Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Biochem Genet ; 62(5): 4174-4190, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38244156

RESUMEN

Colorectal carcinoma (CRC) is one of the most common malignant tumors in the digestive tract. It was found that butyric acid could inhibit the expression of miR-183 to slow down malignant progression of CRC in the early stage. However, its regulatory mechanism remains unclear. This study screened the IC50 value of butyrate on inhibition of CRC cells malignant progression. Its inhibitory effects were detected by MTT assay, colony formation experiment, Transwell migration experiment, and apoptosis evaluation by flow cytometry. Next, the expressions of miR-183 and DNAJB4 were, respectively, determined in butyrate treated and miR-183 analog or si-DNAJB4-transfected CRC cells to further detect the role of upregulated miR-183 or silencing DNAJB4 in CRC cells malignant progression. Subsequently, the targeted regulatory relationship between miR-183 and si-DNAJB4 was confirmed by bioinformatic prediction tools and double luciferase report genes analysis method. The regulatory mechanism of butyrate on miR-183/DNAJB4 axis signal pathway was evaluated in molecular level, and verified in nude mouse xerograft tumor model and immunohistochemical analysis tests of Ki67 positive rates. The results displayed that butyrate with increased concentration can hinder the proliferation and improve apoptosis of CRC cells by decreasing the expression of miR-183. Thus, butyrate reduces miR-183 expression and increases DNAJB4 expression via the miR-183/DNAJB4 axis, ultimately inhibiting the malignant progression and increasing apoptosis of CRC. While over expression of miR-183 downregulate the expression of DNAJB4, which can reverse the inhibitory effect of butyrate.


Asunto(s)
Ácido Butírico , Proliferación Celular , Ratones Desnudos , MicroARNs , MicroARNs/genética , MicroARNs/metabolismo , Humanos , Animales , Proliferación Celular/efectos de los fármacos , Ratones , Ácido Butírico/farmacología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/genética , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Int J Mol Med ; 53(3)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38214365

RESUMEN

Following the publication of this article, a concerned reader drew to the Editor's attention that, in Fig. 9C on p. 2478 showing the results of Transwell invasion assay experiments, unexpected areas of similarity were identified in terms of the cellular patterns revealed both within the data panels for the six different experiments portrayed in this figure, and comparing among them. After having conducted an internal investigation, the Editor of International Journal of Molecular Medicine has reached the conclusion that the overlapping sections of data shown in this figure were unlikely to have arisen by coincidence. Therefore, on the grounds of a lack of confidence in the integrity of these data, the Editor has decided that the article should be retracted from the publication. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused, and thanks the interested reader for drawing this matter to our attention. [International Journal of Molecular Medicine 42: 2469­2480, 2018; DOI: 10.3892/ijmm.2018.3853].

3.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1025411

RESUMEN

Objective:To study the expression levels and clinical significance of microR-NA-183-5p(miR-183-5p)and thioesterase superfamily member 4(THEM4)in colon cancer tissues.Methods:A total of 96 patients with colon cancer who in Hebei China Petroleum Central Hospital gathered as the research objects.During the course of radical resection of colon cancer patients,the colon cancer tissues and adjacent normal tissues were collected.The relative expression levels of miR-183-5p and THEM4 mRNA in colon cancer tissues and adjacent normal tissues were detected.Analysis of the correlation between miR-183-5pand THEM4mRNA in colon cancer and their relation-ship with prognosis.COX regression was used to analyze the risk factors affecting the prognosis of pa-tients with colon cancer.Results:Compared with adjacent normal tissues,the expression level of miR-183-5p in colon cancer tissues increased(P<0.05),and the expression level of THEM4 mRNA decreased(P<0.05).MiR-183-5p was negatively correlated with THEM4 mRNA expression in colon cancer tissue(r=-0.529,P<0.05).The survival rate of the high expression group of miR-183-5p lower than that of the low expression group(P<0.05),the survival rate of the high expression group of THEM4 was obviously higher than that of the low expression group(P<0.05).TNM stage(Ⅲ-Ⅳ),high expres-sion of miR-183-5p and low expression of THEM4 were risk factors for poor prognosis in patients with colon cancer(P<0.05).Conclusion:The expression level of miR-183-5p in cancer tissues of patients with colon cancer is increased,and the expression level of THEM4 is decreased,both are closely relat-ed to the clinicopathological characteristics and prognosis of patients.

4.
Cell Biol Toxicol ; 39(6): 2953-2970, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37296288

RESUMEN

Paclitaxel treatment has been applied for late-stage nasopharyngeal carcinoma (NPC), but therapy failure usually occurs due to paclitaxel resistance. Besides, microRNAs (miRs) delivered by extracellular vesicles (EVs) have been demonstrated as promising biomarkers affecting cancer development. Our work clarified the role of bioinformatically predicted miR-183-5p, which could be delivered by EVs, in the paclitaxel resistance of NPC. Downstream targets of miR-183-5p were predicted in publicly available databases, followed by GO enrichment analysis. A confirmatory dual-luciferase reporter assay determined the targeting relationship between miR-183-5p and P-glycoprotein (P-gp). The shuttling of extracellular miR-183-5p was identified by immunofluorescence. EVs transferred miR-183-5p from paclitaxel-sensitive NPC cells to paclitaxel-resistant NPC cells. Furthermore, overexpression of miR-183-5p and under-expression of P-gp occurred in clinical samples and cells of NPC. High expression of miR-183-5p corresponded to better survival of paclitaxel-treated patients. The effects of manipulated expression of miR-183-5p on NPC cell activities, tumor growth, and paclitaxel resistance were investigated in vitro and in vivo. Its effect was achieved through negatively regulating drug transporters P-gp. Ectopically expressed miR-183-5p enhanced the cancer-suppressive effects of paclitaxel by targeting P-gp, corresponding to diminished cell viability and tumor growth. Taken together, this work goes to elucidate the mechanical actions of miR-183-5p delivered by EVs and its significant contribution towards paclitaxel sensitivity to NPC. 1. This study provides mechanistic insight into the role of miR-183-5p-containing EVs in NPC. 2. The intercellular transportation of miR-183-5p is mediated by EVs in NPC. 3. Overexpressing miR-183-5p facilitates the anti-tumor effects of paclitaxel in NPC. 4. miR-183-5p suppresses paclitaxel resistance of NPC cells by inhibiting P-gp.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Neoplasias Nasofaríngeas , Humanos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Carcinoma Nasofaríngeo/genética , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/genética , MicroARNs/genética , MicroARNs/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Vesículas Extracelulares/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica
5.
J Cell Mol Med ; 27(9): 1179-1191, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36988055

RESUMEN

As a common hypertensive complication of pregnancy, preeclampsia (PE) remains one of the leading causes of maternal and fetal with high morbidity and mortality worldwide. Much research has identified the vital functions of insulin-like growth factor 1 (IGF-1) in PE treatment. However, the combined roles and molecular mechanism of IGF-1 and microRNAs (miRNAs) underlying PE remain unclear. Therefore, we first measured the expression of IGF-1, zinc finger E-box binding homeobox 1 (ZEB1) and microRNA-183 (miR-183) expression in the placenta tissues of patients with PE by Western blot analysis and RT-qPCR. Interactions among IGF-1, ZEB1 and miR-183 were assessed by Western blot analysis, ChIP-PCR and dual-luciferase reporter gene assay. The effect of IGF-1 on the biological characteristics of trophoblast cells was investigated by CCK-8, colony formation assay and in vitro angiogenesis experiments after cells were transfected with si-IGF-1. Finally, a mouse eclampsia model induced by knockdown of IGF-1 was established to confirm the in vitro effect of IGF-1 on PE. We found that IGF-1, ZEB1 and miR-183 were highly expressed in the placental tissues of patients with PE. The knockdown of IGF-1 resulted in reduced proliferation and invasion of trophoblast cells and was accompanied by inhibited angiogenesis. ZEB1 was positively regulated by IGF-1 via ERK/MAPK pathway, which in turn inhibited miR-153 expression by binding to the miR-183 promoter. The in vitro experiments further confirmed that IGF-1 knockdown could induce PE. To sum up, IGF-1 knockdown elevated expression of miR-183 by downregulating ZEB1, thereby promoting deterioration of PE.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , MicroARNs , Preeclampsia , Animales , Femenino , Humanos , Ratones , Embarazo , Apoptosis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Genes Homeobox , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Placenta/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Regulación hacia Arriba/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Dedos de Zinc
6.
Exp Ther Med ; 25(3): 106, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36778043

