Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Intervalo de año de publicación
1.
Int Immunopharmacol ; 130: 111519, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38442573

RESUMEN

This study investigates the molecular mechanisms by which extracellular vesicles (EVs) derived from adipose-derived mesenchymal stem cells (ADSCs) promote M2 polarization of macrophages and thus reduce lung injury caused by sepsis. High-throughput sequencing was used to identify differentially expressed genes related to long non-coding RNA (lncRNA) in ADSC-derived EVs (ADSC-EVs) in sepsis lung tissue. Weighted gene co-expression network analysis (WGCNA) was employed to predict the downstream target genes of the lncRNA DLEU2. The RNAInter database predicted miRNAs that interact with DLEU2 and LXN. Functional and pathway enrichment analyses were performed using GO and KEGG analysis. A mouse model of sepsis was established, and treatment with a placebo or ADSC-EVs was administered, followed by RT-qPCR analysis. ADSC-EVs were isolated and identified. In vitro cell experiments were conducted using the mouse lung epithelial cell line MLE-12, mouse macrophage cell line RAW264.7, and mouse lung epithelial cell line (LEPC). ADSC-EVs were co-cultured with RAW264.7 and MLE-12/LEPC cells to study the regulatory mechanism of the lncRNA DLEU2. Cell viability, proliferation, and apoptosis of lung injury cells were assessed using CCK-8, EdU, and flow cytometry. ELISA was used to measure the levels of inflammatory cytokines in the sepsis mouse model, flow cytometry was performed to determine the number of M1 and M2 macrophages, lung tissue pathology was evaluated by H&E staining, and immunohistochemistry was conducted to examine the expression of proliferation- and apoptosis-related proteins. High-throughput sequencing and bioinformatics analysis revealed enrichment of the lncRNA DLEU2 in ADSC-EVs in sepsis lung tissue. Animal and in vitro cell experiments showed increased expression of the lncRNA DLEU2 in sepsis lung tissue after treatment with ADSC-EVs. Furthermore, ADSC-EVs were found to transfer the lncRNA DLEU2 to macrophages, promoting M2 polarization, reducing inflammation response in lung injury cells, and enhancing their viability, proliferation, and apoptosis inhibition. Further functional experiments indicated that lncRNA DLEU2 promotes M2 polarization of macrophages by regulating miR-106a-5p/LXN, thereby enhancing the viability and proliferation of lung injury cells and inhibiting apoptosis. Overexpression of miR-106a-5p could reverse the biological effects of ADSC-EVs-DLEU2 on MLE-12 and LEPC in vitro cell models. Lastly, in vivo animal experiments confirmed that ADSC-EVs-DLEU2 promotes high expression of LXN by inhibiting the expression of miR-106a-5p, further facilitating M2 macrophage polarization and reducing lung edema, thus alleviating sepsis-induced lung injury. lncRNA DLEU2 in ADSC-EVs may promote M2 polarization of macrophages and enhance the viability and proliferation of lung injury cells while inhibiting inflammation and apoptosis reactions, thus ameliorating sepsis-induced lung injury in a mechanism involving the regulation of the miR-106a-5p/LXN axis.


Asunto(s)
Lesión Pulmonar , MicroARNs , Proteínas del Tejido Nervioso , ARN Largo no Codificante , Sepsis , Animales , Ratones , Apoptosis/genética , Modelos Animales de Enfermedad , Lesión Pulmonar/microbiología , Lesión Pulmonar/terapia , MicroARNs/genética , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/genética , Sepsis/complicaciones , Sepsis/genética , Proteínas del Tejido Nervioso/genética , Células Madre Mesenquimatosas , Exosomas , Masculino , Ratones Endogámicos C57BL
2.
Int Immunopharmacol ; 130: 111739, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38442574

RESUMEN

Persistent type (T) 2 airway inflammation plays an important role in the development of severe asthma. However, the molecular mechanisms leading to T2 severe asthma have yet to be fully clarified. Human normal lung epithelial cells (BEAS-2B cells) were transfected with LINC00158/BCL11B plasmid/small interfering RNA (siRNA). Levels of epithelial-mesenchymal transition (EMT)-related markers were measured using real-time qPCR (RT-qPCR) and western blot. A dual luciferase reporter assay was used to validate the targeting relationship between LINC00158 and BCL11B. The effects of LINC00158-lentivirus vector-mediated overexpression and dexamethasone on ovalbumin (OVA)/lipopolysaccharide (LPS)-induced severe asthma were investigated in mice in vivo. Our study showed that overexpression of LINC00158/BCL11B inhibited the levels of EMT-related proteins, apoptosis, and promoted the proliferation of BEAS-2B cells. BCL11B was a direct target of LINC00158. And LINC00158 targeted BCL11B to regulate EMT, apoptosis, and cell proliferation of BEAS-2B cells. Compared with severe asthma mice, LINC00158 overexpression alleviated OVA/LPS-induced airway hyperresponsiveness and airway inflammation, including reductions in T helper 2 cells factors in lung tissue and BALF, serum total- and OVA-specific IgE, inflammatory cell infiltration, and goblet cells hyperplasia. In addition, LINC00158 overexpression alleviated airway remodeling, including reduced plasma TGF-ß1 and collagen fiber deposition, as well as suppression of EMT. Additionally, overexpression of LINC00158 enhanced the therapeutic effect of dexamethasone in severe asthmatic mice models. LINC00158 regulates BEAS-2B cell biological function by targeting BCL11B. LINC00158 ameliorates T2 severe asthma in vivo and provides new insights into the clinical treatment of severe asthma.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma , ARN Largo no Codificante , Proteínas Represoras , Proteínas Supresoras de Tumor , Animales , Humanos , Ratones , Asma/inmunología , Asma/terapia , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Lipopolisacáridos , Pulmón/metabolismo , Ratones Endogámicos BALB C , Ovalbúmina , Proteínas Represoras/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Supresoras de Tumor/genética , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/genética , Transfección
3.
Ann N Y Acad Sci ; 1506(1): 118-141, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34791665

RESUMEN

The human transcriptome contains many types of noncoding RNAs, which rival the number of protein-coding species. From long noncoding RNAs (lncRNAs) that are over 200 nucleotides long to piwi-interacting RNAs (piRNAs) of only 20 nucleotides, noncoding RNAs play important roles in regulating transcription, epigenetic modifications, translation, and cell signaling. Roles for noncoding RNAs in disease mechanisms are also being uncovered, and several species have been identified as potential drug targets. On May 11-14, 2021, the Keystone eSymposium "Noncoding RNAs: Biology and Applications" brought together researchers working in RNA biology, structure, and technologies to accelerate both the understanding of RNA basic biology and the translation of those findings into clinical applications.


Asunto(s)
Congresos como Asunto/tendencias , Epigénesis Genética/genética , Marcación de Gen/tendencias , ARN no Traducido/administración & dosificación , ARN no Traducido/genética , Informe de Investigación , Animales , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/tendencias , Marcación de Gen/métodos , Humanos , MicroARNs/administración & dosificación , MicroARNs/genética , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Pequeño no Traducido/administración & dosificación , ARN Pequeño no Traducido/genética , Transducción de Señal/genética
4.
Neurochem Res ; 46(12): 3365-3374, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34514556

RESUMEN

Parkinson's disease (PD) is an age-related neurodegenerative disease caused by the loss of dopaminergic neurons in the substantia nigra. LncRNA MIAT has been shown to be critical in Alzheimer's disease, but its role and mechanism in PD are still unknown. Differentiated PC12 cells were treated with 1-methyl-4-phenylpyridinium (MPP+) to establish in vitro cell injury model of PD. MTT, Annexin V-PI double staining test and Western blot were used to detect cell viability and apoptosis. Reactive oxygen species (ROS), superoxide dismutase (SOD) and phospholipid hydroperoxide glutathione peroxidase (GSH-PX) kits were used to evaluate oxidative stress in cells. These results showed that LncRNA MIAT was down-regulated in MPP+-induced PC12 cells. Overexpression of LncRNA MIAT remarkably increased cell viability, inhibited cell apoptosis and oxidative stress in MPP+-treated cells. In addition, we proved that miR-132 is a target of LncRNA MIAT. Overexpression of miR-132 could reverse the positive effect of LncRNA MIAT overexpression on MPP+-induced cell oxidative stress injury. SIRT1 is a target of miR-132 and silencing of SIRT1 attunated the positive effect of LncRNA MIAT overexpression on oxidative stress injury in MPP+-induced PC12 cells. In conclusion, this study indicated that LncRNA MIAT suppressed MPP+-induced oxidative stress injury by regulating miR-132/SIRT1 axis in PC12 cells.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , ARN Largo no Codificante/administración & dosificación , Sirtuina 1/metabolismo , Animales , Apoptosis , Proliferación Celular , Herbicidas/toxicidad , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Células PC12 , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Sustancias Protectoras/farmacología , ARN Largo no Codificante/genética , Ratas , Sirtuina 1/genética
5.
Neurochem Int ; 150: 105173, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34453976

