Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
Life Sci ; 296: 120317, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35026214

RESUMEN

AIMS: Angelol-A (Ang-A), a kind of coumarins, is isolated from the roots of Angelica pubescens f. biserrata. However, AA exerts antitumor effects and molecular mechanism on cervical cancer cells is unknown. MAIN METHODS: Cell viability was determined using the MTT assay, and the cell cycle phase was assessed by PI staining with flow cytometry. Ang-A-treated cells with/without Antago-miR-29a-3p (miR-29a-3p inhibitor) or U0126 (MEK inhibitor) were assessed for the expression of miR-29a-3p, in vitro migration/invasion, and angiogenesis using qRT-PCR, a chemotaxis assay, and tube formation assay, respectively. The expression of mitogen-activated protein kinases/MMP2/MMP9/VEGFA was determined by western blot analysis with applicable antibodies. KEY FINDINGS: Ang-A significantly inhibited MMP2 and VEGFA expression, cell migration, and invasive motility in human cervical cancer cells. Conditioned medium inhibited tube formation in HUVECs. Ang-A principally inhibited invasive motility and angiogenesis by upregulating the expression of miR-29a-3p that targets the VEGFA-3' UTR. The role of miR-29a-3p was confirmed using Antago-miR-29a-3p, which reversed the Ang-A-inhibited expression of MMP2 and VEGFA, invasive motility, and angiogenesis in human cervical cancer cells. The ERK pathway was implicated in mediating the metastatic and angiogenic action of Ang-A. Combined treatment with Ang-A treated and U0126 exerted a synergistic inhibitory effect on the expression of MMP2 and VEGFA and the metastatic and angiogenic properties of human cervical cancer cells. SIGNIFICANCE: These findings are the first to indicate that in human cervical cancer cells, Ang-A exerts anti-metastatic and anti-angiogenic effects via targeting the miR-29a-3p/MMP2/VEGFA axis, mediated through the ERK pathway.


Asunto(s)
Inhibidores de la Angiogénesis , Antineoplásicos Fitogénicos , Neoplasias del Cuello Uterino , Femenino , Humanos , Angelica/química , Inhibidores de la Angiogénesis/farmacología , Antagomirs/genética , Antagomirs/farmacología , Antineoplásicos Fitogénicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fosforilación/efectos de los fármacos , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Cardiovasc Res ; 118(2): 556-572, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33483746

RESUMEN

AIMS: Myocarditis is associated with formidable symptoms and increased risk of adverse outcomes. Current approaches mostly rely on symptomatic treatments, warranting novel concepts for clinical practice. The aim of this study was to investigate the microRNA (miRNA) expression profile of Balb/c mice with experimental autoimmune myocarditis (EAM), choose a representative miRNA to antagonize after review of available literature and test its effects on myocardial inflammation in vitro and in vivo. METHODS AND RESULTS: Phase 1: EAM was induced in 12 male Balb/c mice, 10 animals served as controls. After sacrifice, next-generation sequencing (NGS) of the miRNA expression profile was performed. Based on these results, H9C2 cells and human ventricular cardiac fibroblasts exposed to lipopolysaccharide (LPS) were treated with the selected candidate antagomiR-21a-5p. Phase 2: EAM was induced in 48 animals. Thereof, 24 animals were either treated with antagomiR-21a-5p or negative control oligonucleotide in a nanoparticle formulation. Transthoracic echocardiography (TTE) was performed on Days 0, 7, 14, and 21. Histopathological examination was performed after sacrifice. Phase 1: EAM resulted in a significant up-regulation of 27 miRNAs, including miR-21a-5p (log2FC: 2.23, adj. P = 0.0026). Transfection with antagomiR-21a-5p resulted in a significant reduction of TNFα, IL-6, and collagen I in vitro. Phase 2: Treatment with antagomiR-21a-5p, formulated in polymeric nanoparticles for systemic injection, significantly attenuated myocardial inflammation (P = 0.001) and fibrosis (P = 0.013), as well as myocardial 'hypertrophy' on TTE. CONCLUSIONS: Silencing of miR-21a-5p results in a significant reduction of the expression of pro-inflammatory cytokines in vitro, as well as a significant attenuation of inflammation, fibrosis and echocardiographic effects of EAM in vivo.


Asunto(s)
Antagomirs/administración & dosificación , Enfermedades Autoinmunes/terapia , Ecocardiografía , MicroARNs/metabolismo , Miocarditis/terapia , Miocitos Cardíacos/metabolismo , Animales , Antagomirs/genética , Antagomirs/metabolismo , Enfermedades Autoinmunes/diagnóstico por imagen , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica , Humanos , Lipopolisacáridos , Masculino , Ratones Endogámicos BALB C , MicroARNs/genética , Miocarditis/diagnóstico por imagen , Miocarditis/genética , Miocarditis/metabolismo , Miocitos Cardíacos/patología , Ratas , Transcriptoma , Transfección , Función Ventricular Izquierda , Remodelación Ventricular
3.
Nucleic Acids Res ; 50(2): 617-634, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34967419

RESUMEN

MicroRNAs (miRNAs) are short endogenously expressed RNAs that have the potential to regulate the expression of any RNA. This potential has led to the publication of several thousand papers each year connecting miRNAs to many different genes and human diseases. By contrast, relatively few papers appear that investigate the molecular mechanism used by miRNAs. There is a disconnect between rigorous understanding of mechanism and the extraordinary diversity of reported roles for miRNAs. Consequences of this disconnect include confusion about the assumptions underlying the basic science of human miRNAs and slow development of therapeutics that target miRNAs. Here, we present an overview of investigations into miRNAs and their impact on gene expression. Progress in our understanding of miRNAs would be aided by a greater focus on the mechanism of miRNAs and a higher burden of evidence on researchers who seek to link expression of a particular miRNA to a biological phenotype.


