Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Front Biosci (Landmark Ed) ; 29(5): 189, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38812317

RESUMO

BACKGROUND: It has been demonstrated that exosomes derived from HPV-16 E7-over-expressiong non-small cell lung cancer (NSCLC) cells (E7 Exo) trigger increased levels of epidermal growth factor receptor (EGFR) and miR-381-3p. The purpose of this investigation was to examine the role of E7 Exo in NSCLC angiogenesis, and to analyze the contribution of exosomal EGFR and miR-381-3p to it. METHODS: The influence of E7 Exo on the proliferation and migration of human umbilical vein endothelial cells (HUVECs) was assessed using colony formation and transwell migration assays. Experiments on both cells and animal models were conducted to evaluate the angiogenic effect of E7 Exo treatment. The involvement of exosomal EGFR and miR-381-3p in NSCLC angiogenesis was further investigated through suppressing exosome release or EGFR activation, or by over-expressing miR-381-3p. RESULTS: Treatment with E7 Exo increased the proliferation, migration, and tube formation capacities of HUVECs, as well as angiogenesis in animal models. The suppression of exosome release or EGFR activation in NSCLC cells decreased the E7-induced enhancements in HUVEC migration and tube formation, and notably reduced vascular endothelial growth factor A (VEGFA) and Ang-1 levels. HUVECs that combined miR-381-3p mimic transfection and E7 Exo treatment exhibited a more significant tube-forming capacity than E7 Exo-treated HUVECs alone, but were reversed by the miR-381-3p inhibitor. CONCLUSION: The angiogenesis induced by HPV-16 E7 in NSCLC is mediated through exosomal EGFR and miR-381-3p.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Movimento Celular , Proliferação de Células , Receptores ErbB , Exossomos , Células Endoteliais da Veia Umbilical Humana , Neoplasias Pulmonares , MicroRNAs , Neovascularização Patológica , Proteínas E7 de Papillomavirus , MicroRNAs/genética , MicroRNAs/metabolismo , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Exossomos/metabolismo , Exossomos/genética , Receptores ErbB/metabolismo , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/irrigação sanguínea , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Animais , Linhagem Celular Tumoral , Camundongos , Camundongos Nus , Papillomavirus Humano 16/genética , Angiogênese
2.
Int Dent J ; 74(4): 823-835, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38685137

RESUMO

PURPOSE: To explore the mechanism underlying autophagy disruption in gingival epithelial cells (GECs) in diabetic individuals. METHODS AND MATERIALS: Bone marrow-derived macrophages (BMDMs) and GECs were extracted from C57/bl and db/db mice, the exosomes (Exo) were isolated from BMDMs. qRT‒PCR and Western blotting were performed to analyse gene expression. The AnimalTFDB database was used to identify relevant transcription factors, and miRNA sequencing was utilised to identify relevant miRNAs with the aid of the TargetScan/miRDB/miRWalk databases. A dual-luciferase assay was conducted to verify intermolecular targeting relationships. RESULTS: Similar to BMDMs, BMDM-derived Exos disrupted autophagy and exerted proinflammatory effects in GEC cocultures, and ATG7 may play a vital role. AnimalTFDB database analysis and dual-luciferase assays indicated that NR5A2 is the most relevant transcription factor that regulates Atg7 expression. SiRNA-NR5A2 transfection blocked autophagy in GECs and exacerbated inflammation, whereas NR5A2 upregulation restored ATG7 expression and ameliorated ExoDM-mediated inflammation. MiRNA sequencing, with TargetScan/miRDB/miRWalk analyses and dual-luciferase assays, confirmed that miR-381-3p is the most relevant miRNA that targets NR5A2. MiR-381-3p mimic transfection blocked autophagy in GECs and exacerbated inflammation, while miR-381-3p inhibitor transfection restored ATG7 expression and attenuated ExoDM-mediated inflammation. CONCLUSION: BMDM-derived Exos, which carry miR-381-3p, inhibit NR5A2 and disrupt autophagy in GECs, increasing periodontal inflammation in diabetes.


Assuntos
Autofagia , Células Epiteliais , Exossomos , Gengiva , Macrófagos , Camundongos Endogâmicos C57BL , MicroRNAs , MicroRNAs/genética , MicroRNAs/metabolismo , Autofagia/genética , Animais , Células Epiteliais/metabolismo , Exossomos/metabolismo , Macrófagos/metabolismo , Camundongos , Gengiva/citologia , Gengiva/metabolismo , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Masculino
3.
Funct Integr Genomics ; 23(1): 43, 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36658407

RESUMO

MicroRNA (miR)-381-3p is the newly discovered tumor-associated miRNA, which is frequently associated with diverse human malignancies; but, it is still unknown about its effect on acute myeloid leukemia (AML) in children. This work focused on exploring miR-381-3p's effect on childhood AML and identifying the possible mechanisms facilitating new treatment development. Using qRT-PCR analysis, miR-381-3p expression remarkably reduced in pediatric AML patients and AML cell lines (HL-60 and U937). Following transfection of miR-381-3p mimic or inhibitor into HL-60 and U937 cells, we conducted MTT assay to evaluate cell proliferation, flow cytometry (FCM) to measured cell apoptosis and cell cycle, whereas Transwell assays to detect cell invasion and migration. Our results demonstrated that miR-381-3p overexpression remarkably repressed cell growth, invasion and migration; additionally, miR-381-3p overexpression resulted in arrest of cell cycle and enhanced cell apoptosis. In contrast, miR-381-3p knockdown led to an opposite effect. Moreover, we predicted miR-381's target gene and validated it by luciferase reporter assay and TargetScan, separately. We identified miR-381-3p's binding site in ROCK1 3'-UTR. As revealed by Western-blot (WB) assay, miR-381-3p overexpression notably suppressed ROCK1 level. Moreover, restoring ROCK1 expression abolished miR-381-3p's inhibition on cell proliferation, invasion and migration. Data in this work indicated the role of miR-381-3p as the tumor suppressor within pediatric AML by targeting ROCK1. Therefore, miR-381-3p might serve as a potential therapeutic target for the treatment of pediatric AML.


Assuntos
Leucemia Mieloide Aguda , MicroRNAs , Humanos , Criança , Regulação para Baixo , MicroRNAs/genética , MicroRNAs/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Linhagem Celular , Proliferação de Células/genética , Linhagem Celular Tumoral , Apoptose/genética , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
4.
Cancer Biother Radiopharm ; 38(6): 396-404, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35029520

RESUMO

Objectives: MicroRNAs possess essential effects on gastric cancer (GC), whereas the underlying mechanisms have not been fully uncovered. The present work focused on investigating the role of miR-381-3p in GC cellular processes and the possible mechanisms. Materials and Methods: miR-381-3p levels within GC tissues and cells were measured through quantitative real-time polymerase chain reaction (qRT-PCR). This study measured cell proliferation, apoptosis, and metastasis through EdU, colony formation, flow cytometry, and Transwell assays separately. TargetScan was adopted to predict the miR-381-3p targets, whereas luciferase reporter assay was adopted for confirmation. Results: miR-381-3p levels were decreased, whereas fibroblast growth factor receptor-2 (FGFR2) expression was increased in GC. miR-381-3p upregulation inhibited proliferation, migration, and invasion and it promoted the apoptosis of GC cells. Further, FGFR2 overexpression partly reversed the miR-381-3p-mediated impacts on GC cellular processes. Conclusions: This study provides an experimental basis, suggesting the potential of using miR-381-3p as the novel marker for GC. Clinical Trial Registration number: 2020-05.


Assuntos
MicroRNAs , Neoplasias Gástricas , Humanos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , MicroRNAs/genética , MicroRNAs/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Neoplasias Gástricas/patologia
5.
Biology (Basel) ; 11(10)2022 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-36290402

RESUMO

Intramuscular fat (IMF) deposition is an important determinant of pork quality and a complex process facilitated by non-coding ceRNAs. In this study, 52 Berkshire × Anqing Sixwhite crossbred pigs were slaughtered to measure eight carcass and pork quality traits. Whole-transcriptome sequencing analysis was performed using longissimus dorsi samples of six low- and high-IMF samples; 34 ceRNA networks, based on 881, 394, 158 differentially expressed (DE) lncRNAs, miRNAs, and mRNAs, were constructed. Following weighted gene co-expression network analysis between the low and high IMF, only one ceRNA, lncRNA4789/miR-381-3p/FABP3, that showed similar DE trend in longissimus dorsi tissue was retained. Dual-luciferase reporter assays further indicated that FABP3 was a direct, functional target of miR-381-3p, where miR-381-3p overexpression inhibited the mRNA and protein expression of FABP3. In addition, overexpressed lncRNA4789 attenuated the effect of miR-381-3p on FABP3 by sponging miR-381-3p. Cell function verification experiment demonstrated that miR-381-3p suppressed IMF deposition by inhibiting preadipocyte cell differentiation and lipid droplet deposition via the suppression of FABP3 expression in the peroxisome proliferator-activated receptor signalling pathway, whereas lncRNA4789 rescued FABP3 expression by sponging miR-381-3p. Our study may aid in identifying novel molecular markers for its optimization in IMF which is of importance in breeding for improving pork quality.

6.
Arch Gynecol Obstet ; 306(6): 2093-2103, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35430713

RESUMO

PURPOSE: Cervical cancer (CC), as one of the most widespread gynecological malignancies in the world, severely threatens women health. Long non-coding RNA (lncRNA) could exert vital functions in assorted cancers, including CC. Although FLVCR heme transporter 1 antisense RNA 1 (FLVCR1-AS1) has been recognized as a critical effector in different cancers, its precise role and mechanisms have never been studied in CC. METHODS: RT-qPCR analysis was done for the measurement of the expression of FLVCR1-AS1, magnesium transporter 1 (MAGT1) and miR-381-3p in CC cells. Supported by western blot analysis, functional assays were done to evaluate the CC cell phenotype, while mechanism assays were done to explore the putative correlation among genes. RESULTS: In CC cells, FLVCR1-AS1 and MAGT1 were upregulated and miR-381-3p was downregulated. FLVCR1-AS1 or MAGT1 knockdown or miR-381-3p augment restrained CC cell proliferation, migration and invasion, but facilitated cell apoptosis. FLVCR1-AS1 sponged miR-381-3p, and MAGT1 was targeted by the FLVCR1-AS1/miR-381-3p axis. It was also revealed that the inhibitory influences of FLVCR1-AS1 silence on CC cell malignant behaviors were countervailed by MAGT1 overexpression. CONCLUSION: FLVCR1-AS1 exacerbated the malignant phenotype of CC cells via the miR-381-3p/MAGT1 axis.


Assuntos
Proteínas de Transporte de Cátions , MicroRNAs , RNA Longo não Codificante , Neoplasias do Colo do Útero , Humanos , Feminino , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Neoplasias do Colo do Útero/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética , Transformação Celular Neoplásica , Linhagem Celular Tumoral , Movimento Celular/genética , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo
7.
Autoimmunity ; 55(4): 233-242, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35481453

RESUMO

BACKGROUND: Circular RNAs (circRNAs) have been reported to be involved in the progression of infantile pneumonia. Here, we investigated the function of circTRHDE in lipopolysaccharide (LPS)-induced cell inflammatory injury to evaluate its role in infantile pneumonia progression. METHODS: The circTRHDE, microRNA (miR)-381-3p and TNF-receptor associated factor 3 (TRAF3) expression were detected by quantitative real-time PCR. LPS-induced WI-38 cells were used to construct an inflammatory injury model. Cell viability, inflammation and apoptosis were measured by cell counting kit assay, ELISA assay and flow cytometry. Caspase3 activity, MDA level and SOD activity were analysed to assess cell apoptosis and oxidative stress. Protein levels were determined using western blot analysis. The interaction between miR-381-3p and circTRHDE or TRAF3 was confirmed by dual-luciferase activity assay and RNA pull-down assay. RESULTS: CircTRHDE had increased expression in infantile pneumonia patients and LPS-induced WI-38 cells. LPS treatment inhibited WI-38 cell viability while promoting inflammation, apoptosis and oxidative stress. However, knockdown of circTRHDE remitted LPS-triggered WI-38 cell injury. CircTRHDE could sponge miR-381-3p to positively regulate TRAF3 expression. MiR-381-3p suppressed LPS-induced WI-38 cell inflammatory injury, and this effect was revoked by TRAF3 overexpression. Also, LPS-induced WI-38 cell inflammatory injury restrained by circTRHDE knockdown also were reversed by miR-318-3p inhibitor or TRAF3 overexpression. CONCLUSION: Our findings demonstrated that circTRHDE might be a target for infantile pneumonia treatment, which relieved LPS-induced cell inflammatory injury by the regulation of the miR-318-3p/TRAF3 axis.


Assuntos
MicroRNAs , Pneumonia , Apoptose/genética , Humanos , Inflamação/genética , Lipopolissacarídeos/efeitos adversos , MicroRNAs/genética , MicroRNAs/metabolismo , Pneumonia/induzido quimicamente , Fator 3 Associado a Receptor de TNF/genética , Fator 3 Associado a Receptor de TNF/metabolismo , Fator 3 Associado a Receptor de TNF/farmacologia
8.
Tissue Cell ; 76: 101791, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35427886

RESUMO

Although microRNAs (miRNAs) exert an important role in the osteogenesis of mesenchymal stem cells (MSCs), the effect of miR-381-3p on the osteogenic differentiation in MBD­MSCs is still unclear. The BMMSCs from patients with MBD (MBD­MSC) or normal participants (Normal­MSC) were isolated and induced to differentiation with dexamethasone. BMMSCs were transfected with miR-381-3p mimic, miR-381-3p inhibitor, and FGF7 siRNA to regulate the expression of miR-381-3p or FGF7. The direct binding between miR-381-3p and FGF7 was predicted and confirmed by bioinformatics prediction and luciferase reporter assay. The effect of miR-381-3p on the osteogenic differentiation of BMMSCs was assessed by RT­qPCR, alizarin Red S staining and western blot assays. Isolated BMMSCs showed the regular morphology, and were positive for CD44, CD90 and CD105 but negative for CD34 and CD45 markers. The calcium deposition and the relative mRNA expression levels of ALP, OC and OPN after induction were markedly enhanced. MiR-381-3p was upregulated in BMMSCs. Also, inhibition of miR-381-3p notably promoted osteogenic differentiation, vice versa. Besides, miR-381-3p could directly target FGF7 and negatively modulate the expression of FGF7. Moreover, inhibition of FGF7 attenuated the increase of the calcium deposition, and the relative mRNA expression of ALP, OC and OPN caused by the downregulation of miR-381-3p. In addition, the miR-381-3p inhibitor-induced the enhancement of the relative protein expressions of FGFR2, p-MEK and p-ERK1/2 were significantly reduced by the co-transfection of si-FGF7. Furthermore, the application of LY3214996, the inhibitor of ERK also verified these outcomes. MiR-381-3p directly targeting FGF7 modulated the osteogenic differentiation via MEK/ERK signaling pathway in BMMSCs.


Assuntos
Fator 7 de Crescimento de Fibroblastos , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais , MicroRNAs , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Cálcio/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Fator 7 de Crescimento de Fibroblastos/genética , Fator 7 de Crescimento de Fibroblastos/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Osteogênese/genética , Pirazóis , Pirróis , RNA Mensageiro/metabolismo
9.
Bioengineered ; 13(3): 6839-6855, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35246016

RESUMO

microRNAs, as small endogenous RNAs, influence umpteen sophisticated cellular biological functions regarding neurodegenerative and cerebrovascular diseases. Here, we interrogated miR-381-3p's influence on BV2 activation and neurotoxicity in ischemic and hypoxic environment. Oxygen-glucose deprivation (OGD) was adopted to induce microglial activation and HT-22 neuron damage. Quantitative polymerase chain reaction (qRT-PCR) was taken to check miR-381-3p expression in OGD-elicited BV2 cells and HT-22 neurons. It transpired that miR-381-3p expression was lowered in BV2 cells and HT-22 cells elicited by OGD. miR-381-3p up-regulation remarkably hampered inflammatory mediator expression in BV2 cells induced by OGD and weakened HT22 neuron apoptosis. In vivo, miR-381-3p expression was abated in HI rats' ischemic lesions, and miR-381-3p up-regulation could ameliorate inflammation and neuron apoptosis in their brain. C-C chemokine receptor type 2 (CCR2) was identified as the downstream target of miR-381-3p, and miR-381-3p suppressed the CCR2/NF-κB pathway to mitigate microglial activation and neurotoxicity. Therefore, we believed that miR-381-3p overexpression exerts anti-inflammation and anti-apoptosis in ischemic brain injury by targeting CCR2.


Assuntos
MicroRNAs , NF-kappa B , Animais , Apoptose/genética , Glucose/metabolismo , Glucose/toxicidade , Hipóxia/metabolismo , Hipóxia/patologia , Inflamação/metabolismo , Isquemia/metabolismo , Isquemia/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Microglia/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Neurônios/metabolismo , Oxigênio , Ratos , Receptores de Quimiocinas/metabolismo
10.
Front Cell Dev Biol ; 10: 853215, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35281083

RESUMO

Retinal progenitor cells (RPCs) transplantation has become a promising therapy for retinal degeneration, which is a major kind of ocular diseases causing blindness. Since RPCs have limited proliferation and differentiation abilities toward retinal neurons, it is urgent to resolve these problems. MicroRNAs have been reported to have vital effects on stem cell fate. In our study, the data showed that overexpression of miR-381-3p repressed Hes1 expression, which promoted RPCs differentiation, especially toward neuronal cells, and inhibited RPCs proliferation. Knockdown of endogenous miR-381-3p increased Hes1 expression to inhibit RPCs differentiation and promote proliferation. In addition, a luciferase assay demonstrated that miR-381-3p directly targeted the Hes1 3' untranslated region (UTR). Taken together, our study demonstrated that miR-381-3p regulated RPCs proliferation and differentiation by targeting Hes1, which provides an experimental basis of RPCs transplantation therapy for retinal degeneration.

11.
Future Oncol ; 18(7): 793-805, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34854318

RESUMO

Aim: This study aimed to identify specific and sensitive exosomal miRNAs in diagnosing patients with colorectal cancer (CRC). Methods: Serum exosomes were isolated from 175 CRC patients and 172 healthy donors by ultracentrifugation and identified by transmission electron microscopy, nanoparticle tracking analysis and western blotting. Exosomal miRNA expression was detected by quantitative PCR and the results analyzed by receiver operating characteristic analysis to illuminate the diagnostic accuracy. Results: Both exosomal miR-377-3p and miR-381-3p were downregulated in CRC patients as well as in early-stage patients compared with healthy donors; they could serve as circulating biomarkers of diagnosis, including early diagnosis, for CRC, possessing favorable diagnostic efficiency. Conclusion: Exosomal miR-377-3p and miR-381-3p levels were downregulated in CRC patients and may be useful as novel and specific biomarkers for the diagnosis of CRC, especially early-stage CRC.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Exossomos/metabolismo , MicroRNAs/metabolismo , Idoso , Biomarcadores Tumorais/sangue , Linhagem Celular Tumoral , Neoplasias Colorretais/sangue , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade
12.
Neuroimmunomodulation ; 29(3): 211-219, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34749366

RESUMO

INTRODUCTION: This study aimed to explore the diagnostic value and effect of miR-381-3p on Alzheimer's disease (AD). METHODS: RT-qPCR was used for the measurement of miR-381-3p levels. Pearson correlation coefficient was used for the correlation analysis. Receiver operating characteristic (ROC) curve was constructed to assess the distinct ability of miR-381-3p for AD. SH-SY5Y cells were treated with Aß25-35 to establish an AD cell model. The role of miR-381-3p on cell proliferation and apoptosis was detected. ELISA was applied to detect the protein levels of inflammatory cytokine expression. The target relationship of miR-381-3p with PTGS2 was verified by luciferase reporter gene assay. RESULTS: Low expression of miR-381-3p was detected in the serum of AD patients and cell models. There was a negative association of serum miR-381-3p with the serum inflammatory cytokines. The ROC curve demonstrated the distinct ability of serum miR-381-3p for AD, with the AUC value of 0.898, with a sensitivity of 87.5%, and a specificity of 77.7%. Overexpression of miR-381-3p reversed the influence of Aß25-35 on cell proliferation and apoptosis, but miR-381-3p downregulation exacerbated the influence. miR-381-3p overexpression inhibited the release of IL-6, IL-1ß, and TNF-α induced by Aß25-35 treatment, whereas miR-381-3p downregulation further promoted the release of inflammatory cytokines. PTGS2 was the target gene of miR-381-3p and was upregulated in AD cell models. CONCLUSION: miR-381-3p is less expressed in the serum of AD patients and has potential diagnostic values for AD. Overexpression of miR-381-3p may attenuate Aß25-35-induced neurotoxicity and inflammatory responses via targeting PTGS2 in SH-SY5Y cells.


Assuntos
Doença de Alzheimer , MicroRNAs , Neuroblastoma , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Ciclo-Oxigenase 2 , Humanos , Inflamação/metabolismo , MicroRNAs/genética
13.
Folia Neuropathol ; 60(4): 436-448, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36734386

RESUMO

INTRODUCTION: Long non-coding RNA (LncRNA) plays a critical role in cerebral ischemia-reperfusion (CI/R) injury. The purpose of the current research was to investigate the regulatory role of the LncRNA human leukocyte antigen complex group 11 (HCG11) in CI/R injury and explore its potential mechanism. MATERIAL AND METHODS: The rat middle cerebral artery occlusion (MCAO) model was established to simulate CI/R injury in vivo. mRNA levels of HCG11, microRNA (miR)-381-3p, tumour protein p53, and neuro-inflammatory factors were detected by quantitative reverse transcription PCR (RT-qPCR). Bederson score and Longa score were assessed for neurological deficits. Triphenyl tetrazolium chloride (TTC) staining was used to examine the cerebral infarct volume. What is more, oxidative stress was evaluated by the commercial kit. Finally, the relationship between HCG11, miR-381-3p, and p53 was verified by a dual-luciferase reporter assay. RESULTS: HCG11 was elevated in MCAO rats. And it competitively bound miR-381-3p and down-regulated the expression of p53. Inhibition of HCG11 inhibited cerebral infarct volume and neurological deficits in MCAO rats, and inhibited the secretion of neuro-inflammation and the over-activation of oxidative stress, exerting the protective effect of CI/R injury. However, inhibition of miR-381-3p in rats significantly weakened the protective effect of depression of HCG11 in MCAO rats, resulting in increased cerebral infarction volume and neurological deficits, elevated neuro-inflammatory secretion, and oxidative stress activation. CONCLUSIONS: The present research shows that LncRNA HCG11 silencing protects against cerebral ischemia/reperfusion injury through miR-381-3p to regulate p53.


Assuntos
MicroRNAs , Neoplasias , RNA Longo não Codificante , Traumatismo por Reperfusão , Animais , Ratos , Apoptose , Infarto da Artéria Cerebral Média/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Traumatismo por Reperfusão/metabolismo , RNA Longo não Codificante/genética , Proteína Supressora de Tumor p53/genética
14.
Pulm Pharmacol Ther ; 71: 102096, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34740750

RESUMO

The present study aimed to investigate the effects of PCI-34051-induced human bronchial epithelial cells (HBECs)-derived exosomes (PCI-Exo) on human bronchial smooth muscle cells (HBSMCs) and the key exosomal miRNAs involved in this process. Blank exosomes (Exo) and PCI-Exo were extracted from HBECs treated with PBS and PCI-34051, respectively. RNA-sequencing was performed to uncover the miRNA expression profile affected by PCI-Exo. The MTT, flow cytometry and TUNEL assays were performed to reveal the effect of PCI-34051 and PCI-Exo on the proliferation and apoptosis of HBSMCs. Western blotting and qRT-PCR were used for detecting protein and mRNA expression. A total of 25 exosomal miRNAs consisted of 17 down-regulated and eight up-regulated miRNAs were differentially expressed among PCI-Exo and Exo. Target genes of the exosomal miRNAs were mainly associated with signal transduction, cell adhesion, microRNAs in cancer, and ECM receptor interaction. miR-381-3p was identified as the most significant upregulated differential miRNA in PCI-Exo after qRT-PCR validation and could be transferred to HBSMCs by PCI-Exo. PCI-Exo treatment inhibited the proliferation but induced the apoptosis of HBSMCs. TGFß3 was identified as a target gene of miR-381-3p which could directly bind to the 3'UTR of TGFß3 mRNA. After transfecting the miR-381-3p mimic into HBSMCs, the proliferation inhibition and apoptosis rate of HBSMCs was significantly increased, and siTGFß3 transfection showed similar effects. Moreover, miR-381-3p overexpression could not only decrease the expression of α-SMA, FN1 and collagen I but also increase that of E-cadherin in HBSMCs. Our findings suggested that PCI-Exo could hinder the proliferation and obviously induce the apoptosis of HBSMCs, and its mechanisms might partly be attributable to the reduction of TGFß3 level by up-regulating exosomal miR-381-3p expression. These results may be vital for the treatment of lung related-diseases, especially asthma.


Assuntos
Exossomos , MicroRNAs , Intervenção Coronária Percutânea , Apoptose , Proliferação de Células , Histona Desacetilases , Humanos , Ácidos Hidroxâmicos , Indóis , MicroRNAs/genética , Miócitos de Músculo Liso , Proteínas Repressoras , Fator de Crescimento Transformador beta3
15.
Bioengineered ; 12(2): 11936-11946, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34784841

RESUMO

Immune response imbalance and cardiac dysfunction caused by sepsis are the main reasons for death in sepsis. This study aimed to confirm the expression and diagnostic possibility of microRNA-381-3p (miR-381-3p) and its mechanism in sepsis. Quantitative real-time PCR (qRT-PCR) and receiver operating characteristic (ROC) were used to reveal the levels and clinical significance of miR-381-3p. Pearson correlation was conducted to provide the correlations between miR-381-3p and several indexes of sepsis. The H9c2 cell models were constructed by lipopolysaccharide (LPS), and cecal ligation and puncture (CLP) was applied to establish the Sprague-Dawley (SD) rat models. Cell Counting Kit-8 (CCK-8) and flow cytometry were the methods to detect the cell viability and death rate of H9c2. Enzyme-linked immunosorbent assay (ELISA) was performed to evaluate the concentration of inflammatory cytokines. The target gene of miR-381-3p was validated via the luciferase report system. The low expression of miR-381-3p was found in the serum of patients with sepsis. The lessened miR-381-3p could be a marker in the discrimination of sepsis patients. Overexpression of miR-381-3p could repress the mRNA expression of HMGB1, inhibit the cell apoptosis and inflammatory response, and motivate the viability of sepsis cells. At the same time, enhanced miR-381-3p promoted the inhibition of inflammation and cardiac dysfunction in the rat model of sepsis. Collectively, reduced levels of serum miR-381-3p can be used as an index to detect sepsis patients. MiR-381-3p restored the inflammatory response and myocardial dysfunction caused by sepsis via HMGB1.


Assuntos
Proteína HMGB1/metabolismo , Inflamação/genética , MicroRNAs/metabolismo , Miocárdio/patologia , Sepse/diagnóstico , Sepse/genética , Animais , Sequência de Bases , Biomarcadores/metabolismo , Cardiotônicos/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Feminino , Regulação da Expressão Gênica , Humanos , Inflamação/complicações , Masculino , Camundongos , MicroRNAs/genética , Pessoa de Meia-Idade , Ligação Proteica , Ratos Sprague-Dawley , Sepse/complicações , Sepse/patologia
16.
Front Oncol ; 11: 727698, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34422671

RESUMO

BACKGROUND: Long non-coding RNAs (lncRNAs) display regulatory function flexibly in tumor onset and developments. Our study aimed to delve into the roles of lncRNA LINC01569 (LINC01569) in colorectal cancer (CRC) progression to study the potential mechanisms. METHODS: The genetic expression profiles of miR-381-3p and LINC01569 were measured by RT-PCR. The subcellular localization of LINC01569 in CRC cells was identified using subcellular fractionation location. Loss-of-function assays were performed to explore the potential effects of LINC01569 on CRC progression. Dual-luciferase reporter analysis was employed to verify the binding connections among LINC01569, miR-381-3p, and RAP2A. RESULTS: LINC01569 expression was distinctly increased in CRC. Curiously, if LINC01569 is removed, CRC cells will not migrate, proliferate, and invade remarkably. Molecular mechanism exploration uncovered that LINC01569 acted as a ceRNA competing with RAP2A to bind with miR-381-3p. Furthermore, rescue experiments corroborated the fact that miR-381-3p suppression reversed the inhibitory actions of LINC01569 knockdown on the expression of RAP2A and CRC progression. CONCLUSION: Overall, our findings indicate that LINC01569 plays a key role in CRC development by means of aiming at the miR-381-3p/RAP2A axis and can be equivalent to an underlying medicinal target to save CRC patients.

17.
World J Surg Oncol ; 19(1): 230, 2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-34362391

RESUMO

BACKGROUND: Accumulating evidence indicates that miRNAs are involved in multiple cellular functions and participate in various cancer development and progression, including breast cancer. METHODS: We aimed to investigate the role of miR-381-3p in breast cancer. The expression level of miR-381-3p and EMT transcription factors was examined by quantitative real-time PCR (qRT-PCR). The effects of miR-381-3p on breast cancer proliferation and invasion were determined by Cell Counting Kit-8 (CCK-8), colony formation, and transwell assays. The regulation of miR-381-3p on its targets was determined by dual-luciferase analysis, qRT-PCR, and western blot. RESULTS: We found that the expression of miR-381-3p was significantly decreased in breast cancer tissues and cell lines. Overexpression of miR-381-3p inhibited breast cancer proliferation and invasion, whereas knockdown of miR-381-3p promoted cell proliferation and invasion in MDA-MB-231 and SKBR3 cells. Mechanistically, overexpression of miR-381-3p inhibited breast cancer epithelial-mesenchymal transition (EMT). Both Sox4 and Twist1 were confirmed as targets of miR-381-3p. Moreover, transforming growth factor-ß (TGF-ß) could reverse the effects of miR-381-3p on breast cancer progression. CONCLUSIONS: Our observation suggests that miR-381-3p inhibits breast cancer progression and EMT by regulating the TGF-ß signaling via targeting Sox4 and Twist1.


Assuntos
Neoplasias da Mama , Transição Epitelial-Mesenquimal , MicroRNAs , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , Proteínas Nucleares , Prognóstico , Fatores de Transcrição SOXC , Proteína 1 Relacionada a Twist
18.
RNA Biol ; 18(sup1): 355-368, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34241580

RESUMO

Melanoma is considered as the most frequent primary malignancy occurring in skin. Accumulating studies have suggested that long non-coding RNAs (lncRNAs) play critical parts in multiple cancers. In this study, we explored the molecular mechanism of ZBED3 antisense RNA 1 (ZBED3-AS1) in melanoma. We observed that ZBED3-AS1 expression was remarkably up-regulated in melanoma tissues, and high ZBED3-AS1 level was linked to unsatisfactory survival of melanoma patients. Then, we discovered that ZBED3-AS1 was overexpressed in melanoma cells compared with human epidermal melanocytes. In addition, loss-of-function assays verified that ZBED3-AS1 knockdown restrained cell proliferation, migration, epithelial-mesenchymal transition (EMT), and stemness in melanoma. In addition, signal transducer and activator of transcription 3 (STAT3), which also showed tumour-facilitating functions in melanoma, was confirmed as a transcriptional activator of ZBED3-AS1. Moreover, ZBED3-AS1 enhanced the expression of AT-rich interaction domain 4B (ARID4B) through sequestering miR-381-3p. Importantly, we further confirmed that ZBED3-AS1 promoted the malignant progression of melanoma by regulating miR-381-3p/ARID4B axis to activate the phosphatidylinositol 3-kinase/AKT serine/threonine kinase (PI3K/AKT) signalling pathway. In a word, our research might provide a novel therapeutic target for melanoma.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Melanoma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Antissenso/genética , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição/genética , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Ciclo Celular , Movimento Celular , Proliferação de Células , Proteínas de Ligação a DNA/antagonistas & inibidores , Transição Epitelial-Mesenquimal , Humanos , Melanoma/genética , Melanoma/metabolismo , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fosfatidilinositol 3-Quinases/genética , Prognóstico , Proteínas Proto-Oncogênicas c-akt/genética , Fator de Transcrição STAT3/genética , Taxa de Sobrevida , Fatores de Transcrição/antagonistas & inibidores , Células Tumorais Cultivadas
19.
Bioengineered ; 12(1): 4452-4463, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34308746

RESUMO

Dendrobine has been reported to reduce blood lipid levels and apoptosis. The present study was designed to observe the effect of dendrobine in a model of ERS using vascular endothelial cells and to reveal the biological mechanisms and pathways responsible for the therapeutic effects of dendrobine on AS. Human umbilical vein endothelial cells (HUVECs) were pre-treated with various concentrations of dendrobine, followed by treatment with tunicamycin (TM) for the establishment of the cell models of ERS. The proliferation and apoptosis of HUVECs were detected by bromodeoxyuridine staining and flow cytometry, respectively. The target binding association was verified through dual luciferase reporter assay. It was found that TM treatment resulted in a low expression of miR-381-3p. Dendrobine treatment not only promoted the proliferation, but also inhibited the apoptosis of HUVECs induced by TM. The reduced expression of 78-kDa glucose-regulated protein, inositol-requiring enzyme 1, caspase-4, C/EBP homologous protein and caspase-3 was also observed following treatment with dendrobine. Dendrobine reduced the apoptosis of endothelial cells in the model of ERS by increasing miR-381-3p expression, and partially restored the cell proliferation level. This effect was significantly reduced after the expression of miR-381-3p was blocked. On the whole, the present study demonstrated that dendrobine upregulated miR-381-3p expression to inhibit apoptosis induced by ERS in HUVECs and this process was found to be mediated by caspase-4. The findings of the present study may provide new insight into the causes of endothelial cell apoptosis during AS and reveal the potent therapeutic effects of dendrobine in AS.


Assuntos
Alcaloides/farmacologia , Apoptose/efeitos dos fármacos , Caspases Iniciadoras/genética , Estresse do Retículo Endoplasmático/efeitos dos fármacos , MicroRNAs/genética , Caspases Iniciadoras/metabolismo , Linhagem Celular , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , MicroRNAs/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
Front Oncol ; 11: 657650, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33937069

RESUMO

BACKGROUND: We previously reported that the long non-coding RNA (lncRNA) CASC11 promotes colorectal cancer (CRC) progression as an oncogene by binding to HNRNPK. However, it remains unknown whether CASC11 can act as a competitive endogenous RNA (ceRNA) in CRC. In this study, we focused on the role of CASC11 as a ceRNA in CRC by regulating miR-646 and miR-381-3p targeting of RAB11FIP2. METHODS: We identified the target microRNAs (miRNAs) of CASC11 and the target genes of miR-646 and miR-381-3p using bioinformatic methods. A dual-luciferase reporter assay was performed to validate the target relationship. Quantitative real-time PCR (qRT-PCR), western blotting (WB), and immunohistochemistry (IHC) were used to measure the RNA and protein expression levels. Rescue experiments in vitro and in vivo were performed to investigate the influence of the CASC11/miR-646 and miR-381-3p/RAB11FIP2 axis on CRC progression. RESULTS: We found that CASC11 binds to miR-646 and miR-381-3p in the cytoplasm of CRC cells. Moreover, miR-646 and miR-381-3p inhibitors reversed the suppressive effect of CASC11 silencing on CRC growth and metastasis in vitro and in vivo. We further confirmed that RAB11FIP2 is a mutual target of miR-646 and miR-381-3p. The expression levels of CASC11 and RAB11FIP2 in CRC were positively correlated and reciprocally regulated. Further study showed that CASC11 played an important role in regulating PI3K/AKT pathway by miR-646 and miR-381-3p/RAB11FIP2 axis. CONCLUSION: Our study showed that CASC11 promotes the progression of CRC as a ceRNA by sponging miR-646 and miR-381-3p. Thus, CASC11 is a potential biomarker and a therapeutic target of CRC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...