RESUMEN

Accumulating evidence shows that long non-coding RNAs (lncRNAs) are widely involved in cellular processes of myocardial ischemia/reperfusion (I/R). The present study investigated the functions of lncRNA SNHG16 in myocardial I/R and the mechanism mediated by SNHG16. The myocardial I/R rat and cell model and hypoxia/reoxygenation injury (H/R) models of H9C2 cardiomyocytes were established to detect the expression of SNHG16. Cell Counting Kit-8, flow cytometric and western blot assays were conducted to detect cell viability, apoptosis and protein expression. Myocardial cell apoptosis was assessed by TUNEL staining. Dual-luciferase gene reporter was applied to determine the interaction between the molecules. The expressions of SNHG16 were upregulated in myocardial I/R injury models. Inhibition of SNHG16 relieved myocardial I/R injury in vivo and in vitro silencing of SNHG16 alleviated H/R induced cardiomyocyte apoptosis. To explore the regulatory mechanism, it was discovered that SNHG16 directly interacted with miR-183, while forkhead box O1 (FoxO1) was a target of microRNA (miR)-183. Findings from rescue assays revealed that miR-183 inhibitor and upregulation of FOXO1 can rescue the effect of sh-SNHG16 on H/R-induced cardiomyocyte apoptosis. The results indicated that the lncRNA SNHG16/miR-183/FOXO1 axis exacerbated myocardial cell apoptosis in myocardial I/R injury, suggesting SNHG16 as a potential therapeutic target for myocardial I/R injury.

7.
Genes Dev ; 36(9-10): 582-600, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35654454

RESUMEN

One of the mechanisms by which cancer cells acquire hyperinvasive and migratory properties with progressive loss of epithelial markers is the epithelial-to-mesenchymal transition (EMT). We have previously reported that in different cancer types, including nonsmall cell lung cancer (NSCLC), the microRNA-183/96/182 cluster (m96cl) is highly repressed in cells that have undergone EMT. In the present study, we used a novel conditional m96cl mouse to establish that loss of m96cl accelerated the growth of Kras mutant autochthonous lung adenocarcinomas. In contrast, ectopic expression of the m96cl in NSCLC cells results in a robust suppression of migration and invasion in vitro, and tumor growth and metastasis in vivo. Detailed immune profiling of the tumors revealed a significant enrichment of activated CD8+ cytotoxic T lymphocytes (CD8+ CTLs) in m96cl-expressing tumors, and m96cl-mediated suppression of tumor growth and metastasis was CD8+ CTL-dependent. Using coculture assays with naïve immune cells, we show that m96cl expression drives paracrine stimulation of CD8+ CTL proliferation and function. Using tumor microenvironment-associated gene expression profiling, we identified that m96cl elevates the interleukin-2 (IL2) signaling pathway and results in increased IL2-mediated paracrine stimulation of CD8+ CTLs. Furthermore, we identified that the m96cl modulates the expression of IL2 in cancer cells by regulating the expression of transcriptional repressors Foxf2 and Zeb1, and thereby alters the levels of secreted IL2 in the tumor microenvironment. Last, we show that in vivo depletion of IL2 abrogates m96cl-mediated activation of CD8+ CTLs and results in loss of metastatic suppression. Therefore, we have identified a novel mechanistic role of the m96cl in the suppression of lung cancer growth and metastasis by inducing an IL2-mediated systemic CD8+ CTL immune response.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Animales , Linfocitos T CD8-positivos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Interleucina-2/genética , Interleucina-2/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Linfocitos T Citotóxicos , Microambiente Tumoral
8.
Exp Neurol ; 356: 114151, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35738418

RESUMEN

BACKGROUND: New data are accumulating on the effects of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) in cerebrovascular diseases. We explored the potential role of KLF3-AS1-containing bone marrow MSC-EVs (BMSC-EVs) in a rat model of subarachnoid hemorrhage (SAH). METHODS: A rat model of SAH was established by endovascular perforation method, into which KLF3-AS1-containing EVs from BMSCs or miR-183-5p mimic were injected. Further, brain microvascular endothelial cells (BMECs) were induced by oxyhemoglobin (OxyHb) to simulate in vitro setting, which were co-cultured with KLF3-AS1-containing EVs from BMSCs. Effects of KLF3-AS1 on neurological deficits in vivo and endothelial cell dysfunction in vitro were investigated. We also performed bioinformatics analysis to predict downstream factors miR-183-5p and TCF7L2, which were verified by RIP, RNA pull-down and luciferase activity assays. RESULTS: BMSC-EVs was demonstrated to alleviate neurological deficits in SAH rats and endothelial cell dysfunction in OxyHb-induced BMECs. In addition, BMSC-EVs were shown to deliver KLF3-AS1 to BMECs, where KLF3-AS1 bound to miR-183-5p and miR-183-5p targeted TCF7L2. In vivo results confirmed that BMSC-EVs regulated the KLF3-AS1/miR-183-5p/TCF7L2 signaling axis to attenuate neurological deficit and endothelial dysfunction after SAH. CONCLUSION: Overall, KLF3-AS1 delivered by BMSC-EVs upregulate TCF7L2 expression by binding to miR-138-5p, thus attenuating neurological deficits and endothelial dysfunction after SAH.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , MicroARNs , Hemorragia Subaracnoidea , Animales , Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Ratas , Hemorragia Subaracnoidea/metabolismo
9.
Genes Genomics ; 44(8): 1017-1029, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35190998

RESUMEN

BACKGROUND: Liver regeneration is a highly orchestrated process concerning the modulation of various microRNAs (miRs). miR-183 was recently found to be involved in the process of liver regeneration, that miR-183 was remarkably up-regulated at 2-6 h after partial hepatectomy. OBJECTIVE: This study was aimed to explore the mechanism of miR-183 in on liver regeneration. METHODS: After partial hepatectomy (PH) or transfection, we measured the changes of miR-183 and programmed cell death protein 6 (PDCD6) levels in rats and the hepatocytes. The histopathology was observed with hematoxylin-eosin staining. The miR-183 mimic and inhibitor plasmids were intravenously injected into rats, and the liver weight/body weight ratio was calculated. The prediction of TargetScan and the validation of luciferase activity assay were employed to confirm the targeting relationship between miR-183 and PDCD6. The viability, apoptosis and cell cycle of transfected rat hepatocyte BRL-3A were determined via MTT and flow cytometry assays. RESULTS: MiR-183 expression showed a contrary tendency with that of PDCD6 during liver regeneration. Enhanced miR-183 in rats could notably increase liver/body weight ratio, while its inhibition did conversely. Overexpressed PDCD6, a target of miR-183, repressed the viability and cell cycle in hepatocytes, whereas its silence led to contrary results. Overexpressed miR-183 in BRL-3A cells enhanced cell viability and promoted the cell cycle yet suppressed apoptosis, whereas its inhibition showed contrary results, which were offset by PDCD6. CONCLUSIONS: Collectively, miR-183 promoted liver regeneration via targeting PDCD6.


Asunto(s)
Regeneración Hepática , MicroARNs/genética , Animales , Proteínas Reguladoras de la Apoptosis , Peso Corporal , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Regeneración Hepática/genética , MicroARNs/metabolismo , Ratas
10.
Bone Joint Res ; 10(8): 548-557, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34463129

RESUMEN

AIMS: MicroRNA-183 (miR-183) is known to play important roles in osteoarthritis (OA) pain. The aims of this study were to explore the specific functions of miR-183 in OA pain and to investigate the underlying mechanisms. METHODS: Clinical samples were collected from patients with OA, and a mouse model of OA pain was constructed by surgically induced destabilization of the medial meniscus (DMM). Reverse transcription quantitative polymerase chain reaction was employed to measure the expression of miR-183, transforming growth factor α (TGFα), C-C motif chemokine ligand 2 (CCL2), proinflammatory cytokines (interleukin (IL)-6, IL-1ß, and tumour necrosis factor-α (TNF-α)), and pain-related factors (transient receptor potential vanilloid subtype-1 (TRPV1), voltage-gated sodium 1.3, 1.7, and 1.8 (Nav1.3, Nav1.7, and Nav1.8)). Expression of miR-183 in the dorsal root ganglia (DRG) of mice was evaluated by in situ hybridization. TGFα, CCL2, and C-C chemokine receptor type 2 (CCR2) levels were examined by immunoblot analysis and interaction between miR-183 and TGFα, determined by luciferase reporter assay. The extent of pain in mice was measured using a behavioural assay, and OA severity assessed by Safranin O and Fast Green staining. Immunofluorescent staining was conducted to examine the infiltration of macrophages in mouse DRG. RESULTS: miR-183 was downregulated in tissue samples from patients and mice with OA. In DMM mice, overexpression of miR-183 inhibited the expression of proinflammatory cytokines (IL-6, IL-1ß, TNF-α) and pain-related factors (TRPV1, Nav1.3, Nav1.7, Nav1.8) in DRG. OA pain was relieved by miR-183-mediated inhibition of macrophage infiltration, and dual luciferase reporter assay demonstrated that miR-183 directly targeted TGFα. CONCLUSION: Our data demonstrate that miR-183 can ameliorate OA pain by inhibiting the TGFα-CCL2/CCR2 signalling axis, providing an excellent therapeutic target for OA treatment. Cite this article: Bone Joint Res 2021;10(8):548-557.

11.
Cancer Cell Int ; 21(1): 145, 2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33653339

RESUMEN

BACKGROUND: Emerging evidence continues to highlight the significant role of microRNAs (miRNAs) in the regulation of cancer growth and metastasis. Herein, the current study aimed to elucidate the role of exosomal miR-183 in prostate cancer development. METHODS: Initially, public microarray-based gene expression profiling of prostate cancer was employed to identify differentially expressed miRNAs. The putative target gene TPM1 of miR-183 was subsequently predicted, followed by the application of a luciferase reporter assay and examination of the expression patterns in prostate cancer patients and cell lines. The effects of miR-183 and TPM1 on processes such as cell proliferation, invasion and migration were evaluated using in vitro gain- and loss-of-function experiments. The effect of PC3 cells-derived exosomal miR-183 was validated in LNCaP cells. In vivo experiments were also performed to examine the effect of miR-183 on prostate tumor growth. RESULTS: High expression of miR-183 accompanied with low expression of TPM1 was detected in prostate cancer. Our data indicated that miR-183 could target and downregulate TPM1, with the overexpression of miR-183 and exosomal miR-183 found to promote cell proliferation, migration, and invasion in prostate cancer. Furthermore, the tumor-promoting effect of exosome-mediated delivery of miR-183 was subsequently confirmed in a tumor xenograft model. CONCLUSIONS: Taken together, the key findings of our study demonstrate that prostate cancer cell-derived exosomal miR-183 enhance prostate cancer cell proliferation, invasion and migration via the downregulation of TPM1, highlighting a promising therapeutic target against prostate cancer.

12.
Exp Ther Med ; 21(4): 300, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33717243

RESUMEN

Atherosclerosis is considered a chronic inflammatory disease, and macrophages function as important mediators in the development of atherogenesis. MicroRNA (miR)-183 is a small non-coding RNA that acts as a novel tumor suppressor and has recently been proposed to affect cardiac hypertrophy. However, the exact role and underlying mechanism of miR-183 in macrophage activation remain unknown. In the present study, miR-183 showed upregulated expression in atheromatous plaques and in bone marrow-derived macrophages (BMDMs) subjected to stimulation with oxidized low-density lipoproteins. Using a miR-183 loss-of-function strategy, it was demonstrated that miR-183 knockdown significantly increased resolving M2 macrophage marker expression but decreased proinflammatory M1 macrophage marker expression, as well as attenuated NF-κB activation. Moreover, decreased foam-cell formation accompanied by upregulation of genes involved in cholesterol efflux and downregulation of genes implicated in cholesterol influx was found in BMDMs transfected with a miR-183 inhibitor. Mechanistically, macrophage activation mediated by miR-183 silencing was partially attributed to direct upregulation of NR4A2 expression in BMDMs. Thus, the present study suggests that neutralizing miR-183 may be a potential therapeutic strategy for the treatment of atherosclerosis.

13.
Life Sci ; 269: 119017, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33450262

RESUMEN

BACKGROUND/AIM: Emerging studies have revealed the mechanism of microRNAs (miRNAs) in acute kidney injury (AKI). However, almost no research focuses on miR-183-3p in AKI. Therefore, this study is started to explore the potential role of miR-183-3p from the perspective of Sirtuin (SIRT1)/forkhead box O3a (FOXO3a)/p53 up-regulated modulator of apoptosis (PUMA) in AKI. METHODS: AKI rat models were established. miR-183-3p, SIRT1, deacetylated FOXO3a and PUMA expression in AKI were detected. The targeting relationship between miR-183-3p and SIRT1 was tested. AKI modeled rats were injected with miR-183-3p- or SIRT1-related sequences to identify their effects on AKI. Rat renal tubular epithelial cells NRK-52E were transfected with miR-183-3p- or SIRT1-related sequences for further exploration of their roles in vitro. RESULTS: Decreased SIRT1 and deacetylated FOXO3a and increased miR-183-3p and PUMA were found in AKI. SIRT1 was targeted by miR-183-3p. Down-regulated miR-183-3p or up-regulated SIRT1 attenuated renal tissue damage and fibrosis, and suppressed renal tubular epithelial cell apoptosis in renal tissues of AKI rats. Down-regulated miR-183-3p or up-regulated SIRT1 promoted proliferation and impaired fibrosis and apoptosis of renal tubular epithelial cells. CONCLUSION: Our study provides evidence that down-regulated miR-183-3p or up-regulated SIRT1 attenuates AKI via PUMA inhibition induced by FOXO3a deacetylation. Thus, miR-183-3p and SIRT1 can be the candidates for therapy of AKI.