Asunto(s)
Silenciador del Gen , MicroARNs/genética , Interferencia de ARN , Animales , Antagomirs/síntesis química , Antagomirs/genética , Antagomirs/uso terapéutico , Emparejamiento Base , Secuencia de Bases , Estudios Clínicos como Asunto , Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos , Variación Genética , Humanos , MicroARNs/síntesis química , MicroARNs/uso terapéutico , Relación Estructura-Actividad , Resultado del Tratamiento
4.
PLoS Genet ; 17(12): e1009934, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34914716

RESUMEN

MicroRNAs (miRNA) are short non-coding RNAs widely implicated in gene regulation. Most metazoan miRNAs utilize the RNase III enzymes Drosha and Dicer for biogenesis. One notable exception is the RNA polymerase II transcription start sites (TSS) miRNAs whose biogenesis does not require Drosha. The functional importance of the TSS-miRNA biogenesis is uncertain. To better understand the function of TSS-miRNAs, we applied a modified Crosslinking, Ligation, and Sequencing of Hybrids on Argonaute (AGO-qCLASH) to identify the targets for TSS-miRNAs in HCT116 colorectal cancer cells with or without DROSHA knockout. We observed that miR-320a hybrids dominate in TSS-miRNA hybrids identified by AGO-qCLASH. Targets for miR-320a are enriched for the eIF2 signaling pathway, a downstream component of the unfolded protein response. Consistently, in miR-320a mimic- and antagomir- transfected cells, differentially expressed gene products are associated with eIF2 signaling. Within the AGO-qCLASH data, we identified the endoplasmic reticulum (ER) chaperone calnexin as a direct miR-320a down-regulated target, thus connecting miR-320a to the unfolded protein response. During ER stress, but not amino acid deprivation, miR-320a up-regulates ATF4, a critical transcription factor for resolving ER stress. In summary, our study investigates the targetome of the TSS-miRNAs in colorectal cancer cells and establishes miR-320a as a regulator of unfolded protein response.


Asunto(s)
Factor de Transcripción Activador 4/genética , Neoplasias Colorrectales/genética , MicroARNs/genética , Ribonucleasa III/genética , Antagomirs/genética , Proteínas Argonautas/genética , Calnexina/genética , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , ARN Helicasas DEAD-box/genética , Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/genética , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Transducción de Señal/genética , Sitio de Iniciación de la Transcripción
5.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830336

RESUMEN

Cleft lip with or without cleft palate (CL/P) is one of the most common congenital birth defects. This study aims to identify novel pathogenic microRNAs associated with cleft palate (CP). Through data analyses of miRNA-sequencing for developing palatal shelves of C57BL/6J mice, we found that miR-449a-3p, miR-449a-5p, miR-449b, miR-449c-3p, and miR-449c-5p were significantly upregulated, and that miR-19a-3p, miR-130a-3p, miR-301a-3p, and miR-486b-5p were significantly downregulated, at embryonic day E14.5 compared to E13.5. Among them, overexpression of the miR-449 family (miR-449a-3p, miR-449a-5p, miR-449b, miR-449c-3p, and miR-449c-5p) and miR-486b-5p resulted in reduced cell proliferation in primary mouse embryonic palatal mesenchymal (MEPM) cells and mouse cranial neural crest cell line O9-1. On the other hand, inhibitors of miR-130a-3p and miR-301a-3p significantly reduced cell proliferation in MEPM and O9-1 cells. Notably, we found that treatment with dexamethasone, a glucocorticoid known to induce CP in mice, suppressed miR-130a-3p expression in both MEPM and O9-1 cells. Moreover, a miR-130a-3p mimic could ameliorate the cell proliferation defect induced by dexamethasone through normalization of Slc24a2 expression. Taken together, our results suggest that miR-130-3p plays a crucial role in dexamethasone-induced CP in mice.


Asunto(s)
Fisura del Paladar/genética , Dexametasona/farmacología , Glucocorticoides/farmacología , MicroARNs/genética , Células Madre Embrionarias de Ratones/efectos de los fármacos , Animales , Antagomirs/genética , Antagomirs/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Fisura del Paladar/inducido químicamente , Fisura del Paladar/metabolismo , Fisura del Paladar/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/clasificación , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Cresta Neural/citología , Cresta Neural/efectos de los fármacos , Cresta Neural/metabolismo , Cultivo Primario de Células , Transducción de Señal , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo
6.
Cells ; 10(7)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34360007

RESUMEN

Since mitochondria are suggested to be important regulators in maintaining cartilage homeostasis, turnover of mitochondria through mitochondrial biogenesis and mitochondrial degradation may play an important role in the pathogenesis of osteoarthritis (OA). Here, we found that mitochondrial dysfunction is closely associated with OA pathogenesis and identified the peroxisome proliferator-activated receptor-gamma co-activator 1-alpha (PGC1α) as a potent regulator. The expression level of PGC1α was significantly decreased under OA conditions, and knockdown of PGC1α dramatically elevated the cartilage degradation by upregulating cartilage degrading enzymes and apoptotic cell death. Interestingly, the knockdown of PGC1α activated the parkin RBR E3 ubiquitin protein ligase (PRKN)-independent selective mitochondria autophagy (mitophagy) pathway through the upregulation of BCL2 and adenovirus E1B 19-kDa-interacting protein 3 (BNIP3). The overexpression of BNIP3 stimulated mitophagy and cartilage degradation by upregulating cartilage-degrading enzymes and chondrocyte death. We identified microRNA (miR)-126-5p as an upstream regulator for PGC1α and confirmed the direct binding between miR-126-5p and 3' untranslated region (UTR) of PGC1α. An in vivo OA mouse model induced by the destabilization of medial meniscus (DMM) surgery, and the delivery of antago-miR-126 via intra-articular injection significantly decreased cartilage degradation. In sum, the loss of PGC1α in chondrocytes due to upregulation of miR-126-5p during OA pathogenesis resulted in the activation of PRKN-independent mitophagy through the upregulation of BNIP3 and stimulated cartilage degradation and apoptotic death of chondrocytes. Therefore, the regulation of PGC1α:BNIP3 mitophagy axis could be of therapeutic benefit to cartilage-degrading diseases.


Asunto(s)
Cartílago Articular/metabolismo , Proteínas de la Membrana/genética , MicroARNs/genética , Proteínas Mitocondriales/genética , Mitofagia/genética , Osteoartritis/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Animales , Antagomirs/genética , Antagomirs/metabolismo , Artroplastia de Reemplazo de Rodilla/métodos , Secuencia de Bases , Cartílago Articular/patología , Condrocitos/metabolismo , Condrocitos/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Meniscos Tibiales/metabolismo , Meniscos Tibiales/patología , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/metabolismo , Osteoartritis/metabolismo , Osteoartritis/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/antagonistas & inhibidores , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
7.
J Mol Cell Cardiol ; 160: 128-141, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34329686

RESUMEN

microRNA-378a (miR-378a) is one of the most highly expressed microRNAs in the heart. However, its role in the human cardiac tissue has not been fully understood. It was observed that miR-378a protects cardiomyocytes from hypertrophic growth by regulation of IGF1R and the expression of downstream kinases. Increased levels of miR-378a were reported in the serum of Duchenne muscular dystrophy (DMD) patients and female carriers of DMD gene-associated mutations with developed cardiomyopathy. In order to shed more light on the role of miR-378a in human cardiomyocytes and its potential involvement in DMD-related cardiomyopathy, we generated two human induced pluripotent stem cell (hiPSC) models; one with deletion of miR-378a and the second one with deletion of DMD exon 50 leading to the DMD phenotype. Our results indicate that lack of miR-378a does not influence the pluripotency of hiPSC and their ability to differentiate into cardiomyocytes (hiPSC-CM). miR-378a-deficient hiPSC-CM exhibited, however, significantly bigger size compared to the isogenic control cells, indicating the role of this miRNA in the hypertrophic growth of human cardiomyocytes. In accordance, the level of NFATc3, phosphoAKT, phosphoERK and ERK was higher in these cells compared to the control counterparts. A similar effect was achieved by silencing miR-378a with antagomirs. Of note, the percentage of cells with nuclear localization of NFATc3 was higher in miR-378a-deficient hiPSC-CM. Analysis of electrophysiological properties and Ca2+ oscillations revealed the decrease in the spike slope velocity and lower frequency of calcium spikes in miR-378a-deficient hiPSC-CM. Interestingly, the level of miR-378a increased gradually during cardiac differentiation of hiPSC. Of note, it was low until day 15 in differentiating DMD-deficient hiPSC-CM and then rose to a similar level as in the isogenic control counterparts. In summary, our findings confirmed the utility of hiPSC-based models for deciphering the role of miR-378a in the control and diseased human cardiomyocytes.


Asunto(s)
Señalización del Calcio/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Antagomirs/genética , Calcio/metabolismo , Cardiomiopatías/complicaciones , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Diferenciación Celular/genética , Aumento de la Célula , Tamaño de la Célula , Distrofina/genética , Distrofina/metabolismo , Exones , Eliminación de Gen , Células HEK293 , Humanos , MicroARNs/genética , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/genética , Receptor IGF Tipo 1/metabolismo , Transfección
8.
Acta Biochim Pol ; 68(2): 201-206, 2021 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-33966370

RESUMEN

Sepsis is a systemic inflammatory response syndrome caused by various pathogenic microorganisms or toxins. Lung damage is one of the causes of death in patients with sepsis. This study aimed to investigate the role of miR-19a-3p and its regulation mechanism in sepsis-induced lung injury. MH-S cells were treated with lipopolysaccharide (LPS) to establish sepsis-induced lung injury cell model. C57BL/6 mice were injected with miR-19a-3p antagomiR and LPS to construct animal model. LPS-treated and control cells were transfected with miR-19a-3p mimic, miR-19a-3p inhibitor or USP13 expression vector . The expression levels of miR-19a-3p and USP13 were examined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. The concentration of inflammatory cytokines was measured with enzyme-linked immunosorbent assay (ELISA). The relationship of miR-19a-3p and USP13 was validated using dual-luciferase reporter assay. The lung damage was assessed with hematoxylin-eosin staining (HE). The results showed that LPS treatment increased the concentration of TNF-α, IL-6 and IL-1ß in MH-S cells. In LPS treated MH-S cells, the level of miR-19a-3p gradually increased over time. Both miR-19a-3p knockdown and USP13 overexpression in MH-S cells inhibited the LPS-induced production of TNF-α, IL-6 and IL-1ß. Moreover, miR-19a-3p negatively regulated the expression of USP13 in MH-S cells. Furthermore, miR-19a-3p inhibitor suppressed lung damage in sepsis model mice. In conclusion, miR-19a-3p knockdown could alleviate sepsis-induced lung injury through enhancing USP13 expression.


Asunto(s)
Lesión Pulmonar/metabolismo , MicroARNs/metabolismo , Sepsis/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Antagomirs/genética , Antagomirs/metabolismo , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen/métodos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/efectos adversos , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Sepsis/complicaciones , Sepsis/patología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33804856

RESUMEN

Cancer is one of the leading causes of death worldwide. Conventional therapies, including surgery, radiation, and chemotherapy have achieved increased survival rates for many types of cancer over the past decades. However, cancer recurrence and/or metastasis to distant organs remain major challenges, resulting in a large, unmet clinical need. Oligonucleotide therapeutics, which include antisense oligonucleotides, small interfering RNAs, and aptamers, show promising clinical outcomes for disease indications such as Duchenne muscular dystrophy, familial amyloid neuropathies, and macular degeneration. While no approved oligonucleotide drug currently exists for any type of cancer, results obtained in preclinical studies and clinical trials are encouraging. Here, we provide an overview of recent developments in the field of oligonucleotide therapeutics in oncology, review current clinical trials, and discuss associated challenges.


Asunto(s)
Antagomirs/genética , Neoplasias/terapia , Oligonucleótidos Antisentido/genética , Tratamiento con ARN de Interferencia/métodos , Animales , Aptámeros de Nucleótidos/genética , Ensayos Clínicos como Asunto , Humanos , ARN Interferente Pequeño/genética
10.
Exp Biol Med (Maywood) ; 246(11): 1274-1286, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33715531

RESUMEN

This study aims to examine whether miR-31 promotes endogenous NSC proliferation and be used for spinal cord injury management. In the present study, the morpholino knockdown of miR-31 induced abnormal neuronal apoptosis in zebrafish, resulting in impaired development of the tail. miR-31 agomir transfection in NSCs increased Nestin expression and decreased ChAT and GFAP expression levels. miR-31 induced the proliferation of mouse NSCs by upregulating the Notch signaling pathway, and more NSCs entered G1; Notch was inhibited by miR-31 inactivation. Injection of a miR-31 agomir into mouse models of spinal cord injury could effectively restore motor functions after spinal cord injury, which was achieved by promoting the proliferation of endogenous NSCs. After the injection of a miR-31 agomir in spinal cord injury mice, the expression of Nestin and GFAP increased, while GFAP expression decreased. In conclusion, the zebrafish experiments prove that a lack of miR-31 will block nervous system development. In spinal cord injury mouse models, miR-31 overexpression might promote spinal cord injury repair.


Asunto(s)
MicroARNs/genética , Neuronas Motoras/fisiología , Células-Madre Neurales/citología , Traumatismos de la Médula Espinal/genética , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Antagomirs/genética , Apoptosis/genética , Proliferación Celular , Modelos Animales de Enfermedad , Embrión no Mamífero , Femenino , Masculino , Ratones Endogámicos ICR , Morfolinos , Neuronas Motoras/patología , Células-Madre Neurales/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Transfección , Pez Cebra/genética
11.
Mol Ther ; 29(6): 2041-2052, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-33609732

RESUMEN

Oligonucleotide therapies offer precision treatments for a variety of neurological diseases, including epilepsy, but their deployment is hampered by the blood-brain barrier (BBB). Previous studies showed that intracerebroventricular injection of an antisense oligonucleotide (antagomir) targeting microRNA-134 (Ant-134) reduced evoked and spontaneous seizures in animal models of epilepsy. In this study, we used assays of serum protein and tracer extravasation to determine that BBB disruption occurring after status epilepticus in mice was sufficient to permit passage of systemically injected Ant-134 into the brain parenchyma. Intraperitoneal and intravenous injection of Ant-134 reached the hippocampus and blocked seizure-induced upregulation of miR-134. A single intraperitoneal injection of Ant-134 at 2 h after status epilepticus in mice resulted in potent suppression of spontaneous recurrent seizures, reaching a 99.5% reduction during recordings at 3 months. The duration of spontaneous seizures, when they occurred, was also reduced in Ant-134-treated mice. In vivo knockdown of LIM kinase-1 (Limk-1) increased seizure frequency in Ant-134-treated mice, implicating de-repression of Limk-1 in the antagomir mechanism. These studies indicate that systemic delivery of Ant-134 reaches the brain and produces long-lasting seizure-suppressive effects after systemic injection in mice when timed with BBB disruption and may be a clinically viable approach for this and other disease-modifying microRNA therapies.


Asunto(s)
Antagomirs/genética , Barrera Hematoencefálica/metabolismo , Epilepsia/genética , Epilepsia/terapia , Animales , Antagomirs/administración & dosificación , Barrera Hematoencefálica/patología , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Silenciador del Gen , Técnicas de Transferencia de Gen , Predisposición Genética a la Enfermedad , Terapia Genética , Ratones , MicroARNs/genética , Interferencia de ARN , Resultado del Tratamiento
12.
Mol Med Rep ; 23(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33576461

RESUMEN

Cognitive impairment is one of the primary features of vascular dementia (VD). However, the specific mechanism underlying the regulation of cognition function in VD is not completely understood. The present study aimed to explore the effects of microRNA (miR)­150 on VD. To determine the effects of miR­150 on cognitive function and hippocampal neurons in VD model rats, rats were subjected to intracerebroventricular injections of miR­150 antagomiR. The Morris water maze test results demonstrated that spatial learning ability was impaired in VD model rats compared with control rats. Moreover, compared with antagomiR negative control (NC), miR­150 antagomiR alleviated cognitive impairment and enhanced memory ability in VD model rats. The triphenyltetrazolium chloride, Nissl staining and immunohistochemistry results further demonstrated that miR­150 knockdown improved the activity of hippocampal neurons in VD model rats compared with the antagomiR NC group. To validate the role of miR­150 in neurons in vitro, the PC12 cell line was used. The flow cytometry and Hoechst 33342/PI double staining results indicated that miR­150 overexpression significantly increased cell apoptosis compared with the mimic NC group. Moreover, the dual­luciferase reporter gene assay results indicated that miR­150 targeted HOXA1 and negatively regulated HOXA1 expression. Therefore, the present study indicated that miR­150 knockdown ameliorated VD symptoms by upregulating HOXA1 expression in vivo and in vitro.


Asunto(s)
Apoptosis/genética , Demencia Vascular/genética , Modelos Animales de Enfermedad , Hipocampo/metabolismo , MicroARNs/genética , Neuronas/metabolismo , Animales , Antagomirs/administración & dosificación , Antagomirs/genética , Cognición/fisiología , Regulación de la Expresión Génica , Hipocampo/citología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Células PC12 , Ratas , Ratas Sprague-Dawley , Aprendizaje Espacial/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Microvasc Res ; 135: 104134, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33428882

RESUMEN

BACKGROUND: Clinical data show that aneurysm rupture causes high mortality in aged men. MicroRNAs (miRNAs) were reported to regulate endothelial progenitor cells (EPCs) which play a vital role in repairing endothelial damage and maintaining vascular integrity. This study identified a novel miRNA regulator for the functions of EPCs in aneurysm repair. METHODS: Abdominal aortic aneurysm (AAA) model was established on Sprague-Dawley rats which later underwent antagomiR-222 treatment. The histopathological changes of AAA rats were examined by hematoxylin-eosin staining. Flow cytometry was performed to quantify EPCs in peripheral blood and identify EPCs isolated from the rat femur. The potential target of miR-222-3p was predicted by TargetScan v7.2 and validated by Dual-luciferase reporter assay. The effects of miR-222-3p and ADIPOR1 on the migration, invasion and tube formation of EPCs were evaluated by wound healing, Transwell and tube formation assays. The expressions of miR-222-3p and ADIPOR1 in aortic aneurysm tissues and EPCs were assessed by qRT-PCR or Western blot. RESULTS: AAA exhibited histopathological abnormality, a decreased number of EPCs in the peripheral blood and an increased miR-222-3p expression. AntagomiR-222 injection reversed all these phenomena in AAA rats. Upregulating miR-222-3p expression inhibited the migration, invasion, and tube formation of EPCs, and the expressions of ADIPOR1 and phosphorylated-AMKP, while downregulating miR-222-3p expression exerted opposite effects in EPCs. ADIPOR1 was identified as a target gene of miR-222-3p. Overexpressing ADIPOR1 abrogated the effects of miR-222-3p upregulation on EPCs. CONCLUSION: Downregulated miR-222-3p prompted the migration, invasion and recruitment of EPCs by targeting ADIPOR1-induced AMKP activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aorta Abdominal/enzimología , Aneurisma de la Aorta Abdominal/enzimología , Movimiento Celular , Células Progenitoras Endoteliales/enzimología , MicroARNs/metabolismo , Neovascularización Fisiológica , Receptores de Adiponectina/metabolismo , Animales , Antagomirs/genética , Antagomirs/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Progenitoras Endoteliales/patología , Activación Enzimática , Humanos , Masculino , MicroARNs/genética , Fosforilación , Ratas Sprague-Dawley , Receptores de Adiponectina/genética , Transducción de Señal
14.
Am J Physiol Heart Circ Physiol ; 320(4): H1348-H1360, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33416455

RESUMEN

Viral myocarditis (VMC) is a life-threatening disease characterized by severe cardiac inflammation generally caused by coxsackievirus B3 (CVB3) infection. Several microRNAs (miRNAs or miRs) are known to play crucial roles in the pathogenesis of VMC. The study aimed to decipher the role of miR-30a-5p in the underlying mechanisms of VMC pathogenesis. We first quantified miR-30a-5p expression in a CVB3-induced mouse VMC model. The physiological characteristics of mouse cardiac tissues were then detected by hematoxylin and eosin (HE) and Picrosirius red staining. We established the correlation between miR-30a-5p and SOCS1, using dual-luciferase gene assay and Pearson's correlation coefficient. The expression of inflammatory factors (IFN-γ, IL-6, IL-10, and IL-13), M1 polarization markers [TNF-α, inducible nitric oxide synthase (iNOS)], M2 polarization markers (Arg-1, IL-10), and myocardial hypertrophy markers [atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP)] was detected by RT-qPCR and Western blot analysis. miR-30a-5p was found to be highly expressed in VMC mice. Silencing of miR-30a-5p improved the cardiac function index and reduced heart weight-to-body weight ratio, myocardial tissue pathological changes and fibrosis degree, serological indexes, as well as proinflammatory factor levels, while enhancing anti-inflammatory factor levels in VMC mice. Furthermore, silencing of miR-30a-5p inhibited M1 polarization of macrophages while promoting M2 polarization in vivo and in vitro. SOCS1 was a target gene of miR-30a-5p, and the aforementioned cardioprotective effects of miR-30a-5p silencing were reversed upon silencing of SOCS1. Overall, this study shows that silencing of miR-30a-5p may promote M2 polarization of macrophages and improve cardiac injury following VMC via SOCS1 upregulation, constituting a potential therapeutic target for VMC treatment.NEW & NOTEWORTHY We found in this study that microRNA (miR)-30a-5p inhibition might improve cardiac injury following viral myocarditis (VMC) by accelerating M2 polarization of macrophages via SOCS1 upregulation. Furthermore, the anti-inflammatory mechanisms of miR-30a-5p inhibition may contribute to the development of new therapeutic strategies for VMC.


Asunto(s)
Infecciones por Coxsackievirus/terapia , Silenciador del Gen , Terapia Genética , Macrófagos/metabolismo , MicroARNs/genética , Miocarditis/terapia , Miocitos Cardíacos/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Animales , Antagomirs/genética , Antagomirs/metabolismo , Células Cultivadas , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/virología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Enterovirus Humano B/patogenicidad , Mediadores de Inflamación/metabolismo , Macrófagos/virología , Masculino , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Miocarditis/genética , Miocarditis/metabolismo , Miocarditis/virología , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología , Fenotipo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/genética
15.
Mol Cell Biol ; 41(2)2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33257506

RESUMEN

Colorectal cancer (CRC) has developed into the third leading cause of cancer-associated death worldwide. Studies have confirmed that circular RNAs (circRNAs) absorb microRNAs (miRNAs) to regulate the function of downstream genes. This study aimed to explore the underlying mechanism of circRNA 100146 in CRC. The expression of circRNA 100146, miRNA 149 (miR-149), and high mobility group AT-Hook 2 (HMGA2) was detected by quantitative real-time PCR (RT-qPCR). A series of biofunctional effects (cell viability, apoptosis, migration/invasion) were evaluated by the use of methyl thiazolyl tetrazolium (MTT), flow cytometry, and transwell assays. Protein levels were measured by Western blot assay. A xenograft model was established for in vivo experiments. The interactions among circRNA 100146, miR-149, and HMGA2 were evaluated by dual-luciferase reporter assay, RNA immunoprecipitation assays, or RNA pulldown assay. circRNA 100146 was upregulated in CRC tissues and cells. circRNA 100146 knockdown inhibited cell proliferation, promoted apoptosis, and suppressed migration and invasion in vitro and impeded tumor growth in vivo Also, miR-149 was negatively regulated by circRNA 100146 and was targeted to HMGA2 and mediated its expression. Moreover, miR-149 interference abrogated the activities of silenced circRNA 100146 in proliferation, apoptosis, migration, and invasion. Furthermore, HMGA2 overexpression abated the effects described above caused by circRNA 100146 silencing, while the mutations on miR-149 binding sites in the 3' untranslated region (3'-UTR) of HMGA2 led to its loss of this ability. circRNA 100146 knockdown repressed proliferation, enhanced apoptosis, and hindered migration and invasion in SW620 and SW480 cells through targeting the miR-149/HMGA2 axis.


Asunto(s)
Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Proteína HMGA2/genética , MicroARNs/genética , ARN Circular/genética , Anciano , Animales , Antagomirs/genética , Antagomirs/metabolismo , Emparejamiento Base , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Femenino , Genes Reporteros , Proteína HMGA2/antagonistas & inhibidores , Proteína HMGA2/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica , ARN Circular/antagonistas & inhibidores , ARN Circular/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Clin Lab Anal ; 35(2): e23608, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33277957

RESUMEN

BACKGROUND: Acute coronary syndrome (ACS) may induce cardiovascular death. The correlation of mast cells related microRNAs (miRs) with risk of ACS has been investigated. We explored regulatory mechanism of miR-335-5p on macrophage innate immune response, atherosclerotic vulnerable plaque formation, and revascularization in ACS in relation to Notch signaling. METHODS: ACS-related gene microarray was collected from Gene Expression Omnibus database. After different agomir or antagomir, or inhibitor of Notch signaling treatment, IL-6, IL-1ß, TNF-α, MCP-1, ICAM-1, and VCAM-1 levels were tested in ACS mice. Additionally, Notch signaling-related genes and matrix metalloproteinases (MMPs) were measured after miR-335-5p interference. Finally, mouse atherosclerosis, lipid accumulation, and the collagen/vessel area ratio of plaque were determined. RESULTS: miR-335-5p targeted JAG1 and mediated Notch signaling in ACS. miR-335-5p up-regulation and Notch signaling inhibition reduced expression of JAG1, Notch pathway-related genes, IL-6, IL-1ß, TNF-α, MCP-1, ICAM-1, VCAM-1, and MMPs, but promote TIMP1 and TIMP2 expression. Additionally, vulnerable plaques were decreased and collagen fiber contents were observed to increase after miR-335-5p overexpression and Notch signaling inhibition. CONCLUSIONS: Overexpression of miR-335-5p inhibited innate immune response of macrophage, reduced atherosclerotic vulnerable plaque formation, and promoted revascularization in ACS mice targeting JAG1 through Notch signaling.


Asunto(s)
Síndrome Coronario Agudo/genética , MicroARNs/genética , Placa Aterosclerótica/genética , Receptores Notch/metabolismo , Regiones no Traducidas 3' , Síndrome Coronario Agudo/tratamiento farmacológico , Síndrome Coronario Agudo/fisiopatología , Animales , Antagomirs/genética , Antagomirs/farmacología , Colágeno/genética , Colágeno/metabolismo , Diaminas/farmacología , Expresión Génica , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Lípidos/sangre , Lípidos/genética , Masculino , Ratones Endogámicos C57BL , Placa Aterosclerótica/fisiopatología , Receptores Notch/genética , Tiazoles/farmacología
17.
Aging (Albany NY) ; 12(23): 23609-23618, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33352533

RESUMEN

BACKGROUND: Myocardial infarction (MI), a common presentation for cardiovascular disease, is caused by reduction of blood flow and oxygen supply and is one of the main causes of death worldwide. MicroRNAs participate in multiple physiological and pathological processed and play crucial role in myocardial infarction. RESULTS: qRT-PCR analysis showed that expression level of miR-19a-3p was increased in serum of patient with MI. In vitro study indicated that the miR-19a-3p level was upregulated in response to H2O2 treatment and transferred by exosome, and then, uptake occurred in endothelial cells. Furthermore, western blot and immunostaining showed that treatment of exosome enriched miR-19a-3p suppressed the proliferation of endothelial cells and induced cell death, which was inhibited by AMO-19 transfection. Administration of antagomiR-19a-3p promoted angiogenesis and improved heart function of MI mice. Moreover, miR-19a-3p overexpression downregulated the protein level of HIF-1α and transfection of si-HIF-1α reversed the promotion of endothelial cells proliferation caused by AMO-19 transfection. In addition, antagomiR-19a-3p treatment accelerated angiogenesis and infection of AAV5-shHIF-1α inhibited that effect in MI mice. CONCLUSIONS: In conclusion, our finding indicated that miR-19a-3p inhibited endothelial cells proliferation and angiogenesis via targeting HIF-1α and attenuated heart function of mice after MI, and suggested a new mechanism of cell-to-cell communication between cardiomyocytes and endothelial cells.


Asunto(s)
Antagomirs/metabolismo , Células Endoteliales/metabolismo , Exosomas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , MicroARNs/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Animales , Antagomirs/genética , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/ultraestructura , Exosomas/genética , Exosomas/ultraestructura , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/ultraestructura , Comunicación Paracrina , Recuperación de la Función , Transducción de Señal
18.
Acta Biochim Pol ; 67(4): 453-460, 2020 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-33284571

RESUMEN

Asthma is considered as a general term for various chronic inflammatory diseases of the respiratory tract. Growing evidences have supported that microRNAs were involved in mediating cell proliferation, migration, and other cellular functions. MiR-149 has been found to take part in the development of various cancers. However, whether miR-149 participated in the proliferation and migration of transforming growth factor beta 1 (TGF-ß1)-induced airway smooth muscle cells was still unknown. In this study, the expression level of miR-149 in human airway smooth muscle cells (ASMCs) was decreased after TGF-ß1 treatment in vitro. Additionally, the over-expression of miR-149 obviously suppressed proliferation and migration in human ASMCs. Besides, we found that overexpression of miR-149 could inhibit the expression of transient receptor potential melastatin 7 (TRPM7) both in protein and gene levels. Furthermore, we demonstrated that miR-149 could inhibit the cell proliferation and migration in human ASMCs by targeting TRPM7 through modulating mitogen-activated protein kinases (MAPKs) signaling pathway. Taken together, we strongly supported that miR-149 might be a key inhibitor of asthma by targeting TRMP7. Therefore, our finding suggests a promising biomarker for the development of further targeted therapies for asthma.


Asunto(s)
MicroARNs/genética , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética , Canales Catiónicos TRPM/genética , Factor de Crecimiento Transformador beta1/farmacología , Antagomirs/genética , Antagomirs/metabolismo , Emparejamiento Base , Secuencia de Bases , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica , Células HEK293 , Humanos , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Oligorribonucleótidos/genética , Oligorribonucleótidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sistema Respiratorio/citología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Canales Catiónicos TRPM/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Acta Biochim Pol ; 67(4): 587-593, 2020 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-33332780

RESUMEN

BACKGROUND: MicroRNAs play a crucial role in diabetic peripheral neuropathic pain (DPNP). miR-590-3p is a novel miRNA and involved in multiple diseases. However, the pathological mechanism of miR-590-3p in DPNP needs to be elucidated. MATERIALS AND METHODS: The db/db mice and db/m mice were selected to mimic diabetes and control, respectively, for in vivo studies. The miR-590-3p agomir was injected into db/db mice and pain-related behavioral tests were performed. The interaction of miR-590-3p with target gene was confirmed by dual-luciferase reporter assay. The expression of target gene was determined by qRT-PCR and western blot assay. The levels of inflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: miR-590-3p was down-regulated in diabetic peripheral neuropathy mice. More importantly, miR-590-3p agomir alleviated pain-related behavior, reduced TNF-α, IL-1ß and IL-6 concentrations, and inhibited neural infiltration by immune cells in db/db mice. Interestingly, RAP1A was predicted to be the target of miR-590-3p by Targetscan, and was actually regulated by miR-590-3p. Finally, the rescue experiments proved that overexpression of RAP1A partially abrogated the suppressive impact of miR-590-3p on T cells proliferation and migration. CONCLUSION: miR-590-3p ameliorates DPNP via targeting RAP1A and inhibiting T cells infiltration, indicating that exogenous miR-590-3p may be a potential candidate for clinical treatment of DPNP.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Neuropatías Diabéticas/genética , MicroARNs/genética , Neuralgia/genética , Linfocitos T/inmunología , Proteínas de Unión al GTP rap1/genética , Animales , Antagomirs/genética , Antagomirs/metabolismo , Emparejamiento Base , Secuencia de Bases , Movimiento Celular , Proliferación Celular , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Neuropatías Diabéticas/inmunología , Neuropatías Diabéticas/patología , Modelos Animales de Enfermedad , Ganglios Espinales/inmunología , Ganglios Espinales/patología , Regulación de la Expresión Génica , Genes Reporteros , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/inmunología , Neuralgia/inmunología , Neuralgia/patología , Neuralgia/prevención & control , Oligorribonucleótidos/genética , Oligorribonucleótidos/metabolismo , Transducción de Señal , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas de Unión al GTP rap1/inmunología
20.
J Heart Lung Transplant ; 39(12): 1476-1490, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33067103

RESUMEN

BACKGROUND: Our hypothesis is that the immunomodulatory capacities of mesenchymal stem cell‒derived extracellular vesicles (EVs) can be enhanced by specific microRNAs (miRNAs) to effectively attenuate post-transplant lung ischemia‒reperfusion (IR) injury. METHODS: The expression of miR-206 was analyzed in bronchoalveolar lavage (BAL) fluid of patients on Days 0 and 1 after lung transplantation. Lung IR injury was evaluated in C57BL/6 mice using a left lung hilar-ligation model with or without treatment with EVs or antagomiR-206‒enriched EVs. Murine lung tissue was used for miRNA microarray hybridization analysis, and cytokine expression, lung injury, and edema were evaluated. A donation after circulatory death and murine orthotopic lung transplantation model was used to evaluate the protection by enriched EVs against lung IR injury. In vitro studies analyzed type II epithelial cell activation after coculturing with EVs. RESULTS: A significant upregulation of miR-206 was observed in the BAL fluid of patients on Day 1 after lung transplantation compared with Day 0 and in murine lungs after IR injury compared with sham. Treatment with antagomiR-206‒enriched EVs attenuated lung dysfunction, injury, and edema compared with treatment with EVs alone after murine lung IR injury. Enriched EVs reduced lung injury and neutrophil infiltration as well as improved allograft oxygenation after murine orthotopic lung transplantation. Enriched EVs significantly decreased proinflammatory cytokines, especially epithelial cell‒dependent CXCL1 expression, in the in vivo and in vitro IR injury models. CONCLUSIONS: EVs can be used as biomimetic nanovehicles for protective immunomodulation by enriching them with antagomiR-206 to mitigate epithelial cell activation and neutrophil infiltration in the lungs after IR injury.


Asunto(s)
Antagomirs/genética , Quimiocina CXCL1/genética , Regulación de la Expresión Génica , Lesión Pulmonar/prevención & control , MicroARNs/genética , Daño por Reperfusión/prevención & control , Animales , Antagomirs/biosíntesis , Líquido del Lavado Bronquioalveolar , Quimiocina CXCL1/biosíntesis , Modelos Animales de Enfermedad , Humanos , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Trasplante de Pulmón , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , ARN/genética , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...