DBA/1 mice were randomly divided into three groups: Normal (wild-type mice; n=11), collagen-induced arthritis (CIA; n=12) and CIA treated with UC-MSCs (n=11; 5x106 UC-MSCs per week for 3 weeks). Arthritis and skeletal muscle cachexia were evaluated until the end of the experiment on day 84. It was indicated that both the CIA and UC-MSC groups had lower body weights compared with the normal mice. Clinical arthritis scores, hind ankle diameters, synovitis and bone erosion progressively increased and were similar between the CIA and UC-MSC groups. Although there was no difference in food intake among the three groups, the normalized food intake of normal group was significantly higher than CIA group and UC-MSC group from day 42 onwards; there was no significance on day 77 but this could be neglected. Furthermore, gastrocnemius muscle weight in the UC-MSC group was significantly reduced compared with that in the CIA and normal groups. The UC-MSC group had higher levels of proinflammatory cytokines, such as TNF-α, IL-6 and IL-1ß than those in the CIA group. However, the other cytokines assessed and the fibrosis indices in the CIA and UC-MSC groups were not different from those in the control group and there was no inflammatory cell infiltration. Thus, frequent injections of high-dose UC-MSCs slightly aggravated synovitis and muscle cachexia in the murine CIA model and should therefore be avoided in the treatment of arthritis.

8.
Diabetol Metab Syndr ; 13(1): 15, 2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514420

RESUMEN

BACKGROUND: Hypertriglyceridemia (HTG) is considered an independent risk factor for major adverse cardiovascular events (MACE). METHODS: This study analyzed the effects of various agents on MACE risk reduction in HTG (serum triglyceride ≥ 150 mg/dl) populations by performing a network meta-analysis. We performed a frequentist network meta-analysis to conduct direct and indirect comparisons of interventions. PubMed, EMBASE, and the Cochrane library were searched for trials until Jul 6, 2020. Randomized controlled trials that reported MACE associated with agents in entire HTG populations or in subgroups were included. The primary outcome was MACE. RESULTS: Of the 2005 articles screened, 21 trials including 56,471 patients were included in the analysis. The network meta-analysis results for MACE risk based on frequency data showed that eicosapentaenoic acid (EPA) (OR: 1.32; 95% CI 1.19-1.46), gemfibrozil (OR: 1.53; 95% CI 1.20-1.95), niacin plus clofibrate (OR: 2.00; 95% CI 1.23-3.25), pravastatin (OR: 1.32; 95% CI 1.15-1.52), simvastatin (OR: 2.38; 95% CI 1.55-3.66), and atorvastatin (OR: 0.55; 95% CI 0.37-0.82) significantly reduced the risk of MACE compared to the control conditions. In the subgroup analysis of HTG patients with triglycerides ≥ 200 mg/dL, bezafibrate (OR: 0.56; 95% CI 0.33-0.94), EPA (OR: 0.72; 95% CI 0.62-0.82), and pravastatin (OR: 1.33; 95% CI 1.01-1.75) significantly reduced the MACE risk. CONCLUSIONS: Simvastatin had a clear advantage in reducing the risk of MACE in the entire HTG population analyzed in this meta-analysis. EPA, but not omega-3 fatty acid, was considered an effective HTG intervention. Among fibrates, gemfibrozil was most effective, though bezafibrate may significantly reduce the risk of MACE in populations with triglyceride levels of 200-300 mg/dL. Trial registration retrospectively registered in PROSPERO (CRD42020213705).

9.
Front Mol Biosci ; 7: 622540, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33644114

RESUMEN

Cardiovascular disease is the main disease that affects human life span. In recent years, the disease has been increasingly addressed at the molecular levels, for example, pseudogenes are now known to be involved in the pathogenesis and development of cardiovascular diseases. Pseudogenes are non-coding homologs of protein-coding genes and were once called "junk gene." Since they are highly homologous to their functional parental genes, it is somewhat difficult to distinguish them. With the development of sequencing technology and bioinformatics, pseudogenes have become readily identifiable. Recent studies indicate that pseudogenes are closely related to cardiovascular diseases. This review provides an overview of pseudogenes and their roles in the pathogenesis of cardiovascular diseases. This new knowledge adds to our understanding of cardiovascular disease at the molecular level and will help develop new biomarkers and therapeutic approaches designed to prevent and treat the disease.

10.
Aging (Albany NY) ; 12(24): 25120-25137, 2020 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-33342766

RESUMEN

Among all diabetes mellitus-associated cardiovascular diseases, morbidity of diabetic myocardium with ischemia reperfusion injury (D-IRI) is increasing year by year. We aimed to discover a therapeutic biomarker and investigate its mechanism in D-IRI. High-fat diet and streptozotocin-induced diabetes rats were operated with IRI or sham. Recombined lentiviral vector encoding Apelin was injected into D-IRI rat via tail vein. Cardiac function, infarct size, cellular death and oxidative stress were major outcome measures. Cardiomyocyte ischemia reperfusion injury was more serious in D-IRI rats than in non-diabetes ischemia reperfusion injury (ND-IRI) rats. The secretion of NTproBNP was increased in D-IRI compared with ND-IRI. Bcl-2 expression was decreased, and Bax and cleaved caspase-3 expression was increased in D-IRI rats compared with ND-IRI rats, which were reversed after treatment with Apelin. Apelin-upregulation improved cardiomyocyte ischemia reperfusion injury and decreased NT-proBNP levels in D-IRI rats. Apelin overexpression enhanced PI3K and eNOS levels while reduced those of p38-MAPK and iNOS in D-IRI rats. Apelin overexpression protected against D-IRI through inhibiting apoptosis and oxidative stress via PI3K and p38MAPK signaling pathways in D-IRI rats. These findings provide critical new insight into understanding of Apelin's cardio-protective effects, which may become a novel therapeutic target for the diabetic IRI patients.


Asunto(s)
Apelina/genética , Apoptosis/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Daño por Reperfusión Miocárdica/genética , Estrés Oxidativo/genética , Animales , Apelina/metabolismo , Caspasa 3/metabolismo , Muerte Celular/genética , Terapia Genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Péptido Natriurético Encefálico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fragmentos de Péptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Proteína X Asociada a bcl-2/metabolismo
11.
Biomed Pharmacother ; 108: 1809-1815, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30372886

RESUMEN

Oxidized LDL (ox-LDL) is the key risk factor of developing atherosclerosis. In endothelial cells, exposure of ox-LDL causes endothelial dysfunction and injury. In this study, we investigated the role of receptor interacting protein 1 (RIP1), one of the kinases involved in apoptosis and necroptosis mediated by the death receptor tumor necrosis factor receptor (TNFR), in endothelial dysfunction. We show that RIP1 is responsively induced in human umbilical vein endothelial cells (HUVECs) upon ox-LDL treatment. Blockage of RIP1 activity by its antagonist, necrostatin-1, ameliorates ox-LDL-induced nitric oxide (NO) reduction and induction of vascular adhesion molecules, including vascular cell adhesion molecule 1 (VCAM-1) and E-selectin, as well as adhesion of immune cells to endothelial cells. Mechanistically, we show that inactivation of RIP1 by necrostatin-1 suppressed nuclear factor κB (NF-κB) cascade signals, including activation of IKKα, nuclear factor kappa B inhibitor protein α (IκBα), accumulation of nuclear p65 and NF-κB promoter activity. Silencing of RIP1 largely attenuates the action of ox-LDL on the expression of vascular adhesion molecules and adhesion of immune cells to endothelial cells. Collectively, our data indicate that the response of RIP1 to ox-LDL and its activation are required for ox-LDL-induced endothelial injury.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/patología , Imidazoles/farmacología , Indoles/farmacología , Lipoproteínas LDL/toxicidad , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Adhesión Celular , Selectina E/metabolismo , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Inflamación/patología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Células U937 , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
J Am Heart Assoc ; 5(1)2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26738788

RESUMEN

BACKGROUND: Circulating angiogenic cells (CACs) are peripheral blood cells whose functional capacity inversely correlates with cardiovascular risk and that have therapeutic benefits in animal models of cardiovascular disease. However, donor age and disease state influence the efficacy of autologous cell therapy. We sought to determine whether age or coronary artery disease (CAD) impairs the therapeutic potential of CACs for myocardial infarction (MI) and whether the use of ex vivo gene therapy to overexpress endothelial nitric oxide (NO) synthase (eNOS) overcomes these defects. METHODS AND RESULTS: We recruited 40 volunteers varying by sex, age (< or ≥45 years), and CAD and subjected their CACs to well-established functional tests. Age and CAD were associated with reduced CAC intrinsic migration (but not specific response to vascular endothelial growth factor, adherence of CACs to endothelial tubes, eNOS mRNA and protein levels, and NO production. To determine how CAC function influences therapeutic potential, we injected the 2 most functional and the 2 least functional CAC isolates into mouse hearts post MI. The high-function isolates substantially improved cardiac function, whereas the low-function isolates led to cardiac function only slightly better than vehicle control. Transduction of the worst isolate with eNOS cDNA adenovirus increased NO production, migration, and cardiac function of post-MI mice implanted with the CACs. Transduction of the best isolate with eNOS small interfering RNA adenovirus reduced all of these capabilities. CONCLUSIONS: Age and CAD impair multiple functions of CACs and limit therapeutic potential for the treatment of MI. eNOS gene therapy in CACs from older donors or those with CAD has the potential to improve autologous cell therapy outcomes.


Asunto(s)
Enfermedad de la Arteria Coronaria/enzimología , Células Progenitoras Endoteliales/enzimología , Células Progenitoras Endoteliales/trasplante , Infarto del Miocardio/cirugía , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Trasplante de Células Madre/métodos , Adulto , Anciano , Animales , Estudios de Casos y Controles , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Enfermedad de la Arteria Coronaria/diagnóstico , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones SCID , Persona de Mediana Edad , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Óxido Nítrico Sintasa de Tipo III/genética , Fenotipo , Interferencia de ARN , Recuperación de la Función , Regeneración , Transducción de Señal , Factores de Tiempo , Transducción Genética , Transfección
13.
Mol Med Rep ; 12(4): 5911-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239104

RESUMEN

The growth of the heart during mammalian embryonic development is primarily dependent on an increase in the number of cardiomyocytes (CM). However, shortly following birth, CMs cease proliferating and further growth of the myocardium is achieved via hypertrophic expansion of the existing CM population. The cyclin-dependent kinase inhibitor 2A (Cdkn2a) locus encodes overlapping genes for two tumor suppressor proteins, p16INK4a and p19 alternative reading frame (ARF). To determine whether decreased Cdkn2a gene expression results in improved cardiac regeneration in vitro and in vivo following cardiac injury, the proliferation of CMs isolated from Cdkn2a knockout (KO) and wild­type (WT) mice in vitro and in vivo were evaluated following generation of ischemia reperfusion (IR) injury. The KO mice demonstrated enhanced CM proliferation not only in vitro, but also in vivo. Furthermore, heart function was improved and scar size was decreased in the KO mice compared with that of the WT mice. The results also indicated that microRNA (miR)­1 and miR­195 expression levels associated with cell proliferation were reduced following IR injury in KO mice compared with those of WT mice. These results suggested that the inactivation of INK4a and ARF stimulated CM proliferation and promoted cardiac repair.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Daño por Reperfusión Miocárdica/genética , Cicatrización de Heridas/genética , Animales , Proliferación Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Electrocardiografía , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Ratones , Ratones Noqueados , MicroARNs/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Función Ventricular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA