Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Cell Mol Med ; 26(4): 977-989, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35014178

RESUMO

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression at the post-transcriptional level. MiRNAs are involved in the development and progression of a wide range of cancers. Among such cancer-associated miRNAs, miR-381 has been a major focus of research. The expression pattern and role of miR-381 vary among different cancer types. MiR-381 modulates various cellular behaviours in cancer, including proliferation, apoptosis, cell cycle progression, migration and invasion. MiR-381 is also involved in angiogenesis and lymphangiogenesis, as well as in the resistance to chemotherapy and radiotherapy. MiR-381 itself is regulated by several factors, such as long noncoding RNAs, circular RNAs and cytokines. Aberrant expression of miR-381 in blood samples indicates that it can be used as a diagnostic marker in cancer. Tissue miR-381 expression may serve as a prognostic factor for the clinicopathological characteristics of cancers and survival of patients. Metformin and icaritin regulate miR-381 expression and present anticancer properties. This review comprehensively summarizes the effect of miR-381 on tumour biological behaviours, as well as the clinical application potential of miR-381 for the treatment of cancer.


Assuntos
MicroRNAs , Neoplasias , RNA Longo não Codificante , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Neoplasias/patologia , RNA Circular
2.
J Cell Mol Med ; 25(4): 2098-2109, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33314611

RESUMO

Dexmedetomidine (Dex) possesses analgesic and anaesthetic values and reported being used in cerebral ischaemic injury therapeutics. Accumulating studies have determined the effect of microRNAs (miRNAs) on the cerebral ischaemic injury. Thus, the present study aimed to unravel the molecular mechanism of miR-381 and Dex in cerebral ischaemic injury. For this purpose, the cerebral ischaemic injury rat model was established by induction of middle cerebral artery occlusion (MCAO) and expression of miR-381 and IRF4 was determined. Thereafter, MCAO rats were treated with Dex, miR-381 mimic, miR-381 inhibitor and oe-IRF4 respectively, followed by evaluation of neurological function. Furthermore, neuron cells were isolated from the hippocampus of rats and subjected to oxygen-glucose deprivation (OGD). Then, OGD-treated neuron cells and primary neuron cells were examined by gain- and loss-of-function assay. Neuron cell apoptosis was detected using TUNEL staining and flow cytometry. The correlation between interferon regulatory factor 4 (IRF4) and interleukin (IL)-9 was detected. Our results showed down-regulated miR-38 and up-regulated IRF4 in MCAO rats. Besides, IRF4 was targeted by miR-381 in neuron cells. Dex and overexpressed miR-381, or silenced IRF4 improved the neurological function and inhibited neuron cell apoptosis in MCAO rats. Additionally, in MCAO rats, Dex was found to increase the miR-381 expression and reduced IRF4 expression to decrease the IL-9 expression, which suppressed the inflammatory response and cell apoptosis both in vivo and in vitro. Importantly, our study demonstrated that Dex elevated the expression of miR-381, which ultimately results in the inhibition of inflammation response in rats with cerebral ischaemic injury.


Assuntos
Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Dexmedetomidina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fatores Reguladores de Interferon/metabolismo , MicroRNAs/genética , Animais , Apoptose/efeitos dos fármacos , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Glucose/metabolismo , Fatores Reguladores de Interferon/genética , Interleucina-9/genética , Interleucina-9/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oxigênio/metabolismo , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Interferência de RNA , Ratos , Transdução de Sinais/efeitos dos fármacos
3.
J Biomed Sci ; 28(1): 53, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34266430

RESUMO

OBJECTIVE: Researchers have revealed the combined functions of long noncoding RNAs (lncRNAs) and microRNA (miRNAs) in polycystic ovary syndrome (PCOS). This study aimed to understand the role of nuclear-enriched abundant transcript 1 (NEAT1) and miR-381 involving insulin-like growth factor 1 (IGF1) in PCOS. METHODS: PCOS rat model was established by dehydroepiandrosterone induction. NEAT1, miR-381 and IGF1 expression in ovarian granulosa cells of PCOS patients and ovarian tissues of PCOS rats were tested. Bioinformatics website and dual luciferase reporter gene assay were utilized to verify the relationship between NEAT1 and miR-381 and that between miR-381 and IGF1. Levels of sex hormone, pathological changes and ovarian granulosa cell apoptosis in ovarian tissues of PCOS rats were detected. Ovarian granulosa cell proliferation and apoptosis were analyzed in vitro. RESULTS: NEAT1 and IGF1 expression increased while miR-381 expression decreased in the ovarian granulosa cells of patients with PCOS and the ovarian tissues of PCOS rats. In in vivo experiments, interference with NEAT1 improved the levels of sex hormones, alleviated pathological changes and suppressed ovarian granulosa cell apoptosis in the ovarian tissues of PCOS rats. In in vitro cell experiments, interference with NEAT1 suppressed apoptosis and enhanced cell proliferation of ovarian granulosa cells. NEAT1 interference-mediated effect would be reversed by up-regulating miR-381. NEAT1 acted as a ceRNA to adsorb miR-381 to target IGF1. Overexpression of IGF1 reversed the inhibitory effect of miR-381 on ovarian granulosa cell apoptosis. CONCLUSION: Interference with NEAT1 increases miR-381 and reduces IGF1 levels, effectively improving the levels of sex hormones and reducing the pathological damage of ovarian tissue in rats with PCOS.