Asunto(s)
Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína Forkhead Box O3/metabolismo , MicroARNs/metabolismo , Sirtuina 1/metabolismo , Acetilación , Animales , Apoptosis , Secuencia de Bases , Línea Celular , Proliferación Celular , Regulación hacia Abajo/genética , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Fibrosis , Silenciador del Gen , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , MicroARNs/genética , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/genética
14.
Am J Transl Res ; 12(10): 6250-6263, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194027

RESUMEN

BACKGROUND: Cerebral ischemia/reperfusion (I/R) injury is a severe complication during the treatment of patients with stroke. It has been shown that the expression of SNHG15 was increased in patients with ischemic stroke (IS). However, the function and regulatory mechanism of SNHG15 in IS remains unclear. METHODS: An oxygen glucose deprivation/reoxygenation (OGD/R) cell model was use to establish an in vitro model of I/R injury. RT-qPCR assay was used to detect the level of SNHG15 in OGD/R-treated SH-SY5Y cells. Meanwhile, middle cerebral artery occlusion (MCAO) was used to establish an in vivo model of cerebral I/R injury. RESULTS: The expression of SNHG15 was upregulated in OGD/R-treated SH-SY5Y cells. Downregulation of SNHG15 during reperfusion reduced cell death in OGD/R-treated SH-SY5Y cells. In addition, SNHG15 knockdown suppressed OGD/R-induced apoptosis in SY-SY5Y cells by attenuating intracellular ROS generation and reducing mitochondrial membrane potential (MMP) lost. In addition, SNHG15 knockdown promoted cell cycle transition in SY-SY5Y cells after OGD/R insult accompany with PI3K/Akt signaling activation. Meanwhile, mechanism investigations suggested SNHG15 knockdown downregulated the expression of FOXO1 through acting as a competitive 'sponge' of miR-183-5p. Most importantly, knockdown of SNHG15 expression in vivo inhibited neuronal apoptosis and decreased infarct area in MCAO rats. CONCLUSION: Thus, the present study indicated that SNHG15 knockdown protected against cerebral I/R injury via targeting miR-183-5p/FOXO1 axis, which may represent a potential therapeutic option for the treatment of cerebral IS.

15.
Mol Cell Biol ; 41(1)2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33139493

RESUMEN

Preeclampsia (PE) is a hypertensive disorder of uncertain etiology that is the leading cause of maternal and fetal morbidity or mortality. The dysregulation of microRNAs (miRNAs) has been highlighted as a potential factor involved in the development of PE. Therefore, our study investigated a novel miRNA, miRNA 183 (miR-183), and its underlying association with PE. Expression of miR-183, forkhead box P1 (FOXP1), and G protein subunit gamma 7 (GNG7) in placental tissues of patients with PE was determined. Gain- and loss-of-function experiments were conducted to explore modulatory effects of miR-183, FOXP1, and GNG7 on the viability, invasion, and angiogenesis of trophoblast cells in PE. Finally, we undertook in vivo studies to explore effects of FOXP1 in the PE model. The results revealed suppressed expression of FOXP1 and significant elevations in miR-183 and GNG7 expression in placental tissues of PE patients. FOXP1 was observed to promote proliferation, invasion, and angiogenesis in human chorionic trophoblastic cells. miR-183 resulted in depletion of FOXP1 expression, while FOXP1 was capable of restraining GNG7 expression and promoting the mTOR pathway. The findings confirmed the effects of FOXP1 on PE. In conclusion, miR-183 exhibits an inhibitory role in PE through suppression of FOXP1 and upregulation of GNG7.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Subunidades gamma de la Proteína de Unión al GTP/biosíntesis , MicroARNs/metabolismo , Preeclampsia/metabolismo , Proteínas Represoras/biosíntesis , Adulto , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Expresión Génica , Humanos , Ratones , MicroARNs/genética , Placenta/metabolismo , Preeclampsia/genética , Preeclampsia/patología , Embarazo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patología
16.
Biosci Rep ; 40(10)2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33078826

RESUMEN

Hepatocellular carcinoma (HCC) remains one of the most common malignant tumors worldwide. The present study aimed to investigate the biological role of microRNA-183-5p (miR-183-5p), a novel tumor-related microRNA (miRNA), in HCC and illuminate the possible molecular mechanisms. The expression patterns of miR-183-5p in clinical samples were characterized using qPCR analysis. Kaplan-Meier survival curve was applied to evaluate the correlation between miR-183-5p expression and overall survival of HCC patients. Effects of miR-183-5p knockdown on HCC cell proliferation, apoptosis, migration and invasion capabilities were determined via Cell Counting Kit-8 (CCK8) assays, flow cytometry, scratch wound healing assays and Transwell invasion assays, respectively. Mouse neoplasm transplantation models were established to assess the effects of miR-183-5p knockdown on tumor growth in vivo. Bioinformatics analysis, dual-luciferase reporter assays and rescue assays were performed for mechanistic researches. Results showed that miR-183-5p was highly expressed in tumorous tissues compared with adjacent normal tissues. Elevated miR-183-5p expression correlated with shorter overall survival of HCC patients. Moreover, miR-183-5p knockdown significantly suppressed proliferation, survival, migration and invasion of HCC cells compared with negative control treatment. Consistently, miR-183-5p knockdown restrained tumor growth in vivo. Furthermore, programmed cell death factor 4 (PDCD4) was identified as a direct target of miR-183-5p. Additionally, PDCD4 down-regulation was observed to abrogate the inhibitory effects of miR-183-5p knockdown on malignant phenotypes of HCC cells. Collectively, our data suggest that miR-183-5p may exert an oncogenic role in HCC through directly targeting PDCD4. The current study may offer some new insights into understanding the role of miR-183-5p in HCC.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroARNs/metabolismo , Proteínas de Unión al ARN/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/cirugía , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Estudios de Cohortes , Biología Computacional , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hepatectomía , Humanos , Estimación de Kaplan-Meier , Hígado/patología , Hígado/cirugía , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/cirugía , Masculino , Ratones , Persona de Mediana Edad , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Stem Cells Cloning ; 13: 79-89, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32982315

RESUMEN

INTRODUCTION: Hearing Loss (HL) is known as the most common sensory processing disorder across the world. An effective treatment which has been currently used for patients suffering from this condition is cochlear implant (CI). The major limitation of this treatment is the need for a healthy auditory neuron (AN). Accordingly, mesenchymal cells (MCs) are regarded as good candidates for cell-based therapeutic approaches. The present study aimed to investigate the potentials of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) for differentiation towards ANs along with using treatments with growth factors and microRNA (miRNA) transfection in vitro. METHODS: To this end, neurospheres derived from hBM-MSCs were treated via basic fibroblast growth factor (bFGF), neurotrophin-3 (NT-3), and brain-derived neurotrophic factor (BDNF) as growth factors N2 and B27 supplements, as well as miRNA-96, -182, -183 transfected into hBM-MSCs in order to evaluate the differentiation of such cells into ANs. RESULTS: Treatments with growth factors demonstrated a significant increase in neurogenin 1 (Ngn1) and sex determining region Y-box 2 (SOX2) markers; but tubulin, microtubule-associated protein 2 (MAP2), and GATA binding protein 3 (GATA3) markers were not statistically significant. The findings also revealed that miRNA-182 expression in miRNA-183 family could boost the expressions of some AN marker (ie, Ngn1, SOX2, peripherin, and nestin) in vitro. DISCUSSION: It can be concluded that miRNA is probably a good substitute for growth factors used in differentiating into ANs. Transdifferentiation of hBM-MSCs into ANs, which does not occur under normal conditions, may be thus facilitated by miRNAs, especially miRNA-182, or via a combination of miRNA and growth factors.

18.
Aging (Albany NY) ; 12(13): 12869-12895, 2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32602850

RESUMEN

Differences in microRNA (miRNA) expression after intracerebral hemorrhage (ICH) have been reported in human and animal models, and miRNAs are being investigated as a new treatment for inflammation and oxidative stress after ICH. In this study, we found that microRNA-183-5p expression was decreased in the mouse brain after ICH. To investigate the effect of miRNA-183-5p on injury and repair of brain tissue after ICH, saline, miRNA-183-5p agomir, or miRNA-183-5p antagomir were injected into the lateral ventricles of 8-week-old mice with collagenase-induced ICH. Three days after ICH, mice treated with exogenous miRNA-183-5p showed less brain edema, neurobehavioral defects, inflammation, oxidative stress, and ferrous deposition than control mice. In addition, by alternately treating mice with a heme oxygenase-1 (HO-1) inducer, a HO-1 inhibitor, a nuclear factor erythroid 2-related factor (Nrf2) activator, and Nrf2 knockout, we demonstrated an indirect, HO-1-dependent regulatory relationship between miRNA-183-5p and Nrf2. Our results indicate that miRNA-183-5p and HO-1 are promising therapeutic targets for controlling inflammation and oxidative damage after hemorrhagic stroke.


Asunto(s)
Hemorragia Cerebral/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Hemo-Oxigenasa 1/análisis , Hemo-Oxigenasa 1/genética , Inflamación , Masculino , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL
19.
J Neuroinflammation ; 17(1): 225, 2020 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32723328

RESUMEN

BACKGROUND: Neuropathic pain is related to the sustained activation of neuroglial cells and the production of proinflammatory cytokines in the spinal dorsal horn. However, the clinical efficacy of currently available treatments is very limited. The transcription factor nuclear factor κB (NF-κB) is a ubiquitously expressed protein family and considered to be crucial in autoimmunity. Thus, our study aimed to examine the influence of NF-κB p65 in chronic constriction injury (CCI)-induced neuropathic pain as well as its underlying mechanism. METHODS: A rat model of neuropathic pain was established by CCI induction followed by isolation of microglial cells. The binding of NF-κB p65 to HDAC2, of miR-183 to TXNIP, and of TXNIP to NLRP3 was investigated. Expression of miR-183, NF-κB p65, HDAC2, TXNIP, and NLRP3 was determined with their functions in CCI rats and microglial cells analyzed by gain- and loss-of-function experiments. RESULTS: NF-κB p65 and HDAC2 were upregulated while miR-183 was downregulated in the dorsal horn of the CCI rat spinal cord. NF-κB p65 was bound to the HDAC2 promoter and then increased its expression. HDAC2 reduced miR-183 expression by deacetylation of histone H4. Additionally, miR-183 negatively regulated TXNIP. Mechanistically, NF-κB p65 downregulated the miR-183 expression via the upregulation of HDAC2 and further induced inflammatory response by activating the TXNIP-NLRP3 inflammasome axis, thus aggravating the neuropathic pain in CCI rats and microglial cells. CONCLUSION: These results revealed a novel transcriptional mechanism of interplay between NF-κB and HDAC2 focusing on neuropathic pain via the miR-183/TXNIP/NLRP3 axis.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Histona Desacetilasa 2/biosíntesis , Neuralgia/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas de Ciclo Celular/metabolismo , Constricción Patológica , Ligadura , Masculino , MicroARNs/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas , Ratas Sprague-Dawley , Nervio Ciático/lesiones , Factor de Transcripción ReIA/metabolismo
20.
Aging (Albany NY) ; 12(9): 8352-8371, 2020 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-32364530

RESUMEN

Exosomes play important roles in proliferation and microenvironment modulation of many types of cancers, including colorectal cancer (CRC). However, the inhibitory effect of CRC cells-derived exosomes in angiogenesis has not been fully discussed. In this study, the roles of microRNA-183-5p (miR-183-5p) in abundant in exosomes secreted from the CRC cells were investigated. Initially, microarray analysis was employed to determine the differentially expressed miRNAs. Exosomes isolated from CRC cells were co-cultured with HMEC-1 cells to explore the role of exosomes in angiogenesis. Further, the effects of CRC cell-derived exosomal miR-183-5p on proliferation, invasion and tube formation abilities of HMEC-1 cells were assessed. The preventative effect of exosomal miR-183-5p in vivo was measured in nude mice. Initially, it was found that FOXO1 was downregulated while miR-183-5p was upregulated in CRC. Additionally, the inhibition of miR-183-5p was suggested to suppress proliferation, invasion and tube formation abilities of HMEC-1 cells through upregulating FOXO1. Then, in vitro assays demonstrated that CRC cell-derived exosomes overexpressing miR-183-5p contributed to an enhanced proliferation, invasion and tube formation abilities of HMEC-1 cells. Furthermore, in vivo experiments confirmed the tumor-promotive effects of CRC cell-derived exosomal miR-183-5p. Collectively, our study demonstrates that the CRC cell-derived exosomes overexpressing miR-183-5p aggravates CRC through the regulation of FOXO1. Exosomes overexpressing miR-183-5p might be a potential treatment biomarker for CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Proteína Forkhead Box O1/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Neovascularización Patológica/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación hacia Abajo , Exosomas/metabolismo , Proteína Forkhead Box O1/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA