Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Noncoding RNA Res ; 9(2): 547-559, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38515792

RESUMO

A non-negligible part of our DNA has been proven to be transcribed into non-protein coding RNA and its intricate involvement in several physiological processes has been highly evidenced. The significant biological role of non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) has been variously reported. In the current review, the authors highlight the multifaceted role of myocardial infarction-associated transcript (MIAT), a well-known lncRNA, in hepatocellular carcinoma (HCC). Since its discovery, MIAT has been described as a regulator of carcinogenesis in several malignant tumors and its overexpression predicts poor prognosis in most of them. At the molecular level, MIAT is closely linked to the initiation of metastasis, invasion, cellular migration, and proliferation, as evidenced by several in-vitro and in-vivo models. Thus, MIAT is considered a possible theranostic agent and therapeutic target in several malignancies. In this review, the authors provide a comprehensive overview of the underlying molecular mechanisms of MIAT in terms of its downstream target genes, interaction with other classes of ncRNAs, and potential clinical implications as a diagnostic and/or prognostic biomarker in HCC.

2.
Prev Nutr Food Sci ; 28(3): 224-234, 2023 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-37842247

RESUMO

Nanocurcumin (NaC) and high-intensity interval training (HIIT) play crucial role in weight and inflammation control. The purpose of the current study was to evaluate the separate and combined effects of 8 weeks of Tabata-HIIT and NaC supplementation on the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, long non-coding RNA myocardial infarction associated transcript (lncRNA MIAT) expression, body composition, and cardiorespiratory health in elderly overweight women. A total of 48 healthy overweight elderly women were randomly divided into four groups: NaC, Tabata-HIIT+Pla, Tabata-HIIT+NaC, and placebo. Participants underwent a Tabata HIIT program (2 days per week, at 80∼0% of maximal HR) and NaC supplementation (daily 80 mg in two 40 mg capsules) for 8 weeks. Blood sampling, cardiorespiratory hemodynamic responses, and body composition evaluations were obtained before and after treadmill stress testing at the baseline timepoint and following 8 weeks of intervention. The mRNA of lncRNA-MIAT and NLRP3 were measured by real-time polymerase chain reaction. After 8 weeks, a significant improvement was observed in body composition and cardiorespiratory hemodynamics in the Tabata-HIIT groups compared to the NaC alone and placebo groups (P<0.05). Tabata training, both with and without the addition of nano curcumin supplementation, did not result significant effect on the resting levels of lncRNA-MIAT expression (P>0.05). Nevertheless, NaC supplementation along with Tabata training led to a significant reduction in NLRP3 inflammasome. In addition, NaC supplementation in overweight/preobese women improved systemic inflammation during treadmill stress testing. These findings indicating the suppressive effects of non-pharmacologic interventions on the sympathetic system and downregulation of the inflammasome.

3.
Noncoding RNA ; 9(4)2023 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-37624039

RESUMO

Myocardial Infarction Associated Transcript (MIAT) is a nuclear long non-coding RNA (LncRNA) with four different splicing variants. MIAT dysregulation is associated with carcinogenesis, mainly acting as an oncogene regulating cellular growth, invasion, and metastasis. The aim of the current study is to investigate the role of MIAT in the regulation of T and chronic myeloid leukemic cell survival. To this end, MIAT was silenced using MIAT-specific siRNAs in leukemic cell lines, and functional assays were performed thereafter. This investigation also aims to investigate the effects of MIAT silencing on the expression of core genes involved in cancer. Functional studies and gene expression determination confirm that MIAT knockdown not only affects short- and long-term survival and the apoptosis of leukemic cells but also plays a pivotal role in the alteration of key genes involved in cancer, including c-MYC and HIF-1A. Our observations suggest that MIAT could act as an oncogene and it has the potential to be used not only as a reliable biomarker for leukemia, but also be employed for prognostic and therapeutic purposes.

4.
Exp Neurol ; 357: 114159, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35779615

RESUMO

Activation of an innate immune response serves as a key, contributing factor in perinatal brain injury. The current study sought to evaluate the clinical significance of innate defense regulatory peptide 1018 (IDR-1018)-derived peptide mediating ceRNA regulation network as a biomarker in neonatal mice with hypoxic-ischemic brain damage (HIBD). Firstly, bioinformatics analyses were performed to screen the HIBD-related candidate genes, miRNAs, and lncRNAs. The StarBase, miRDB, and LncBase databases were retrieved to obtain the lncRNA-miRNA-mRNA network, which revealed the ceRNA regulatory network mediated by IDR-1018. Subsequently, RT-qPCR was adopted to determine the expression patterns of MIAT, miR-7a-5p, and Plp2 in neonatal mice with HIBD after treatment with IDR-1018. Moreover, the relationship among mRNA, miRNA, and lncRNA in primary hippocampal neurons was verified by means of dual-luciferase reporter assay and RIP assay. Initial findings demonstrated that Plp2, mmu-miR-7a-5p, and three lncRNAs (MIAT, XIST, and 1700020I14RIK) were related to HIBD. Moreover, IDR-1018 could relieve HIBD in neonatal mice. Plp2 and MIAT were down-regulated, while mmu-miR-7a-5p was up-regulated in the striatum, hippocampus, and cortical tissues of the neonatal mice with HIBD, whereas treatment with the IDR-1018 could revere these trends. Additionally, MIAT acted as a ceRNA of miR-7a-5p to elevate Plp2 expression. In conclusion, our findings highlighted that IDR-1018 relieved HIBD in neonatal mice via the MIAT/miR-7a-5p/Plp2 axis.


Assuntos
Lesões Encefálicas , Hipóxia-Isquemia Encefálica , MicroRNAs , RNA Longo não Codificante , Animais , Animais Recém-Nascidos , Peptídeos Catiônicos Antimicrobianos , Apoptose/genética , Biologia Computacional , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Peptídeos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro
5.
J Inflamm Res ; 15: 3269-3283, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35676970

RESUMO

Purpose: The biological role and mechanism of long noncoding RNA (lncRNA) myocardial infarction-associated transcript (MIAT) in dry eye remain to be illustrated. Pyroptosis is a noticeable form of inflammatory activation, which is characteristic of gasdermin D (GSDMD)-driven cell death. The present study was designed to explore the role of MIAT in pyroptosis and apoptosis induced by hyperosmolarity stress (HS) in human corneal epithelial cells (HCECs). Methods: HCECs were cultured in 70-120 mM hyperosmotic medium for 24 h to create a dry eye model in vitro. The level of the pyroptosis marker GSDMD was measured, and the cell inflammatory response was evaluated by detecting IL-1ß and IL-18 levels. Exogenous caspase-1 inhibitor Ac-YVAD-CHO was used. The pyroptosis in HCECs was examined by caspase-1 activity, immunofluorescent staining, and Western blotting. Flow cytometry was performed to test the apoptosis rate of HCECs. Cell migration and proliferation were detected. The expression of the lncRNA MIAT in HCECs was detected by quantitative real-time PCR. MIAT was knocked down by small interfering RNA (siRNA) transfection. The effects of caspase-1 inhibition on pyroptosis, apoptosis, migration, and proliferation were observed. Results: HS promoted pyroptosis in HCECs by elevating caspase-1, GSDMD, and the active cleavage of GSDMD (N-terminal domain, N-GSDMD), and increased the release of IL-1ß, IL-18, LDH and the rate of apoptosis, with reduced cell migration. These changes were prevented by the inhibition of caspase-1. The expression of MIAT was significantly increased in HCECs exposed to a hyperosmotic medium. Silencing MIAT increased the expression of GSDMD, caspase-1, and inflammatory chemokines IL-1ß and IL-18, and promoted apoptosis while inhibiting migration and proliferation in HCECs. Conclusion: The lncRNA MIAT is involved in HS-induced pyroptosis and apoptosis and the inflammatory response of HCECs and provides a new understanding of the pathogenesis of dry eye.

6.
Genes (Basel) ; 13(1)2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-35052495

RESUMO

Angiogenesis is one of the hallmarks of cancer, and the establishment of new blood vessels is vital to allow for a tumour to grow beyond 1-2 mm in size. The angiogenic switch is the term given to the point where the number or activity of the pro-angiogenic factors exceeds that of the anti-angiogenic factors, resulting in the angiogenic process proceeding, giving rise to new blood vessels accompanied by increased tumour growth, metastasis, and potential drug resistance. Long noncoding ribonucleic acids (lncRNAs) have been found to play a role in the angiogenic switch by regulating gene expression, transcription, translation, and post translation modification. In this regard they play both anti-angiogenic and pro-angiogenic roles. The expression levels of the pro-angiogenic lncRNAs have been found to correlate with patient survival. These lncRNAs are also potential drug targets for the development of therapies that will inhibit or modify tumour angiogenesis. Here we review the roles of lncRNAs in regulating the angiogenic switch. We cover specific examples of both pro and anti-angiogenic lncRNAs and discuss their potential use as both prognostic biomarkers and targets for the development of future therapies.


Assuntos
Neoplasias/patologia , Neoplasias/terapia , Neovascularização Patológica/patologia , RNA Longo não Codificante/genética , Animais , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/genética
7.
Bioengineered ; 13(1): 407-417, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34783297

RESUMO

Dysregulated lipid metabolism of macrophages contributes to thrombosis and antiphospholipid syndrome (APS). The long non-coding RNAs (lncRNA) myocardial infarction-associated transcript 2 (Mirt2) has been reported to inhibit inflammation and lipid accumulation; therefore, this study intended to clarify whether Mirt2 served a role in lipid metabolism. THP-1-derived macrophages with or without Mirt2-knockdown or overexpression, were exposed to oxidized low-density lipoprotein (ox-LDL), then cell migration, lipid accumulation, cholesterol efflux and inflammation were assessed using wound healing, oil red staining, commercial kits and western blot assays. Besides, ML385 was used to treat THP-1-derived macrophages to inhibit nuclear factor erythroid-related factor 2 (NRF2) expression. The expression of proteins involved in the above processes were measured by western blot. Results demonstrated that phorbol 12-myristate 13-acetate (PMA) significantly increased Mirt2 expression in THP-1 cells. Mirt2-knockdown enhanced ox-LDL-induced macrophage migration, lipid accumulation, inflammation, and inhibited cholesterol efflux. By contrast, Mirt2 overexpression displayed the opposite effects. Furthermore, Mirt2-knockdown inhibited NRF2 signaling and enhanced mitogen-activated protein kinase (MAPK) signaling, while Mirt2 overexpression displayed the opposite effects. Finally, the NRF2 inhibitor ML385 significantly reversed the above effects of Mirt2. In summary, Mirt2 served an important role in regulating lipid metabolism in macrophages via inhibiting MAPK signaling and activating the NRF2 signaling pathway.


Assuntos
Colesterol/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/citologia , Fator 2 Relacionado a NF-E2/metabolismo , RNA Longo não Codificante/genética , Acetato de Tetradecanoilforbol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células THP-1 , Regulação para Cima
8.
Bioengineered ; 13(1): 930-940, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34967706

RESUMO

This study attempted to evaluate the role of long non-coding RNA myocardial infarction-associated transcript (LncRNA MIAT) in Parkinson's disease (PD). The mouse model was established through intraperitoneal injection with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and in vitro model was induced by administrating cell with 1-Methyl-4-phenylpyridinium ion (MPP+). Rotarod test was conducted to evaluate the motor coordination of PD mice. In order to investigate the roles of LncRNA MIAT in neuronal inflammation and oxidative stress, MIAT shRNA (shMIAT) was transfected into MPP+-treated cells, and cell viability, cell apoptosis and oxidative stress response were evaluated. To evaluate the interactions between LncRNA MIAT and microRNA-221-3p (miR-221-3p)/TGF-ß1/Nrf2, miR-221-3p mimic, miR-221-3p inhibitor, NC-inhibitor and transforming growth factor-ß1 shRNA (shTGF-ß1) were subsequently transfected into MPP+-treated cells. Dual-luciferase reporter gene assays were performed to determine the interaction of miR-221-3p with MIAT or TGFB receptor 1 (TGFBR1). The expressions of LncRNA MIAT, miR-221-3p, TGFBR1, transforming growth factor (TGF-ß1) and nuclear factor E2-related factor 2 (Nrf2) were measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR) and immunoblotting. As a result, LncRNA MIAT was abundantly expressed in PD mice and cells, while downregulation of LncRNA MIAT promoted the survival of neurons, inhibited apoptosis and oxidative stress in neurons. LncRNA MIAT bound to miR-221-3p, and there was a negative correlation between miR-221-3p and LncRNA MIAT expression. In addition, miR-221-3p targeted TGFBR1 and suppressed TGF-ß1 expression but increased Nrf2 expression. LncRNA MIAT promoted MPP+-induced neuronal injury in PD via regulating TGF-ß1/Nrf2 axis through binding with miR-221-3p.


Assuntos
1-Metil-4-fenilpiridínio/efeitos adversos , MicroRNAs/genética , Fator 2 Relacionado a NF-E2/genética , Doença de Parkinson/fisiopatologia , RNA Longo não Codificante/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Fator de Crescimento Transformador beta1/genética , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , RNA Nuclear Heterogêneo/administração & dosagem , RNA Nuclear Heterogêneo/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Teste de Desempenho do Rota-Rod , Fator de Crescimento Transformador beta1/metabolismo
9.
Exp Ther Med ; 22(5): 1342, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34630696

RESUMO

Long non-coding RNAs (lncRNAs) can function as onco-lncRNAs in several types of human cancer, including retinoblastoma (Rb). The present study investigated the potential role and regulatory mechanism of the lncRNA myocardial infarction-associated transcript (MIAT) in Rb. To do so, the expression levels of MIAT, microRNA (miR)-665, and LIM and SH3 protein 1 (LASP1) in Rb tissues from patients or Rb cells were analysed using reverse transcription quantitative PCR. The interactions between miR-665 and MIAT/LASP1 were confirmed by the dual-luciferase reporter assay. MTT, Transwell (to assess migration and invasion) and western blotting assays were used to explore the functions of the MIAT/miR-665/LASP1 axis on Rb progression in vitro. The results of the present study indicated that MIAT targeted miR-665. In Rb tissues and cell lines, high expression of MIAT was observed, whereas miR-665 was downregulated in Rb tissues. Furthermore, the proliferation and migratory and invasive abilities of Rb Y79 and HXO-RB44 cells were decreased following MIAT downregulation or miR-665 overexpression. In addition, LASP1 was identified as a target gene of miR-665. Both the decreased expression of miR-665 and the elevated expression of LASP1 reversed the suppressive effects of MIAT knockdown on the proliferation and migratory and invasive abilities of Y79 cells. Furthermore, MIAT silencing attenuated the development of Rb by regulating the miR-665/LASP1 axis. Taken together, these findings suggested that MIAT may be considered as a possible therapeutic target for Rb.

10.
Am J Transl Res ; 13(9): 9993-10013, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34650678

RESUMO

To examine the neuroprotective roles of lncRNA-MIAT in Parkinson's disease (PD). RNA sequencing expression profiles were utilized to screen the dysregulated lncRNAs in patients with PD and to explore the underlying molecular mechanisms by which the lncRNAs regulate the pathogenesis of PD. 6-hydroxydopamine-induced SH-SY5Y cell lines and a PD mouse model were used to prove how the overexpressing or knocking-down of MIAT produce a marked effect in both in vitro and in vivo experiments. Subsequently, the subcellular localization of MIAT was detected via RNA fluorescence in situ hybridization (FISH) assays. Quantitative PCR, as well as western blotting, were used to determine the expression levels of the associated genes and proteins. We utilized Cell Counting Kit-8 (CCK8) assays to measure the viability of the cells, and the apoptotic rate was determined using Annexin V-FITC/PI double staining. The expressions of tyrosine hydroxylase (TH) and Parkin were quantified in the substantia nigra using immunohistochemical staining. Also, TUNEL staining was performed to visualize the apoptotic cells in the substantia nigra. Compared with the normal rats, the downregulation of MIAT was observed in the cortex, hippocampus, substantia nigra, and striatum of the PD rats. Overexpression of MIAT exhibited a neuroprotective effect on the SH-SY5Y cells. Through RNA-sequencing of the PD mice treated with an overexpression of MIAT and through a differentially expressed genes analysis, it was hypothesized that MIAT could upregulate the expression of synaptotagmin-1 (SYT1) through sponging of miR-34-5p. Interactions between MIAT, miR-34-5p, and SYT1 were confirmed using RIP and dual-luciferase reporter assays. At the same time, the MIAT overexpression group exhibited elevated Parkin and TH protein levels, increased cell viability but a decreased apoptosis rate of the SH-SY5Y cells in contrast with the negative control (NC) group. In vivo, compared with the NC group, the overexpression of MIAT resulted in an increase in the positive rates of Parkin and TH, and the apoptosis was decreased in the PD mice. The behavioral test results showed that the motor coordination and autonomous activity of the mice were enhanced in the MIAT overexpression group compared with the NC group. LncRNA-MIAT regulates the growth of SHSY5Y cells by sponging miR-34-5p which targets SYT1 and exerts a neuroprotective effect in a mouse model of PD.

11.
J Cardiol ; 78(6): 586-597, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34489160

RESUMO

BACKGROUND: Coronary artery disease (CAD) is a common heart disease with high incidence and mortality. Myocardial ischemia is the main type of CAD, which negatively affects health worldwide. The aim of the present study was to investigate the function and mechanism of myocardial infarction-associated transcript (MIAT) in myocardial ischemia. METHODS: Human cardiomyocytes (HCM) were treated with oxygen-glucose deprivation (OGD) to set the in vitro model and mouse myocardial ischemia/reperfusion (I/R) was set for in vivo model. Cell viability and apoptosis were detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, flow cytometry, and immunofluorescence analysis. Inflammatory cytokines levels were detected by enzyme-linked immunosorbent assay. Gene and protein expressions were identified by quantitative real time-polymerase chain reaction or Western blotting. The interaction of MIAT, miR-181a-5p, and janus kinase 2 (JAK2) was identified by dual-luciferase report assay. Mouse heart tissues histopathological condition were observed by hematoxylin and eosin assays. RESULTS: Expression of MIAT and JAK2 were increased in OGD-treated HCM and mice of I/R model group, and miR-181a-5p was decreased. MIAT silencing could reverse the OGD treatment induced cell proliferation inhibition, cleaved caspase-3 and Bcl2-associated X (Bax) levels increased, while those of B-cell lymphoma-2 (Bcl-2) and mitochondria's cyt-C decreased. Besides, MIAT knockdown attenuated the OGD-induced increase of tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6 levels. Moreover, MIAT targeted miR-181a-5p to enhance the expression of JAK2 and signal Transducer and Activator of Transcription 3 (STAT3), and miR-181a-5p overexpression promoted proliferation, whereas it inhibited apoptosis in OGD-induced cardiomyocytes. Furthermore, the regulatory effects of MIAT knockdown in cell proliferation, apoptosis, and inflammatory injury was reversed by inhibition of miR-181a-5p or overexpression of JAK2 in OGD-treated HCM. Knockdown of MIAT reduced myocardial injury caused by I/R treatment in vivo. CONCLUSION: MIAT knockdown inhibited apoptosis and inflammation by regulating JAK2/STAT3 signaling pathway via targeting miR-181a-5p in myocardial ischemia model. MIAT can be a possible therapeutic target for controlling the progression of myocardial ischemia.


Assuntos
MicroRNAs , Infarto do Miocárdio , RNA Longo não Codificante , Animais , Apoptose , Glucose , Humanos , Janus Quinase 2/metabolismo , Camundongos , MicroRNAs/genética , Miócitos Cardíacos/metabolismo , Oxigênio , RNA Longo não Codificante/genética , Fator de Transcrição STAT3/metabolismo
12.
Brain Res Bull ; 175: 81-89, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34265390

RESUMO

OBJECTIVE: Long non-coding RNAs (lncRNAs) have diagnostic and therapeutic values in the setting of ischemic stroke (IS). Here, we evaluated the value of myocardial infarction-associated transcript (MIAT) in IS with the involvement of microRNA (miR)-874-3p/interleukin (IL) 1B. METHODS: MIAT, miR-874-3p and IL1B levels in serum of patients with IS were measured. A middle cerebral artery occlusion (MCAO) model was established in mice. MCAO mice were injected with Agomir of miR-874-3p, shRNA or overexpression vector of MIAT or siRNA of IL1B. Subsequently, behavioral activities and neurological function of mice were assessed. The number of Nissl bodies, brain damage, neuronal apoptosis and inflammatory factors in brain tissues of mice were measured. The targeting relationship between MIAT and miR-874-3p, as well as that between miR-874-3p and IL1B was explored. RESULTS: In patients with IS, MIAT and IL1B were up-regulated and miR-874-3p was down-regulated. MIAT absorbed miR-874-3p while miR-874-3p targeted IL1B. Silencing of MIAT or IL1B, or promotion of miR-874-3p improved behavioral activities and neurological function of mice, reduced the number of Nissl bodies, as well as improved brain damage, neuronal apoptosis and inflammation. Overexpression of miR-874-3p abrogated up-regulated MIAT-mediated influence on MCAO mice. CONCLUSION: Shortly, this study figures out that MIAT impairs neurological function in IS via up-regulating miR-874-3p-targeted IL1B.


Assuntos
Interleucina-1beta/genética , AVC Isquêmico/genética , AVC Isquêmico/fisiopatologia , MicroRNAs/genética , RNA Longo não Codificante/genética , Animais , Comportamento Animal , Encéfalo/patologia , Feminino , Inativação Gênica , Marcação de Genes , Humanos , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Interleucina-1beta/sangue , AVC Isquêmico/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Regulação para Cima
13.
J Int Med Res ; 49(7): 3000605211031433, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34275376

RESUMO

OBJECTIVE: This study aimed to investigate the role of long noncoding RNA (LncRNA) myocardial infarction-associated transcript (MIAT) in a heart failure (HF) model in vivo and in vitro by regulating the PI3K/Akt signaling pathway. METHODS: We established HF models in vivo and in vitro and evaluated the collagen content of these models and other factors. RESULTS: We found that when LncRNA MIAT was silenced, vascular endothelial growth factor, phosphorylated protein kinase B (Akt), and phosphorylated phosphoinositide 3-kinase (PI3K) mRNA and protein levels were significantly downregulated, which suggested that MIAT activated the PI3K/Akt signaling pathway. Akt and PI3K expression was not significantly changed. We also found that when LncRNA MIAT was silenced, collagen expression was significantly downregulated. This finding suggested that MIAT promoted myocardial fibrosis during the development of HF. The levels of inflammatory factors were also significantly reduced with silencing of LncRNA MIAT. This finding suggested that MIAT promoted the expression of inflammatory factors in myocardial fibrosis by activating the PI3K/Akt signaling pathway. CONCLUSION: This study indicates that silencing LncRNA MIAT may improve myocardial fibrosis and alleviate HF through the PI3K/Akt signaling pathway, which may be helpful for patients with HF to obtain a better therapeutic effect.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Fibrose , Insuficiência Cardíaca/genética , Humanos , Infarto do Miocárdio/genética , Fosfatidilinositol 3-Quinase , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular
14.
Aging (Albany NY) ; 13(8): 11188-11206, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33819189

RESUMO

Long non-coding RNA (lncRNA) myocardial infarction-associated transcript (MIAT) has been widely-demonstrated to function as diagnostic markers for acute myocardial infarction (MI). This study was designed to explore the modulatory role of MIAT and its underlying molecular mechanism in MI. Firstly, MI mouse model was developed via ligation of the descending branch of the left coronary artery, and cell model was established through exposure to hypoxic conditions. Online prediction indicated that MIAT could bind to microRNA-10a-5p (miR-10a-5p), while miR-10a-5p was highlighted to bind to early growth response gene-2 (EGR2). MIAT and EGR2 were subsequently determined to be highly-expressed, whereas miR-10a-5p was found to be poorly-expressed in cardiomyocytes exposed to hypoxia as well as in MI mice using RT-qPCR and Western blot assay. The binding relationships between MIAT and miR-10a-5p, and between miR-10a-5p and EGR2 were further confirmed by dual-luciferase reporter and RNA immunoprecipitation assays. The results of in vitro and in vivo experimentation also suggested that overexpression of miR-10a-5p or silencing of MIAT and EGR2 reduced cardiomyocyte apoptosis and increased ATP content, thus alleviating the impairment of cardiac function following MI. In a word, inhibition of MIAT protects against cardiac dysfunction induced by MI through the crosstalk with miR-10a-5p/EGR2.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/genética , MicroRNAs/metabolismo , Infarto do Miocárdio/genética , Miocárdio/patologia , RNA Longo não Codificante/metabolismo , Animais , Hipóxia Celular/genética , Linhagem Celular , Biologia Computacional , Vasos Coronários/patologia , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , RNA Longo não Codificante/genética , Organismos Livres de Patógenos Específicos , Regulação para Cima
15.
Mol Med Rep ; 23(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33760182

RESUMO

The development of novel treatments for lymphedema is hindered by the poorly understood pathophysiology of the disease. To improve the therapeutic success of treating the disease, the present study aimed to investigate the effects and mechanism of long non­coding RNA myocardial infarction­associated transcript (MIAT) in terms of the differentiation of adipose­derived mesenchymal stem cells (ADMSCs) into lymphatic endothelial cells (LECs). The expression levels of (MIAT), microRNA (miR)­495 and Prospero­related homeobox 1 (Prox1) were measured by reverse transcription­quantitative PCR. The protein expression levels of Prox1, lymphatic vessel endothelial hyaluronan receptor 1 (LYVE­1), vascular endothelial growth factor receptor­3 (VEGFR­3) and podoplanin (PDPL) were detected by western blotting and immunofluorescence. A dual­luciferase reporter assay was also used to detect the interaction between MIAT, miR­495 and Prox1. In addition, migration and tube­formation capabilities were measured by Transwell assay and tube­formation assay, respectively. The results obtained demonstrated that VEGF­C156S (recombinant VEGF­C in which Cys156 was replaced by Ser residue) treatment could efficiently induce the differentiation of ADMSCs into LECs. MIAT expression was upregulated and miR­495 was downregulated during differentiation. Mechanistically, MIAT upregulated Prox1 expression possibly by acting as a molecular sponge for miR­495. Functional analyses indicated that the expression levels of Prox1, LYVE­1, VEGFR­3 and PDPL, and the migration and tube­formation capabilities of ADMSCs induced by VEGF­C156S, were significantly inhibited by silencing MIAT and overexpressing miR­495. Moreover, miR­495 inhibition and Prox1 overexpression reversed the effects of MIAT downregulation and miR­495 upregulation, respectively, on the differentiation of ADMSCs into LECs. Taken together, these results suggested that MIAT may be involved in the differentiation of ADMSCs into LECs, and that the MIAT/miR­495/Prox1 axis may be a novel regulatory mechanism and therapeutic target for the treatment of lymphedema.


Assuntos
Proteínas de Homeodomínio/genética , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Infarto do Miocárdio/genética , RNA Longo não Codificante/genética , Proteínas Supressoras de Tumor/genética , Animais , Diferenciação Celular/genética , Movimento Celular/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas de Transporte Vesicular/genética
16.
Exp Ther Med ; 21(3): 232, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33603840

RESUMO

Accumulating evidence indicates that the long non-coding RNA myocardial infarction associated transcript (lncRNA MIAT) serves an important role in the progression of a number of cancer types. However, the precise molecular mechanism of MIAT in laryngeal squamous cell carcinoma (LSCC) progression remain elusive. The aim of the current study was to assess the effects and to clarify the molecular mechanism of MIAT on the proliferation and invasion of LSCC cells. The expression of MIAT was detected in LSCC tissues and cells using reverse transcription-quantitative PCR. MTT and colony formation assays were performed to examine the effects of MIAT on the proliferation of LSCC cells. Additionally, wound healing and Transwell experiments were employed to examine cellular migration and invasion. Luciferase reporter gene assay was also used to confirm the direct binding between MIAT and microRNA (miR)-613 in LSCC cells. An RNA immunoprecipitation assay was performed to verify the interaction between MIAT and miR-613. In the present study, it was found that the expression of MIAT in LSCC tissues was markedly higher compared with that in adjacent non-tumor tissues. In addition, MIAT expression was also increased in the human LSCC cell lines TU686, TU-177 and AMC-HN-8 compared with that in normal human keratinocytes (HaCaT). Knocking down MIAT expression significantly reduced LSCC cell proliferation and inhibited colony formation, a shown by MTT and colony formation assays, respectively. MIAT knockdown also substantially inhibited the migratory and invasive abilities of LSCC cells, as shown by wound healing and Transwell invasion assays, respectively. Subsequently, luciferase reporter assays verified that MIAT could bind to miR-613, where a negative correlation was observed between the expression of MIAT and miR-613 in LSCC tissues. Suppression of miR-613 partially reversed the inhibitory effects of MIAT knockdown on the proliferation, migration and invasion of LSCC cells. Taken together, the present study identified that MIAT may function as an oncogenic lncRNA to promote LSCC progression, which provides a potential therapeutic target or as a novel diagnostic biomarker for LSCC.

17.
Neural Regen Res ; 16(9): 1877-1881, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33510096

RESUMO

Whether long non-coding RNA myocardial infarction-associated transcript is involved in oxygen-induced retinopathy remains poorly understood. To validate this hypothesis, we established a newborn mouse model of oxygen-induced retinopathy by feeding in an oxygen concentration of 75 ± 2% from postnatal day 8 to postnatal day 12, followed by in normal air. On postnatal day 11, the mice were injected with the myocardial infarction-associated transcript siRNA plasmid via the vitreous cavity to knockdown long non-coding RNA myocardial infarction-associated transcript. Myocardial infarction-associated transcript siRNA transcription significantly inhibited myocardial infarction-associated transcript mRNA expression, reduced the phosphatidylinosital-3-kinase, phosphorylated Akt and vascular endothelial growth factor immunopositivities, protein and mRNA expression, and alleviated the pathological damage to the retina of oxygen-induced retinopathy mouse models. These findings suggest that myocardial infarction-associated transcript is likely involved in the retinal neovascularization in retinopathy of prematurity and that inhibition of myocardial infarction-associated transcript can downregulate phosphatidylinosital-3-kinase, phosphorylated Akt and vascular endothelial growth factor expression levels and inhibit neovascularization. This study was approved by the Animal Ethics Committee of Shengjing Hospital of China Medical University, China (approval No. 2016PS074K) on February 25, 2016.

18.
Biomed Pharmacother ; 133: 111040, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33378948

RESUMO

Myocardial Infarction Associated Transcript (MIAT) is a non-coding transcript which is located on chromosome 22q12.1. This lncRNA can regulate expression of genes at both transcriptional and post-transcriptional stages. It has been firstly recognized as a susceptibility locus for myocardial infarction. Subsequently, its role in the development of several human cancers has been acknowledged. Numerous researches have reported the impact of MIAT silencing on the reduction of cell viability, proliferation and invasion while enhancement of cellular senescence and apoptosis. Consistently, investigations in the xenograft models have verified MIAT role in the promotion of tumor growth. Numerous microRNAs such as miR-214, miR-22-3p, miR-520d-3p, miR-203a, miR-29a-3p, miR-141, miR-150, miR-302, miR-29, and miR-155-5p have functional interactions with this lncRNA. Moreover, dysregulation of MIAT has been associated with abnormal activity of numerous cancer-related signaling cascades such as Hippo, PI3K/Akt/c-Met and Wnt/ß-catenin. In the current review, we explain the role of MIAT in the cancer evolution based on the outcomes of in vitro, in vivo and clinical studies.


Assuntos
Neoplasias/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Invasividade Neoplásica , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/patologia , RNA Longo não Codificante/genética , Transdução de Sinais , Carga Tumoral
19.
Methods Mol Biol ; 2176: 49-56, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32865781

RESUMO

Long noncoding RNAs (lncRNAs) are a class of RNA with 200 nucleotides or longer that are not translated into protein. lncRNAs are highly abundant; a study estimates that at least four times more lncRNAs are typically present than coding RNAs in humans. However, function of more than 95% of human lncRNAs are still unknown. Synthetic antisense oligonucleotides called gapmers are powerful tools for lncRNA loss-of-function studies. Gapmers contain a central DNA part, which activates RNase H-mediated RNA degradation, flanked by modified oligonucleotides, such as 2'-O-methyl RNA (2'OMe), 2'-O-methoxyethyl RNA (2'MOE), constrained ethyl nucleosides (cEt), and locked nucleic acids (LNAs). In contrast to siRNA or RNAi-based methods, antisense oligonucleotide gapmer-based knockdown is often more effective against nuclear-localized lncRNA targets, since RNase H is mainly localized in nuclei. As such, gapmers are also potentially a powerful tool for therapeutics targeting lncRNAs in various diseases, including cancer, cardiovascular diseases, lung fibrosis, and neurological/neuromuscular diseases. This chapter will discuss the development and applications of gapmers for lncRNA loss-of-function studies and tips to design effective antisense oligonucleotides.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Oligonucleotídeos Antissenso , RNA Longo não Codificante/genética , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Técnicas de Silenciamento de Genes/história , Terapia Genética/história , Terapia Genética/métodos , História do Século XX , História do Século XXI , Humanos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Neoplasias/genética , Neoplasias/terapia , Oligonucleotídeos Antissenso/síntese química , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , Fibrose Pulmonar/genética , Fibrose Pulmonar/terapia , RNA Longo não Codificante/antagonistas & inibidores
20.
Exp Ther Med ; 20(2): 1053-1063, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32742346

RESUMO

Age-related cataract (ARC) is a common cause of blindness in elderly individuals. Long non-coding RNA (lncRNA) myocardial infarction associated transcript (MIAT) has been reported to participate in various biological processes in a number of diseases; however, the biological mechanism underlying MIAT during ARC is not completely understood. The expression levels of MIAT, microRNA (miR)-181a and connective tissue growth factor (CTGF) were measured by reverse transcription-quantitative PCR. The protein expression levels of CTGF, α-smooth muscle actin, fibronectin, collagen type I, ERK, phosphorylated (p)-ERK, mitogen-activated protein kinase (MEK), and p-MEK were detected by western blotting. Cell viability and migration were assessed using MTT and Transwell assays, respectively. Moreover, a dual-luciferase reporter assay was performed to investigate the interaction between miR-181a and MIAT or CTGF. MIAT and CTGF were upregulated, while miR-181a was significantly downregulated in ARC tissues compared with normal tissues. MIAT or CTGF knockdown decreased cell viability, migration, epithelial-mesenchymal transition and extracellular matrix production in TGF-ß2-treated SRA01/04 cells. It was hypothesized that miR-181a may be sponged by MIAT and may target CTGF. Furthermore, the miR-181a inhibitor reversed the inhibitory effect of MIAT knockdown on the progression of TGF-ß2-treated SRA01/04 cells. Moreover, CTGF knockdown also reversed MIAT overexpression-mediated progression of TGF-ß2-treated SRA01/04 cells. In addition, MIAT and CTGF regulated the activity of the ERK signaling pathway. The results suggested that MIAT may regulate the progression of ARC via the miR-181a/CTGF/ERK signaling pathway, which may serve as a novel therapeutic target for ARC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...