Assuntos
Apoptose/genética , Proliferação de Células/genética , Regulação para Baixo , MicroRNAs/genética , Síndrome do Ovário Policístico/genética , RNA Longo não Codificante/genética , Animais , Feminino , Células da Granulosa/metabolismo , MicroRNAs/metabolismo , Ovário/metabolismo , Síndrome do Ovário Policístico/metabolismo , RNA Longo não Codificante/metabolismo , Ratos , Ratos Wistar
4.
Cell Mol Neurobiol ; 40(8): 1307-1319, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32297103

RESUMO

Ischemic stroke is a serious disease with limited prevention methods, and various genes and microRNAs (miRNAs) have been found to be dysregulated in the pathogenesis of this disease. This study aims to explore the potential role of miR-381-3p in ischemic stroke, along with its underlying mechanism. A mouse model of ischemic stroke was developed using middle cerebral artery occlusion. Next, the expression of mitogen-activated protein kinase kinase kinase 8 (Map3k8) and CCAAT enhancer binding protein beta (Cebpb) was determined by RT-qPCR. Gain- and loss-of-function approaches were applied to analyze the effects of miR-381-3p, Cebpb and Map3k8 on the biological functions of endothelial progenitor cells (EPCs) with the involvement of the tumor necrosis factor-α (TNF-α) signaling pathway. In addition, dual luciferase reporter gene assay was performed for the analysis of the relationship among miR-381-3p, Map3k8 and Cebpb. Further, rescue experiment was performed with the help of JNK/p38 specific agonist, Anisomycin. Map3k8 and Cebpb were highly expressed in ischemic stroke. Loss-of-function of Map3k8 or Cebpb in EPCs contributed to accelerated proliferation, migration and angiogenesis of EPCs. Next, miR-381-3p downregulated the expression of its two target genes, Map3k8 and Cebpb. miR-381-3p overexpression promoted angiogenesis of EPCs, and inhibited inflammation, which could be reversed by restoration of Map3k8 or Cebpb. Additionally, silencing Map3k8 or Cebpb inhibited the activation of TNF-α signaling pathway. Furthermore, Anisomycin treatment could enhance inflammation and inhibit angiogenesis. Taken together, miR-381-3p downregulates Map3k8 and Cebpb to protect against ischemic stroke, broadening our understanding of the pathogenesis of ischemic stroke.


Assuntos
Inflamação/metabolismo , AVC Isquêmico/metabolismo , MAP Quinase Quinase Quinases/metabolismo , MicroRNAs/genética , Proteínas Proto-Oncogênicas/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica/genética , Inflamação/genética , AVC Isquêmico/genética , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
5.
J Cell Biochem ; 119(4): 3149-3161, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29073721

RESUMO

This study aims to explore whether microRNA-381 (miR-381) mediating CXCR4 affects the renal tubular epithelial cells (RTEC) of renal ischemia reperfusion (I/R) injury. Forty-eight rats were assigned into the I/R (n = 24, successfully established as I/R model) and sham (n = 24) groups. After collecting kidney tissues, immunohistochemistry, and microvascular density (MVD) counting were conducted for CXCR4 positive expression and MVD numbers. RTECs were assigned into the sham, blank, negative control (NC), miR-381 mimics, miR-381 inhibitor, si-CXCR4, and miR-381 inhibitor + si-CXCR4 groups. RT-qPCR and Western blotting were performed for relative expressions in tissues and cells. Cell proliferation and apoptosis were measured by MTT assay and flow cytometry. Results showed that compared with the sham group, positive expression of CXCR4 and MVD number were higher in the I/R group, which exhibited decreased miR-381 and increased expression of CXCR4, stromal cell-derived factor-1 (SDF1), vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1 (HIF-1α) and Tie-2. Dual luciferase reporter gene assay verified that CXCR4 is a target gene of miR-381. MiR-381 expression was lower in the miR-381 inhibitor + si-CXCR4 and miR-381 inhibitor groups and higher in the miR-381 mimics group than the blank and NC groups. Compared with the blank and NC groups, the miR-381 mimics and si-CXCR4 groups exhibited higher cell proliferation but lower cell apoptosis and expression of CXCR4, SDF1, VEGF, HIF-1α, and Tie-2, whereas the miR-381 inhibitor group exhibited the opposite trend. In conclusion, miR-381 may promote RTEC proliferation in rats with renal I/R injury by down-regulating CXCR4.


Assuntos
Injúria Renal Aguda/genética , Regulação para Baixo , Túbulos Renais/citologia , MicroRNAs/genética , Receptores CXCR4/genética , Traumatismo por Reperfusão/genética , Injúria Renal Aguda/metabolismo , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Células Epiteliais/citologia , Nefropatias , Masculino , Ratos , Receptores CXCR4/metabolismo , Traumatismo por Reperfusão/metabolismo
6.
Cell Physiol Biochem ; 46(3): 890-906, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29669322

RESUMO

BACKGROUND/AIMS: Acute cerebral ischemia is a manifestation of cerebral vascular insufficiency and has a high mortality. However, the therapy for acute cerebral ischemia is still limited. This study aimed to investigate the effect of microRNA-381 (miR-381) on the repair of nerve injury in rats with acute cerebral ischemia after cerebral lymphatic blockage (CLB) by targeting leucine-rich repeat C4 protein (LRRC4) through the Stromal cell-derived factor-1/CXC chemokine receptor-4 signaling pathway. METHODS: Rat models of CLB and middle cerebral artery occlusion (MCAO) were established, and 56 Wistar rats were divided into sham, MCAO, CLB + MCAO, CLB + MCAO + miR-381 inhibitor, CLB + MCAO + miR-381 mimic, CLB + MCAO + AMD3100 and CLB + MCAO + miR-381 mimic + AMD3100 groups. Modified neurological severity score (mNSS was used to determine nerve injury, TTC staining to measure infarction volume, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining and flow cytometry to evaluate cell apoptosis, immunofluorescence to measure BrdU-positive cell number, enzyme-linked immunosorbent assay (ELISA) to determine contents of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), interleukin-10 (IL-10), nerve growth factor (NGF) and neurite outgrowth inhibitor -A (Nogo-A), Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting to evaluate expression of miR-381, LRRC4, SDF-1, CXCR4, pERK, Slit2 and vascular endothelial growth factor (VEGF). RESULTS: LRRC4 was a target gene of miR-381. Compared with the results in the CLB + MCAO group, mNSS, infarction volume, apoptosis rate and TNF-α, IL-1ß, IL-6 and Nogo-A contents as well as LRRC4 expression in the CLB + MCAO + miR-381 inhibitor and CLB + MCAO + AMD3100 groups were increased (those in the CLB + MCAO + AMD3100 group > those in the CLB + MCAO + miR-381 mimic + AMD3100 group), while BrdU-positive cell number, contents of NGF and IL-10, and expression of SDF-1, CXCR4, pERK, Slit2 and VEGF in brain tissues were decreased (those in the CLB + MCAO + AMD3100 group < those in the CLB + MCAO + miR-381 mimic + AMD3100 group). The results in the CLB + MCAO + mimic group were opposite of those in the CLB + MCAO + miR-381 inhibitor and CLB + MCAO + AMD3100 groups. CONCLUSION: Taken together, we concluded that up-regulation of miR-381 promoted nerve injury repair in acute cerebral ischemia rats after CLB by negatively regulating LRRC4 through activating the SDF-1/CXCR4 signaling pathway.


Assuntos
Isquemia Encefálica/patologia , Quimiocina CXCL12/metabolismo , MicroRNAs/metabolismo , Proteínas/metabolismo , Receptores CXCR4/metabolismo , Animais , Benzilaminas , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Quimiocina CXCL12/genética , Ciclamos , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Hipocampo/patologia , Infarto da Artéria Cerebral Média/complicações , Interleucina-1beta/análise , Proteínas de Repetições Ricas em Leucina , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas/antagonistas & inibidores , Proteínas/genética , Ratos , Ratos Wistar , Receptores CXCR4/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/análise , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Int J Mol Sci ; 17(9)2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27563877

RESUMO

Chondrocyte hypertrophy, regulated by Runt-related transcription factor 2 (RUNX2) and matrix metalloproteinase 13 (MMP13), is a crucial step in cartilage degeneration and osteoarthritis (OA) pathogenesis. We previously demonstrated that microRNA-381 (miR-381) promotes MMP13 expression during chondrogenesis and contributes to cartilage degeneration; however, the mechanism underlying this process remained unclear. In this study, we observed divergent expression of miR-381 and histone deacetylase 4 (HDAC4), an enzyme that directly inhibits RUNX2 and MMP13 expression, during late-stage chondrogenesis of ATDC5 cells, as well as in prehypertrophic and hypertrophic chondrocytes during long bone development in E16.5 mouse embryos. We therefore investigated whether this miRNA regulates HDAC4 expression during chondrogenesis. Notably, overexpression of miR-381 inhibited HDAC4 expression but promoted RUNX2 expression. Moreover, transfection of SW1353 cells with an miR-381 mimic suppressed the activity of a reporter construct containing the 3'-untranslated region (3'-UTR) of HDAC4. Conversely, treatment with a miR-381 inhibitor yielded increased HDAC4 expression and decreased RUNX2 expression. Lastly, knockdown of HDAC4 expression resulted in increased RUNX2 and MMP13 expression in SW1353 cells. Collectively, our results indicate that miR-381 epigenetically regulates MMP13 and RUNX2 expression via targeting of HDAC4, thereby suggesting the possibilities of inhibiting miR-381 to control chondrocyte hypertrophy and cartilage degeneration.


Assuntos
Condrócitos/citologia , Condrócitos/metabolismo , Histona Desacetilases/metabolismo , Hipertrofia/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Linhagem Celular , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Histona Desacetilases/genética , Hipertrofia/genética , Imuno-Histoquímica , Hibridização In Situ , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , MicroRNAs/genética
8.
Mol Med Rep ; 30(3)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38994774

RESUMO

Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that the certain of the cell proliferation assay data shown in Fig. 4C on p. 1444 were strikingly similar to data appearing in different form in another article written by different authors at different research institutes, which had already been submitted for publication [Shi N, Shan B, Song Y, Chu H and Qian L: Circular RNA circ­PRKCI functions as a competitive endogenous RNA to regulate AKT3 expression by sponging miR­3680­3p in esophageal squamous cell carcinoma. J Cell Biochem 120: 10021­10030, 2019]. Owing to the fact that the contentious data in the above article were already under consideration for publication prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. 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. [Molecular Medicine Reports 21: 1439­1448, 2020; DOI: 10.3892/mmr.2020.10957].

9.
Zhongguo Zhen Jiu ; 44(2): 175-181, 2024 Feb 12.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38373763

RESUMO

OBJECTIVES: To investigate the effects of electroacupuncture (EA) on the miR-381, leucine-rich repeat C4 protein (LRRC4), and downstream stromal cell-derived factor-1 (SDF-1)/CXC chemokine receptor 4 (CXCR4) signaling pathway in rat model of ischemic stroke, and to explore the mechanism by which EA improves neurological damage following ischemic stroke. METHODS: Among 50 SPF male SD rats, 10 rats were randomly selected into a sham surgery group, and the remaining rats were used to establish the middle cerebral artery occlusion (MCAO) model. The 30 successfully modeled rats were randomly divided into a model group, an EA group, and an agonist group, with 10 rats in each group. The rats in the EA group received EA at "Baihui" (GV 20) and "Dazhui" (GV 14), with disperse-dense wave, a frequency of 2 Hz/10 Hz, and a current intensity of 1 mA, 30 min per session, once daily for a total of 14 days. The rats in the agonist group received miR-381 agonist injections into the lateral ventricle, with 10 µL per injection, every 7 days for a total of 2 injections. After intervention, ZeaLonga neurobehavioral deficit score was observed in each group. HE staining was performed to observe the morphological changes in the ischemic brain tissue of rats in each group. ELISA was used to measure the levels of tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and nerve growth factor (NGF) in serum. Western blot was employed to detect the protein expression of LRRC4, SDF-1, CXCR4, and extracellular regulated protein kinase 1 (ERK1) in the ischemic brain tissue. Real-time PCR was utilized to assess the expression of miR-381 and LRRC4, SDF-1, CXCR4, ERK1 mRNA in the ischemic brain tissue. RESULTS: After intervention, the brain tissue showed disordered cell arrangement, reduced quantity, and significant interstitial edema, with numerous vacuoles in the model group. The pathological changes mentioned above were alleviated in the brain tissue of rats in the EA group and the agonist group. Compared with the sham surgery group, the rats in the model group exhibited increased ZeaLonga neurobehavioral deficit scores, elevated levels of serum TNF-α and IL-6 (P<0.01), and decreased serum NGF level (P<0.01);the protein expression of SDF-1, CXCR4 and ERK1 in ischemic brain tissue was reduced (P<0.01), while LRRC4 protein expression was increased (P<0.01);the expression of miR-381, as well as SDF-1, CXCR4 and ERK1 mRNA in ischemic brain tissue was decreased (P<0.01), while LRRC4 mRNA expression was increased (P<0.01). Compared with the model group, the rats in the EA group and the agonist group showed decreased ZeaLonga neurobehavioral deficit scores and reduced levels of serum TNF-α and IL-6 (P<0.05, P<0.01), and increased serum NGF levels (P<0.05, P<0.01); the protein expression of SDF-1, CXCR4 and ERK1 in ischemic brain tissue was increased (P<0.01), while LRRC4 protein expression was decreased (P<0.01);the expression of miR-381, as well as SDF-1, CXCR4 and ERK1 mRNA in ischemic brain tissue was increased (P<0.05, P<0.01), while LRRC4 mRNA expression was decreased (P<0.01). CONCLUSIONS: EA at "Baihui" (GV 20) and "Dazhui" (GV 14) may promote the repair of neurological damage following ischemic stroke by up-regulating miR-381 to selectively inhibit LRRC4 expression, thereby activating the SDF-1/CXCR4 signaling pathway.


Assuntos
Isquemia Encefálica , Eletroacupuntura , AVC Isquêmico , MicroRNAs , Ratos , Masculino , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Isquemia Encefálica/metabolismo , Ratos Sprague-Dawley , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Fator de Necrose Tumoral alfa/genética , Interleucina-6 , Fator de Crescimento Neural , Transdução de Sinais , MicroRNAs/genética , RNA Mensageiro
10.
Oncol Rep ; 50(5)2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37800629

RESUMO

Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that certain of the western blotting data shown in Fig. 5C, and the cell migration and invasion data shown in Figs. 3C and D and 6B and C were strikingly similar to data that had already appeared in other articles. Owing to the fact that the contentious data in the above article had already been published prior to its submission to Oncology Reports, the Editor has decided that this paper should be retracted from the Journal. 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. [Oncology Reports 38: 3071­3077, 2017; DOI: 10.3892/or.2017.5956].

11.
ACS Chem Neurosci ; 14(11): 1981-1991, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37166273

RESUMO

As a common and disabling disease of the elderly, the standard therapies of Parkinson's disease (PD) fail to curb the ongoing neurodegeneration, thus calling for newer strategies. This work was conducted to examine the effect of microRNA-381 (miR-381) on oxidative stress injury to dopaminergic neurons in PD in vivo and in vitro. We established an in vivo mouse model of PD using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and an in vitro cell model of PD by treating dopaminergic neuron MN9D cells with 1-methyl-4-phenylpyridinium (MPP+). It was established that miR-381 was poorly expressed in the substantia nigra pars compacta (SNc) of MPTP-lesioned mice. The motor function of MPTP-lesioned mice was evaluated in the presence of ectopic miR-381 expression, and oxidative stress and dopaminergic neuron injury were also characterized. Restoration of miR-381 was demonstrated to diminish oxidative stress and damage in dopaminergic neurons, accompanied by enhanced motor functions. Mechanistically, the putative binding sites of miR-381 were retrieved through the starBase database, and the luciferase activity assay confirmed that it bound to EGR1 and repressed its expression, which then upregulated the expression of PTEN and p53. The neuroprotective effects of miR-381 on the motor function and dopaminergic neuronal damage were counteracted by ectopic EGR1 expression. Together, this study indicates that the binding of miR-381 to EGR1 upregulates PTEN/p53 to alleviate PD, which provides novel insights for a neuroprotective mechanism in PD.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce , MicroRNAs , Doença de Parkinson , Animais , Camundongos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , 1-Metil-4-fenilpiridínio/toxicidade , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Estresse Oxidativo , Doença de Parkinson/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo
12.
Bioengineered ; 13(3): 5737-5755, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35184652

RESUMO

Metastasis and chemoresistance are the leading causes of death in patients with hepatocellular carcinoma (HCC). microRNAs (miRNAs or miRs) may be useful as diagnostic, therapeutic and prognostic markers for HCC. In this study, we set out to investigate the possible role of miR-381 in HCC development and chemoresistance along with the related mechanism. Microarray-based gene expression profiling was carried out to analyze the expression of SET domain bifurcated 1 (SETDB1) and histone methyltransferase enhancer of zeste homolog 2 (EZH2) followed by validation in clinical HCC tissues and cells. The potential binding between miR-381 and SETDB1 was found and verified. Then, the role of SETDB1 in HCC in relation to miR-381 and protein kinase B (AKT) pathway was explored through gain- and loss-of-function approaches. After expression determination of EZH2, SETDB1, miR-381, and AKT pathway-related factors, their reactions were analyzed and their functional roles in HCC progression and chemoresistance were investigated in vitro and in vivo. SETDB1 was aberrantly upregulated in clinical HCC tissues and cells. This upregulation activated AKT pathway by promoting its tri-methylation on K64. SETDB1 promoted the proliferation, migration and chemoresistance through the AKT pathway in HCC cells. In a xenograft mouse model, SETDB1 promoted HCC cell tumorigenesis in vivo by activating the AKT pathway. Furthermore, EZH2 suppressed miR-381 by catalyzing the activity of H3K27me3 on its promoter region. In conclusion, EZH2 suppressed miR-381 expression by promoting H3K27me3 activity on its promoter region to facilitate SETDB1 expression, thereby activating the AKT pathway to promote hepatocarcinogenesis and chemoresistance.


Assuntos
Carcinoma Hepatocelular , Proteína Potenciadora do Homólogo 2 de Zeste , Histona-Lisina N-Metiltransferase , Neoplasias Hepáticas , MicroRNAs , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Nus , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
13.
Bioengineered ; 12(2): 12036-12048, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34753384

RESUMO

Our study aimed to investigate the clinical significance and biological functions of Spindlin1 (SPIN1) in colorectal cancer (CRC) tumorigenesis and progression, as well as the mechanism underlying its upregulation. The expression of SPIN1 was detected by immunohistochemistry and western blotting assays. Bioinformatics prediction and dual-luciferase reporter assays were used to determine whether microRNA-381 (miR-381) could target SPIN1. A series of cell functional experiments were performed to investigate whether the miR-381-mediated regulation of SPIN1 is involved in the progression and aggressiveness of CRC cells via the Wnt/ß-catenin pathway. Our results showed that SPIN1 is frequently overexpressed in CRC tissues and cell lines, and its upregulation is positively correlated with disease progression and lymph node metastasis. Moreover, SPIN1 depletion suppresses cell growth, migration, and invasion through inactivation of the Wnt/ß-catenin signaling pathway, which recapitulates the effects of miR-381 upregulation. Moreover, SPIN1 is a target gene of miR-381, and miR-381 is downregulated in CRC. Furthermore, the reintroduction of SPIN1 partially abolished the miR-381-mediated inhibitory effects in CRC cells. In summary, our data revealed that the miR-381/SPIN1 axis greatly contributes to CRC tumorigenesis by orchestrating the Wnt/ß-catenin pathway, thereby representing actionable therapeutic targets for colorectal cancer patients.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , MicroRNAs/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Via de Sinalização Wnt , Sequência de Bases , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Proteínas Associadas aos Microtúbulos/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Fosfoproteínas/genética , Regulação para Cima/genética , Via de Sinalização Wnt/genética
14.
Int Immunopharmacol ; 92: 107326, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33461162

RESUMO

OBJECTIVE: Over the years, roles of microRNAs (miRNAs) in development of human diseases has been broadly investigated, while the role of dexmedetomidine (DEX) regulating miR-381-5p in myocardial ischemia-reperfusion injury (MIRI) remains largely unknown. Thus, we aimed to identify the effect of DEX on MIRI via mediating miR-381-5p. METHODS: The MIRI mice models were established by the ligation of the left anterior descending (LAD) artery and treated with miR-381-5p agomir, silenced chitinase-3-like 1 protein (CHI3L1) and DEX. The cardiac function, serum inflammatory factors, pathological changes and cardiomyocyte apoptosis of the mice' myocardial tissues were measured. The targeting relationship between miR-381-5p and CHI3L1 was predicted. RESULTS: MiR-381-5p expression was decreased while CHI3L1 expression was increased in myocardial tissues of MIRI mice. DEX preconditioning could improve cardiac function and attenuate the pathological changes, cardiomyocyte apoptosis in myocardial tissues and inflammatory response in serum of MIRI mice. MiR-381-5p agomir improved the protective impact of DEX on myocardial injury in MIRI mice. Moreover, there existed a target relation between miR-381-5p and CHI3L1. CONCLUSION: Our study demonstrated that upregulated miR-381-5p strengthens the effect of DEX preconditioning to protect against MIRI in mouse models by inhibiting CHI3L1.


Assuntos
Proteína 1 Semelhante à Quitinase-3/metabolismo , Dexmedetomidina/farmacologia , MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Animais , Apoptose , Proteína 1 Semelhante à Quitinase-3/genética , Modelos Animais de Doenças , Hipnóticos e Sedativos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Transdução de Sinais , Regulação para Cima
15.
Life Sci ; 267: 118902, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33340525

RESUMO

OBJECTIVE: Based on the theory that long non-coding RNAs (lncRNAs) sponge microRNAs (miRNAs) to engage in cervical cancer development, this work was set out to investigate the possible role of lncRNA taurine upregulated gene 1 (TUG1) and miR-381-3p in the development of cervical cancer. METHODS: TUG1, miR-381-3p and murine double minute 2 (MDM2) expression were measured in cervical cancer tissues and cells. The nexus between TUG1 and clinicopathological features of cervical cancer was discussed. The biological functions of TUG1, miR-381-3p and MDM2 on cervical cancer cell process were interpreted via gain- and loss-of-function experiments. Also, tumor xenograft in nude mice was conducted in vivo. The interactions between TUG1, miR-381-3p and MDM2 were identified. RESULTS: TUG1 and MDM2 raised while miR-381-3p reduced in cervical cancer. TUG1 expression was related to tumor size, differentiation, international federation of gynecology and obstetrics stage and lymph node metastasis of cervical cancer. Restored miR-381-3p, depleted TUG1 or reduced MDM2 decreased viability, colony-forming, migration and invasion abilities, and facilitated apoptosis of cervical cancer cells. Xenografted tumors grew slowly upon injection with restored miR-381-3p and depleted TUG1. TUG1 bound to miR-381-3p and miR-381-3p targeted MDM2. CONCLUSION: On all accounts, this present study provides evidence that silencing TUG1 depressed cervical cancer cell progression through miR-381-3p/MDM2 axis, highlighting a theoretical basis for cervical cancer treatment.