RESUMEN

The neuroprotective role of human adipose-derived stems cells (hASCs) has raised great interest in regenerative medicine due to their ability to modulate their surrounding environment. Our group has demonstrated that exosomes derived from hASC (hASCexo) are a cell-free regenerative approach to long term recovery following traumatic brain injury (TBI). Previously, we demonstrated the efficacy of exosome treatment with intravenous delivery at 3 h post TBI in rats. Here, we show efficacy of exosomes through intranasal delivery at 48 h post TBI in mice lengthening the therapeutic window of treatment and therefore increasing possible translation to clinical studies. Our findings demonstrate significant recovery of motor impairment assessed by an elevated body swing test in mice treated with exosomes containing MALAT1 compared to both TBI mice without exosomes and exosomes depleted of MALAT1. Significant cognitive improvement was seen in the reversal trial of 8 arm radial arm water maze in mice treated with exosomes containing MALAT1. Furthermore, cortical damage was significantly reduced in mice treated with exosomes containing MALAT1 as well as decreased MHCII+ staining of microglial cells. Mice without exosomes or treated with exosomes depleted of MALAT1 did not show similar recovery. Results demonstrate both inflammation related genes and NRTK3 (TrkC) are target genes modulated by hASC exosomes and further that MALAT1 in hASC exosomes regulates expression of full length TrkC thereby activating the MAPK pathway and promoting recovery. Exosomes are a promising therapeutic approach following TBI with a therapeutic window of at least 48 h and contain long noncoding RNA's, specifically MALAT1 that play a vital role in the mechanism of action.


Asunto(s)
Tejido Adiposo/trasplante , Lesiones Traumáticas del Encéfalo/terapia , Disfunción Cognitiva/terapia , Exosomas/trasplante , Trastornos Motores/terapia , Trasplante de Células Madre/métodos , Tejido Adiposo/metabolismo , Administración Intranasal , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Células Cultivadas , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Exosomas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Motores/metabolismo , Trastornos Motores/patología , ARN Largo no Codificante/administración & dosificación , Tiempo de Tratamiento
6.
Nat Commun ; 12(1): 3319, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083547

RESUMEN

Long noncoding RNAs are widely implicated in diverse disease processes. Nonetheless, their regulatory roles in bone resorption are undefined. Here, we identify lncRNA Nron as a critical suppressor of bone resorption. We demonstrate that osteoclastic Nron knockout mice exhibit an osteopenia phenotype with elevated bone resorption activity. Conversely, osteoclastic Nron transgenic mice exhibit lower bone resorption and higher bone mass. Furthermore, the pharmacological overexpression of Nron inhibits bone resorption, while caused apparent side effects in mice. To minimize the side effects, we further identify a functional motif of Nron. The delivery of Nron functional motif to osteoclasts effectively reverses bone loss without obvious side effects. Mechanistically, the functional motif of Nron interacts with E3 ubiquitin ligase CUL4B to regulate ERα stability. These results indicate that Nron is a key bone resorption suppressor, and the lncRNA functional motif could potentially be utilized to treat diseases with less risk of side effects.


Asunto(s)
Osteoporosis/genética , Osteoporosis/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Animales , Enfermedades Óseas Metabólicas/etiología , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/prevención & control , Proteínas Cullin/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Fémur/diagnóstico por imagen , Fémur/metabolismo , Fémur/patología , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoporosis/terapia , ARN Largo no Codificante/administración & dosificación , Ubiquitinación , Regulación hacia Arriba , Microtomografía por Rayos X
7.
Mol Neurobiol ; 58(8): 3712-3728, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33818737

RESUMEN

Stroke is a major cause of premature mortality and disability around the world. Therefore, identification of cellular and molecular processes implicated in the pathogenesis and progression of ischemic stroke has become a priority. Long non-coding RNAs (lncRNAs) are emerging as significant players in the pathophysiology of cerebral ischemia. They are involved in different signalling pathways of cellular processes like cell apoptosis, autophagy, angiogenesis, inflammation, and cell death, impacting the progression of cerebral damage. Exploring the functions of these lncRNAs and their mechanism of action may help in the development of promising treatment strategies. In this review, the current knowledge of lncRNAs in ischemic stroke, focusing on the mechanism by which they cause cellular apoptosis, inflammation, and microglial activation, has been summarized. Very few lncRNAs have been functionally annotated. Therefore, the therapies based on lncRNAs still face many hurdles since the potential targets are likely to increase with the identification of new ones. Majority of experiments involving the identification and function of lncRNAs have been carried out in animal models, and the role of lncRNAs in human stroke presents a challenge. However, mitigating these issues through more rational experimental design might lead to the development of lncRNA-based stroke therapies to treat ischemic stroke.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/terapia , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/genética , Animales , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Muerte Celular/fisiología , Humanos , Accidente Cerebrovascular Isquémico/metabolismo , ARN Largo no Codificante/metabolismo
8.
Int J Nanomedicine ; 16: 1051-1066, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33603368

RESUMEN

BACKGROUND: This study was aimed to prepare a novel magnetic thermosensitive cationic liposome drug carrier for the codelivery of Oxaliplatin (OXA) and antisense lncRNA of MDC1 (MDC1-AS) to Cervical cancer cells and evaluate the efficiency of this drug carrier and its antitumor effects on Cervical cancer. METHODS: Thermosensitive magnetic cationic liposomes were prepared using thin-film hydration method. The OXA and MDC1-AS vectors were loaded into the codelivery system, and the in vitro OXA thermosensitive release activity, efficiency of MDC1-AS regulating MDC1, in vitro cytotoxicity, and in vivo antitumor activity were determined. RESULTS: The codelivery system had desirable targeted delivery efficacy, OXA thermosensitive release, and MDC1-AS regulating MDC1. Codelivery of OXA and MDC1-AS enhanced the inhibition of cervical cancer cell growth in vitro and in vivo, compared with single drug delivery. CONCLUSION: The novel codelivery of OXA and MDC1-AS magnetic thermosensitive cationic liposome drug carrier can be applied in the combined chemotherapy and gene therapy for cervical cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/genética , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Fenómenos Magnéticos , Terapia Molecular Dirigida , Oxaliplatino/uso terapéutico , ARN Largo no Codificante/administración & dosificación , Neoplasias del Cuello Uterino/terapia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Cationes , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Liberación de Fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Liposomas , Ratones Endogámicos BALB C , Ratones Desnudos , Oxaliplatino/farmacología , Tamaño de la Partícula , Electricidad Estática , Temperatura , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
9.
RNA Biol ; 18(11): 1546-1554, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33427561

RESUMEN

Pathological changes involving TDP-43 protein ('TDP-43 proteinopathy') are typical for several neurodegenerative diseases, including frontotemporal lobar degeneration (FTLD). FTLD-TDP cases are characterized by increased binding of TDP-43 to an abundant lncRNA, NEAT1, in the cortex. However it is unclear whether enhanced TDP-43-NEAT1 interaction represents a protective mechanism. We show that accumulation of human TDP-43 leads to upregulation of the constitutive NEAT1 isoform, NEAT1_1, in cultured cells and in the brains of transgenic mice. Further, we demonstrate that overexpression of NEAT1_1 ameliorates TDP-43 toxicity in Drosophila and yeast models of TDP-43 proteinopathy. Thus, NEAT1_1 upregulation may be protective in TDP-43 proteinopathies affecting the brain. Approaches to boost NEAT1_1 expression in the CNS may prove useful in the treatment of these conditions.


Asunto(s)
Esclerosis Amiotrófica Lateral/prevención & control , Encéfalo/metabolismo , Proteínas de Unión al ADN/toxicidad , Demencia Frontotemporal/prevención & control , Neuroblastoma/prevención & control , ARN Largo no Codificante/genética , Proteinopatías TDP-43/prevención & control , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Drosophila melanogaster , Demencia Frontotemporal/etiología , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroblastoma/etiología , Neuroblastoma/metabolismo , Neuroblastoma/patología , ARN Largo no Codificante/administración & dosificación , Saccharomyces cerevisiae , Proteinopatías TDP-43/etiología , Proteinopatías TDP-43/metabolismo , Proteinopatías TDP-43/patología
10.
Anticancer Drugs ; 32(2): 178-188, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32826414

RESUMEN

Ultrasound-mediated nanobubble destruction (UMND), which can utilize the physical energy of ultrasound irradiation to improve the transfer efficiency to target cells is becoming one of the most promising carriers for gene delivery. The purpose of this study was to establish cell-penetrating peptide (CPP)-loaded nanobubbles (CNBs) connected with long intergenic nonprotein coding RNA 00511-small interfering RNA (LINC00511-siRNA) and evaluate its feasibility for improving the chemosensitivity of triple-negative breast cancer in vitro. First, fluorescence imaging confirmed the loading of siLINC00511 on CNBs, and the CNBs-siLINC00511 were characterized by the Zetasizer Nano ZS90 analyzer and transmission electron microscopy. Next, cell counting kit 8 assay was used to detect the inhibitory activity of cisplatin on the proliferation of MDA-MB-231 cells, and the 50% inhibition concentration value before and after transfer was calculated. Finally, the silencing effect of siLINC00511 was evaluated in vitro using an apoptosis assay, transwell assay, real time-PCR and western blotting. UMND combined with CNBs could effectively transfer the siRNA to MDA-MB-231 cells, thus evidently reducing the expression of LINC00511. Furthermore, inhibitory activity of cisplatin on MDA-MB-231 cells was enhanced after downregulation of LINC00511 expression. Downregulation of LINC00511 alters expression of cell cycle-related (CDK 6) and apoptosis-related (Bcl-2 and Bax) proteins in MDA-MB-231 cells. These results suggested that siRNA-CNBs may be an ideal vector for the treatment of tumors, with high efficiency RNA interference under the combined action of UMND. It may provide a new therapeutic method for triple negative breast cancer.


Asunto(s)
Péptidos de Penetración Celular/farmacología , Cisplatino/farmacología , ARN Largo no Codificante/farmacología , ARN Interferente Pequeño/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ondas Ultrasónicas , Línea Celular Tumoral , Péptidos de Penetración Celular/administración & dosificación , Cisplatino/administración & dosificación , Quinasa 6 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo , Femenino , Vectores Genéticos , Humanos , Concentración 50 Inhibidora , Nanopartículas/química , Polímeros/química , ARN Largo no Codificante/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Neoplasias de la Mama Triple Negativas/patología
11.
Mol Pharm ; 17(11): 4040-4066, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32902291

RESUMEN

Nucleic acid therapeutics for RNA interference (RNAi) are gaining attention in the treatment and management of several kinds of the so-called "undruggable" tumors via targeting specific molecular pathways or oncogenes. Synthetic ribonucleic acid (RNAs) oligonucleotides like siRNA, miRNA, shRNA, and lncRNA have shown potential as novel therapeutics. However, the delivery of such oligonucleotides is significantly hampered by their physiochemical (such as hydrophilicity, negative charge, and instability) and biopharmaceutical features (in vivo serum stability, fast renal clearance, interaction with extracellular proteins, and hindrance in cellular internalization) that markedly reduce their biological activity. Recently, several nanocarriers have evolved as suitable non-viral vectors for oligonucleotide delivery, which are known to either complex or conjugate with these oligonucleotides efficiently and also overcome the extracellular and intracellular barriers, thereby allowing access to the tumoral micro-environment for the better and desired outcome in glioblastoma multiforme (GBM). This Review focuses on the up-to-date advancements in the field of RNAi nanotherapeutics utilized for GBM treatment.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Terapia Genética/métodos , Glioblastoma/tratamiento farmacológico , MicroARNs/administración & dosificación , Nanoconjugados/química , Oligonucleótidos/administración & dosificación , Interferencia de ARN , ARN Largo no Codificante/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Animales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , MicroARNs/química , MicroARNs/genética , Oligonucleótidos/química , Oligonucleótidos/genética , ARN Largo no Codificante/química , ARN Largo no Codificante/genética , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Biomed Pharmacother ; 131: 110572, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32836073

RESUMEN

Long non-coding RNA (lncRNA) was once considered to be the "noise" of genome transcription without biological function. However, increasing evidence shows that lncRNA is dynamically expressed in developmental stage or disease status, playing a regulatory role in the process of gene expression and translation. In recent years, lncRNA is considered to be a core node of functional regulatory networks that controls cardiac and also involves in multiple process of heart failure such as myocardial hypertrophy, fibrosis, angiogenesis, etc., which would be a therapeutic target for diseases. In fact, it is the development of technology that has improved our understanding of lncRNAs and broadened our perspective on heart failure. From transcriptional "noise" to star molecule, progress of lncRNAs can't be achieved without the combination of multidisciplinary technologies, especially the emergence of high-throughput approach. Thus, here, we review the strategies and technologies available for the exploration lncRNAs and try to yield insights into the prospect of lncRNAs in clinical diagnosis and treatment in heart failure.


Asunto(s)
Tecnología Biomédica/tendencias , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/terapia , ARN Largo no Codificante/biosíntesis , Animales , Tecnología Biomédica/métodos , Insuficiencia Cardíaca/genética , Humanos , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/genética
13.
Cancer Biomark ; 28(2): 231-246, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508321

RESUMEN

Chemotherapy is one of the primary treatments used against cancer. Cisplatin is a conventional chemotherapy drug used to treat osteosarcoma; however, due to the development of cisplatin resistance, advantageous therapeutic outcomes and prognosis of osteosarcoma remain low. Thus, investigation of the specific targeted therapies to circumvent the anti-chemoresistance of osteosarcoma depends on understanding the molecular mechanisms underlying cisplatin resistance. Tumor cells display an increased utilization of glycolysis rather than oxidative phosphorylation. This phenomenon is called the "Warburg effect," which presents a survival advantage for tumor cells, leading to chemoresistance. To date, the molecular mechanism underlying osteosarcoma cisplatin resistance remains to be fully elucidated. In this study, we reported the significant down-regulation of the long noncoding RNA-Suppressing Androgen Receptor in Renal Cell Carcinoma (lncRNA-SARCC) in the cells of osteosarcoma and in the specimens from osteosarcoma patients. Moreover, we observed a negative correlation between the lncRNA-SARCC and cisplatin resistance in the osteosarcoma tissues. Overexpression of the lncRNA-SARCC sensitizes osteosarcoma cells to cisplatin. From microarray analysis, we screened several miRNAs, which are significantly regulated by the lncRNA-SARCC in osteosarcoma cells, and revealed that lncRNA-SARCC promoted microRNA-43 (miR-143) expression in osteosarcoma. Interestingly, miR-143 showed the same expression pattern with the lncRNA-SARCC in osteosarcoma patient specimens. By establishing a cisplatin-resistant cell line from Sarcoma Osteogenic-2 (Saos-2), we found the cisplatin-resistant cells with down-regulated expressions of the lncRNA-SARCC and miR-143, but with a higher glycolysis rate compared to that in parental cells. We identified the glycolysis key enzyme, Hexokinase 2 (HK2), as a direct target for miR-143 in osteosarcoma. Restoration of the HK2 expression in the lncRNA-SARCC-overexpressing osteosarcoma cells reversed cisplatin resistance, suggesting that lncRNA-SARCC-mediated cisplatin sensitivity may be via glycolysis in the miR-143-inhibited osteosarcoma cells. Finally, results from both in vitro and in vivo xenograft models demonstrated that the lncRNA-SARCC was an effective therapeutic agent for overcoming cisplatin resistance in osteosarcoma. Our findings suggest an essential axis of the lncRNA-SARCC-miR-143-HK2 in regulation of osteosarcoma chemosensitivity, presenting the lncRNA-SARCC as a new therapeutic target against cisplatin-resistant osteosarcoma.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Hexoquinasa/genética , MicroARNs/metabolismo , Osteosarcoma/tratamiento farmacológico , ARN Largo no Codificante/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Huesos/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/uso terapéutico , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Glucólisis/efectos de los fármacos , Glucólisis/genética , Humanos , Inyecciones Intraperitoneales , Ratones , Osteosarcoma/genética , Osteosarcoma/patología , ARN Largo no Codificante/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancer Lett ; 479: 42-53, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32200038

RESUMEN

Triple-negative breast cancer (TNBC) has special characteristics of significant aggressiveness, and strong potential for metastasis and recurrence; currently there are no targeted drugs for TNBC. Abnormal activation of epithelial-mesenchymal transition (EMT) plays an important role in these malignant behaviors of TNBC. In the crosstalk among the multiple EMT-associated signaling pathways, many miRNAs participate in regulating pathway activity, where they act as "traffic lights" at the intersection of these pathways. In this study, we used miRNA microarray technology to detect differentially expressed miRNAs related to EMT in TNBC, and we identified and verified 9 highly expressed oncogenic miRNAs (OncomiRs). High expression of these OncomiRs in clinical breast cancer tissues affected the prognosis of patients, and inhibition of their expression blocked EMT in TNBC cell lines and suppressed cancer cell proliferation and migration. We constructed an oncolytic adenovirus (AdSVP-lncRNAi9) armed with an artificially-designed interfering lncRNA (lncRNAi9), which exhibited an activity to block EMT in TNBC cells by disrupting the functions of multiple OncomiRs; the efficacy of such a treatment for TNBC was demonstrated in cytology and animal experiments. This research provides a new candidate oncolytic virotherapy for treating highly malignant refractory TNBC.


Asunto(s)
Dependovirus/genética , Viroterapia Oncolítica/métodos , ARN Largo no Codificante/administración & dosificación , Neoplasias de la Mama Triple Negativas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dependovirus/fisiología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Células MCF-7 , Ratones , Persona de Mediana Edad , ARN Largo no Codificante/genética , ARN Largo no Codificante/farmacología , Neoplasias de la Mama Triple Negativas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Cell Biochem ; 121(10): 4261-4270, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31909503

RESUMEN

This article aimed to reveal the mechanism of long noncoding RNA (lncRNA) urothelial cancer-associated 1 (UCA1) regulated astrocyte activation in temporal lobe epilepsy (TLE) rats via mediating the activation of the JAK/STAT signaling pathway. A model of TLE was established based on rats via kainic acid (KA) injection. All rats were divided into the Sham group (without any treatments), KA group, normal control (NC; injection with empty vector) + KA group, and UCA1 + KA group. The Morris water maze was used to test the learning and memory ability of rats, and the expression of UCA1 in the hippocampus was determined by quantitative real time polymerase chain reaction (qRT-PCR). Surviving neurons were counted by Nissl staining, and expression levels of glial cells glial fibrillary acidic protein (GFAP), p-JAK1, and p-STAT3 and glutamate/aspartate transporter (GLAST) were analyzed by immunofluorescence and Western blot analysis. A rat model of TLE was established by intraperitoneal injection of KA. qRT-PCR and fluorescence analyses showed that UCA1 inhibited astrocyte activation in the hippocampus of epileptic rats. Meanwhile, the Morris water maze analysis indicated that UCA1 improved the learning and memory in epilepsy rats. Moreover, the Nissl staining showed that UCA1 might have a protective effect on neuronal injury induced by KA injection. Furthermore, the immunofluorescence and Western blot analysis revealed that the overexpression of UCA1 inhibited KA-induced abnormal elevation of GLAST, astrocyte activation of the JAK/STAT signaling pathway, as well as hippocampus of epilepsy rats. UCA1 inhibited hippocampal astrocyte activation and JAK/STAT/GLAST expression in TLE rats and improved the adverse reactions caused by epilepsy.


Asunto(s)
Astrocitos/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Janus Quinasa 1/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Animales , Conducta Animal , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/inducido químicamente , Vectores Genéticos/administración & dosificación , Hipocampo/metabolismo , Ácido Kaínico/efectos adversos , Masculino , Memoria , Prueba del Laberinto Acuático de Morris , Neuroglía/metabolismo , Neuronas/metabolismo , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/genética , Ratas , Ratas Sprague-Dawley
16.
Inflammation ; 43(2): 605-618, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31900829

RESUMEN

Both long non-coding RNA (lncRNA) RMRP and heat shock protein (HSP) 70 have been known to play crucial roles in inflammation. The present study investigated the roles of lncRNA RMRP and HSP70 protein 4 (HSPA4) in lipopolysaccharide (LPS)-induced sepsis. The C57BL/6 mice were treated with LPS, following which the cardiomyocytes were isolated for in vitro experiments. Further, a cardiac muscle cell line, HL-1 was transfected with plasmids expressing RMRP and HSPA4, si-NC, si-HSPA4, miR-1-5p mimic, and controls in vitro. Cell apoptosis, mitochondrial membrane potential (MMP), and levels of intracellular reactive oxygen species (ROS), mRNAs, and proteins were detected in the transfected mice tissues and cells. The LPS treatment significantly reduced the expression levels of RMRP, MMP, and mitochondrial cytochrome C. Moreover, it enhanced the cardiomyocyte apoptosis, intracellular ROS levels, cytoplasm cytochrome C levels, and the expression of caspase-3 and caspase-9 and nuclear factor κB (NF-κB) p65 subunit. The predicted RMRP-miR-1-5p-HSPA4 network was validated by co-transfection experiments in vitro in HL-1 cells. The transfection of miR-1-5p-treated cells with pcDNA-RMRP enhanced the levels of the protein HSPA4; however, no change at the mRNA level was observed. Moreover, miR-1-5p mimic attenuated the protective effect of pcDNA-HSPA4 against LPS-induced mitochondrial damage and apoptosis. In addition, we observed that silencing of HSPA4 increased the expression of nuclear p65; however, this effect could be reversed by co-transfection with pcDNA-RMRP. The lncRNA RMRP axis acts as a sponge for miR-1-5p. RMRP inhibits LPS-induced apoptosis of cardiomyocytes and mitochondrial damage by suppressing the post-transcriptional regulatory function of miR-1-5p on HSPA4. We believe that RMRP exhibits therapeutic potential for LPS-induced myocardial dysfunction both in vitro and in vivo.


Asunto(s)
Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , MicroARNs/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , ARN Largo no Codificante/administración & dosificación , Sepsis/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células Cultivadas , Proteínas HSP70 de Choque Térmico/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/metabolismo , Sepsis/inducido químicamente , Sepsis/metabolismo
17.
J Neuroimmune Pharmacol ; 15(3): 390-399, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31325121

RESUMEN

Long non-coding RNAs (lncRNAs), including long intergenic non-coding RNAs (lincRNAs), play an important regulatory role in controlling various biological processes. Both in vitro and in vivo studies have demonstrated that lincRNA-Cox2 plays a global regulatory role in regulating the expression of immune genes. Extracellular vesicles (EVs) are cell-derived nanosized membrane vesicles that have gained increasing attention in recent years due to their ability to efficiently deliver therapeutics to specific target organs or cell types. In this study, we found that lincRNA-Cox2 controls the expression of a set of cell cycle genes in lipopolysaccharide (LPS)-stimulated microglial cells. Our in vitro study suggested that knocking down lincRNA-Cox2 reversed LPS-induced microglial proliferation. In addition, our in vivo study demonstrated that intranasally delivered lincRNA-Cox2-siRNA loaded EVs could reach the brain resulting in a significant decrease in the expression of lincRNA-Cox2 in the microglia. Importantly, lincRNA-Cox2-siRNA loaded EVs also decreased LPS-induced microglial proliferation in mice. These findings indicate that intranasal delivery of EV-loaded small RNA could be developed as therapeutics for treatment of a multitude of CNS disorders.


Asunto(s)
Ciclooxigenasa 2/genética , Vesículas Extracelulares , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , ARN Largo no Codificante/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Administración Intranasal , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Genes cdc/genética , Ratones , Ratones Endogámicos C57BL , Nanopartículas
18.
Eur Rev Med Pharmacol Sci ; 23(12): 5366-5373, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31298389

RESUMEN

OBJECTIVE: To investigate the effects of long non-coding ribonucleic acid (lncRNA) small nucleolar RNA host gene 1 (SNHG1) on the neuronal apoptosis in rats with cerebral infarction through the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. MATERIALS AND METHODS: Male Sprague Dawley (SD) rats were divided into M group (model control group), N group (rat model of cerebral infarction) and R group (rat model of cerebral infarction plus lncRNA SNHG1) and then treated accordingly. 2,3,5-triphenyl tetrazolium chloride (TTC) staining was applied to detect the percentage of cerebral infarct volume and apoptosis of brain cells in the three groups of rats; hematoxylin and eosin (HE) staining was utilized to observe the pathological morphology of brain tissues, and Western blotting was performed to measure the protein levels of phosphorylated PI3K (p-PI3K) and p-Akt in the brain tissues. RESULTS: The degree of neurological deficit in the N group was much higher than that in the M group (p<0.05), and it was decreased markedly in the R group compared with that in the N group, with statistically significant differences (p<0.05). In comparison with that in the M group, the cell apoptosis was aggravated notably in the N group and alleviated remarkably in the R group, and the differences were statistically significant (p<0.05). In the N group, the cerebral infarct volume accounted for 33.67% of the whole brain volume, and mild cerebral infarction was detected in the R group, with a percentage of cerebral infarct volume of 20.15%. N group had a more prominent increase in the cerebral infarct volume than the R group (p<0.05). Compared with those in the M group, the pyknotic nuclei and neuron staining of brain tissues were increased significantly, and the neuronal cell injury was aggravated in the N group, while markedly reduced pyknotic nuclei and neuron staining (p<0.05), as well as mild neuronal cell injury (p<0.05), were detected in the R group. The levels of p-PI3K and p-Akt proteins in the brain tissues declined remarkably in the N group compared with those in the R group (p<0.05). CONCLUSIONS: The protective effect of lncRNA SNHG1 on the rats with cerebral infarction is correlated with the activation of the PI3K/Akt signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , ARN Largo no Codificante/administración & dosificación , Animales , Apoptosis/genética , Apoptosis/inmunología , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/patología , Masculino , Neuronas/efectos de los fármacos , Neuronas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología
19.
Eur Rev Med Pharmacol Sci ; 22(17): 5647-5651, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30229841

RESUMEN

OBJECTIVE: We investigate the effect of long non-coding RNA H19 in acute myocardial infarction (AMI) and the underlying mechanism. MATERIALS AND METHODS: C57BL/6 mice were subjected to AMI and injected with lentivirus pcDNA-H19. After AMI procedures for 3 weeks, cardiac function was detected by echocardiography. The infarct size was stained by triphenyltetrazolium chloride. H19 expression in mice was measured by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). Protein expressions of LC3, Beclin-1, and ATG-7 in mice were measured by Western blot. RESULTS: Our results indicated that H19 expression was significantly downregulated in the infarcted myocardium. Overexpression of H19 after injection with pcDNA-H19 in mice could reduce infarct size and improve cardiac function through upregulating the ratio of LC3-II/I and expressions of Beclin-1 and ATG-7. CONCLUSIONS: Overexpression of H19 could protect AMI in mice via activating autophagy.


Asunto(s)
Autofagia , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/administración & dosificación , Animales , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal , Función Ventricular Izquierda
20.
Medicine (Baltimore) ; 97(36): e12131, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30200102

RESUMEN

This study aimed to assess the effect of long noncoding RNAs (lncRNAs) taurine-upregulated gene 1 (TUG1) on cells proliferation and apoptosis as well as its targeting genes in epithelial ovarian cancer (EOC) cells.Blank mimic, lncRNA TUG1 mimic, blank inhibitor, and lncRNA TUG1 inhibitor plasmids were transfected into SK-OV-3 (SKOV3) cells. Rescue experiment was performed by the transfection of lncRNA TUG1 inhibitor and Aurora kinase A (AURKA) mimic plasmids into SKOV3 cells. Cell counting kit-8 (CKK-8), annexin V-FITC (AV)-propidium iodide (PI) (AV-PI), quantitative polymerase chain reaction (qPCR), and western blot assays were performed to detect cells proliferation, apoptosis, RNA expression, and protein expression respectively.Cells proliferation was increased in lncRNA TUG1 mimic group and decreased in lncRNA TUG1 inhibitor group than normal control (NC) groups. Cells apoptosis rate was repressed after treatment with lncRNA TUG1 mimic and promoted after treatment with lncRNA TUG1 inhibitor. AURKA expression but not CLDN3, SERPINE1, or ETS1 expression was adversely regulated by lncRNA TUG1 mimic and inhibitor. After transferring lncRNA TUG1 (-) and AURKA (+) plasmids, cells proliferation was increased, while cells apoptosis rate was decreased in AURKA mimic (+)/lncRNA TUG1 inhibitor (-) group than NC (+)/lncRNA TUG1 (-) group, which suggested lncRNA TUG1 regulated cells proliferation and cells apoptosis through targeting AURKA.LncRNA TUG1 promotes cells proliferation and inhibits cells apoptosis through regulating AURKA in EOC cells.


Asunto(s)
Apoptosis/fisiología , Aurora Quinasa A/metabolismo , Proliferación Celular/fisiología , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/metabolismo , ARN Largo no Codificante/metabolismo , Aurora Quinasa A/administración & dosificación , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Claudina-3/metabolismo , Humanos , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , ARN Largo no Codificante/administración & dosificación , ARN Largo no Codificante/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...