Assuntos
MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/biossíntese , RNA Longo não Codificante/metabolismo , Neoplasias do Colo do Útero/metabolismo , Adulto , Idoso , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Sobrevivência Celular/genética , Feminino , Humanos , Camundongos , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Longo não Codificante/genética , Ativação Transcricional , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Life Sci ; 254: 117672, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32304760

RESUMO

AIM: MicroRNAs (miRNAs) function as oncogenes or tumor suppressors in an extensive variety of human cancers, including prostate cancer (PCa). Herein, the aim of this study is to investigate the effect of miR-381 on autophagy and apoptosis of PCa cells through the reelin (RELN)-mediated the PI3K/AKT/mTOR signaling pathway. METHODS: PCa-related differentially expressed genes and regulatory miRNA were retrieved according to microarray-based analysis and online mRNA-miRNA interaction analysis. The regulatory relationship between miR-381 and RELN was verified by bioinformatics prediction and dual-luciferase reporter gene assay. Gain- and loss-of-function experiments were applied to analyze the effects of miR-381 and RELN on the abilities of proliferation, apoptosis as well as autophagy of PCa cells. Furthermore, xenograft tumor was developed in nude mice to examine the tumorigenic ability of PCa. KEY FINDINGS: RELN, which was found to be up-regulated in PCa, was identified as a target of miR-381. Overexpressed miR-381 suppressed PCa cell proliferation while promoted autophagy and apoptosis of PCa cells. In addition, miR-381 inhibited the activation of the PI3K/AKT/mTOR signaling pathway by targeting RELN. In vivo experiments also confirmed that overexpression of miR-381 could inhibit the tumorigenic ability of PCa. CONCLUSION: Up-regulation of miR-381 inhibits the expression of RELN, which in turn inhibits the activation of the PI3K/AKT/mTOR signaling pathway, thereby inhibiting PCa cell proliferation, and promoting PCa cell apoptosis and autophagy.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , MicroRNAs/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Humanos , Masculino , Proteína Reelina
17.
Oncol Lett ; 19(3): 1899-1905, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32194685

RESUMO

The present study aimed to investigate the role of the long non-coding RNA EGFR-AS1 in bladder cancer (BC). In this study gene expression of both BC and non-tumor tissues from BC patients were measured by quantitative PCR. Cell transfections were performed to analyze gene interactions in HT-1197 cells. Transwell assays were performed to analyze cell invasion and migration of HT-1197 cells. It was revealed that epidermal growth factor receptor-antisense RNA 1 (EGFR-AS1) was upregulated in BC and positively associated with rho associated coiled-coil containing protein kinase 2 (ROCK2). Analysis of data collected in follow-ups indicated that EGFR-AS1 expression was significantly associated with poorer overall survival of patients with BC. Moreover, in bladder cancer cells, EGFR-AS1 overexpression mediated the upregulation of ROCK2, while microRNA (miR)-381 mediated the downregulation of ROCK2. However, EGFR-AS1 and ROCK2 failed to affect each other. Bioinformatics analysis indicated that miR-381 binds EGFR-AS1. In addition, EGFR-AS1 and ROCK2 overexpression resulted in the promotion of cell invasiveness and migration of HT-1197 BC cells. Conversely, miR-381 was revealed to partially reverse the effect of EGFR-AS1 overexpression. Therefore, EGFR-AS1 may sponge miR-381 to upregulate ROCK2 in BC, thereby promoting cell invasion and migration.

18.
Mol Med Rep ; 21(3): 1439-1448, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32016478

RESUMO

Coronary heart disease (CHD) is the leading cause of human morbidity and mortality worldwide. MicroRNA (miRNA) profiling is an innovative method of identifying biomarkers for many diseases and may be a powerful tool in the diagnosis and treatment of CHD. The present study aimed to analyze the effects of miRNA (miR)­381 on the inflammatory damage of endothelial cells during CHD. A total of 21 patients with CHD and 21 healthy control patients were enrolled in this study. Reverse transcription­quantitative PCR, western blotting and immunofluorescence assays were conducted to examine the expression levels of miR­381, C­X­C chemokine receptor type 4 (CXCR4), Bcl­2, Bax, Cleaved­Caspases­3 and ­9, p38, ERK1/2 and JNK. Cell Counting Kit­8, EdU and flow cytometry experiments were performed to evaluate cell proliferation and apoptosis. An ELISA was adopted to determine the expressions of inflammatory factors (interleukins­8, ­6 and ­1ß, and tumor necrosis factor­α). In addition, a dual­luciferase reporter assay was used to determine the relationship between miR­381 and CXCR4. Decreased miR­381 expression and increased CXCR4 expression in the plasma were observed in the CHD group compared with the normal group, which indicated a negative relationship between miR­381 and CXCR4. Overexpression of miR­381 significantly promoted the proliferation and inhibited the apoptosis of oxidized low­density lipoprotein (OX­LDL)­induced human umbilical vein endothelial cells (HUVECs) through mitogen­activated protein kinase pathway by targeting and inhibiting CXCR4. Furthermore, overexpression of miR­381 reduced the release of inflammatory factors in OX­LDL­induced HUVECs. By contrast, reduced expression of miR­381 exerted the opposite effects, which were subsequently reversed by silencing CXCR4 expression. Results from the present study indicated that miR­381 was a CHD­related factor that may serve as a potential molecular target for CHD treatment.


Assuntos
Doença das Coronárias/genética , MicroRNAs/genética , Receptores CXCR4/genética , Transdução de Sinais , Idoso , Apoptose , Proliferação de Células , Doença das Coronárias/fisiopatologia , Doença das Coronárias/terapia , Feminino , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Receptores CXCR4/sangue
19.
Regen Ther ; 14: 322-329, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32467829

RESUMO

INTRODUCTION: Ropivacaine has been regularly used because of its good anesthetic and analgesic effects, but it may exert neurotoxic effects on neurocyte. Dexmedetomidine has presented special advantages in the fields of neuroprotection, and it also could improve peripheral nerve block combining with ropivacaine. However, if dexmedetomidine could repair neurocyte injury induced by ropivacaine, and the specific mechanism remain unclear. METHODS: Western blotting and qRT-PCR were applied for measuring expression of protein and mRNA, respectively. Flow cytometry was used for assessing apoptosis. Cell proliferation was detected using Cell Counting Kit-8 (CCK-8) and colony formation assays. Transwell assay was applied to measure the migration and invasion of cells. Dual luciferase reporter assay was applied for confirming the binding site between microRNA-381 (miR-381) and Leucine-rich repeat C4 protein (LRRC4). RESULTS: The viability of PC12 cells increased with raising the concentration of dexmedetomidine (0 µM, 10 µM, 50 µM, 100 µM). Dexmedetomidine reversed role of ropivacaine (0 mM, 0.1 mM, 0.5 mM, 1 mM) by upragulating the expression of miR-381 and suppressing the expression of LRRC4 in PC12 cells. miR-381 can directly interact with target gene LRRC4 and negatively regulate its expression. Dexmedetomidine promoted the proliferation, migration, and invasion and inhibited apoptosis of PC12 cells by suppressing LRRC4 via up-regulating the expressions of miR-381 and further activated SDF-1/CXCR4 signaling pathway. CONCLUSIONS: Dexmedetomidine could protect PC12 cells from ropivacaine injury through miR-381/LRRC4/SDF-1/CXCR4 signaling pathway. This study may provide new therapeutic strategy targeting miR-381/LRRC4/SDF-1/CXCR4 signaling pathway about the prevention of ropivacaine induced neurocyte injury.

20.
Oncol Lett ; 18(2): 2066-2072, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31423279

RESUMO

Prostate cancer (PCa) is the most frequently diagnosed malignancy in men and its incidence has increased rapidly worldwide. Notably, the molecular mechanisms underlying prostate tumorigenesis have not been fully identified. The levels of microRNA (miR)-381 have been explored in numerous types of malignancy; however, the expression levels and biological function of miR-381 in PCa remain largely unknown. In the present study, reverse-transcription polymerase chain reaction was used to detect the expression levels of miR-381 in PCa cells and normal prostate epithelial cells. Subsequently, miR-381 antisense oligonucleotides and mimics were transfected into LNCaP PCa cells. Bioinformatics analysis was performed to identify the potential target genes of miR-381. Protein expression analysis, dual-luciferase reporter assay and a rescue assay were used to confirm the target of miR-381. The data suggested that the expression levels of miR-381 were significantly decreased in PCa cells compared with in normal prostatic epithelial cells. Furthermore, transfection of LNCaP cells with miR-381 mimics suppressed their proliferation, migration and invasion. In addition, bioinformatics analysis suggested that the androgen receptor (AR) was a target gene of miR-381. miR-381 suppressed the expression levels of AR by directly binding to its 3'-untranslated region. Furthermore, transfection with an AR plasmid partially attenuated miR-381-induced inhibition of cell proliferation, migration and invasion. The results of the present study suggested that miR-381 may act as a tumor suppressor in PCa by directly targeting the AR.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa