Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
1.
J Cell Mol Med ; 28(13): e18527, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38984969

RESUMO

Infected bone defects (IBDs) are the common condition in the clinical practice of orthopaedics. Although surgery and anti-infective medicine are the firstly chosen treatments, in many cases, patients experience a prolonged bone union process after anti-infective treatment. Epimedium-Curculigo herb pair (ECP) has been proved to be effective for bone repair. However, the mechanisms of ECP in IBDs are insufficiency. In this study, Effect of ECP in IBDs was verified by micro-CT and histological examination. Qualitative and quantitative analysis of the main components in ECP containing medicated serum (ECP-CS) were performed. The network pharmacological approaches were then applied to predict potential pathways for ECP associated with bone repair. In addition, the mechanism of ECP regulating LncRNA MALAT1/miRNA-34a-5p/SMAD2 signalling axis was evaluated by molecular biology experiments. In vivo experiments indicated that ECP could significantly promote bone repair. The results of the chemical components analysis and the pathway identification revealed that TGF-ß signalling pathway was related to ECP. The results of in vitro experiments indicated that ECP-CS could reverse the damage caused by LPS through inhibiting the expressions of LncRNA MALAT1 and SMAD2, and improving the expressions of miR-34a-5p, ALP, RUNX2 and Collagen type І in osteoblasts significantly. This research showed that ECP could regulate the TGF-ß/SMADs signalling pathway to promote bone repair. Meanwhile, ECP could alleviate LPS-induced bone loss by modulating the signalling axis of LncRNA MALAT1/miRNA-34a-5p/ SMAD2 in IBDs.


Assuntos
Epimedium , MicroRNAs , Osteoblastos , RNA Longo não Codificante , Transdução de Sinais , Proteína Smad2 , MicroRNAs/genética , MicroRNAs/metabolismo , Osteoblastos/metabolismo , Osteoblastos/efeitos dos fármacos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Animais , Proteína Smad2/metabolismo , Proteína Smad2/genética , Camundongos , Epimedium/química , Transdução de Sinais/efeitos dos fármacos , Masculino , Regeneração Óssea/efeitos dos fármacos , Humanos , Regulação da Expressão Gênica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética
2.
BMC Endocr Disord ; 24(1): 28, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38439031

RESUMO

OBJECTIVE: This study aimed to examine the diagnostic predictive value of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1(MALAT1) and NOD-like receptor protein 3(NLRP3) expression in patients with type 2 diabetes mellitus(T2DM) and lower extremity atherosclerosis disease (LEAD). METHODS: A total of 162 T2DM patients were divided into T2DM with LEAD group (T2DM + LEAD group) and T2DM alone group (T2DM group). The lncRNA MALAT1 and NLRP3 expression levels were measured in peripheral blood, and their correlation was examined. Least absolute shrinkage and selection operator (LASSO) regression model was used to screen for the best predictors of LEAD, and multivariate logistic regression was used to establish a predictive model and construct the nomogram. The effectiveness of the nomogram was assessed using the receiver operating characteristic (ROC) curve, area under the curve (AUC), calibration curve, and decision curve analysis (DCA). RESULTS: The levels of the lncRNA MALAT1 and NLRP3 in the T2DM + LEAD group were significantly greater than those in the T2DM group (P <0.001), and the level of the lncRNA MALAT1 was positively correlated with that of NLRP3 (r = 0.453, P<0.001). The results of the LASSO combined with the logistic regression analysis showed that age, smoking, systolic blood pressure (SBP), NLRP3, and MALAT1 were the influencing factors of T2DM with LEAD(P<0.05). ROC curve analysis comparison: The discriminatory ability of the model (AUC = 0.898), MALAT1 (AUC = 0.804), and NLRP3 (AUC = 0.794) was greater than that of the other indicators, and the predictive value of the model was the greatest. Calibration curve: The nomogram model was consistent in predicting the occurrence of LEAD in patients with T2DM (Cindex = 0.898). Decision curve: The net benefit rates obtained from using the predictive models for clinical intervention decision-making were greater than those obtained from using the individual factors within the model. CONCLUSION: MALAT1 and NLRP3 expression increased significantly in T2DM patients with LEAD, while revealing the correlation between MALAT1 and NLRP3. The lncRNA MALAT1 was found as a potential biomarker for T2DM with LEAD.


Assuntos
Aterosclerose , Diabetes Mellitus Tipo 2 , RNA Longo não Codificante , Humanos , Aterosclerose/diagnóstico , Aterosclerose/genética , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Extremidade Inferior , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , RNA Longo não Codificante/genética
3.
Exp Cell Res ; 429(1): 113650, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37209990

RESUMO

The retina may undergo structural and functional damage as a result of hypoxia, which could lead to permanent blindness. As competing endogenous RNAs (ceRNAs), lncRNAs are essential in eye disorders. The biological function of lncRNA MALAT 1 and its potential mechanisms in hypoxic-ischemic retinal diseases are still unknown. MALAT 1 and miR-625-3p expression alterations in hypoxia-treated RPE cells were examined using qRT-PCR. The target binding relationships between MALAT 1 and miR-625-3p, as well as between miR-625-3p and HIF-1α, were identified utilizing bioinformatics analysis and dual luciferase reporter assay. We observed that si-MALAT 1 and miR-625-3p mimic both reduced apoptosis and epithelial-mesenchymal transition (EMT) in hypoxic RPE cells, whereas si-MALAT 1 was reversed by miR-625-3p inhibitor. Further, we carried out a mechanistic investigation, and rescue assays demonstrated that MALAT 1 sponging miR-625-3p influenced HIF-1α expression and consequently took part in the NF-κB/Snail signaling pathway, which regulated apoptosis and EMT. In conclusion, our research had shown that the MALAT 1/miR-625-3p/HIF-1α axis drove the progression of hypoxic-ischemic retinal disorders and may serve as a promising predictive biomarker for their therapeutic and diagnostic targets.


Assuntos
MicroRNAs , RNA Longo não Codificante , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , Transição Epitelial-Mesenquimal/genética , Movimento Celular/genética , Transdução de Sinais/genética , Hipóxia , Proliferação de Células/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo
4.
Clin Oral Investig ; 28(4): 219, 2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38492123

RESUMO

OBJECTIVES: This study aimed to investigate the regulatory roles of lncRNA MALAT1, miR-124-3p, and IGF2BP1 in osteogenic differentiation of periodontal ligament stem cells (PDLSCs). MATERIALS AND METHODS: We characterized PDLSCs by employing quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses to evaluate the expression of key osteogenic markers including ALPL, SPP1, and RUNX2. Manipulation of lncRNA MALAT1 and miR-124-3p expression levels was achieved through transfection techniques. In addition, early osteogenic differentiation was assessed via Alkaline phosphatase (ALP) staining, and mineral deposition was quantified using Alizarin Red S (ARS) staining. Cellular localization of lncRNA MALAT1 was determined through Fluorescence In Situ Hybridization (FISH). To elucidate the intricate regulatory network, we conducted dual-luciferase reporter assays to decipher the binding interactions between lncRNA MALAT1 and miR-124-3P as well as between miR-124-3P and IGF2BP1. RESULTS: Overexpression of lncRNA MALAT1 robustly promoted osteogenesis in PDLSCs, while its knockdown significantly inhibited the process. We confirmed the direct interaction between miR-124-3p and lncRNA MALAT1, underscoring its role in impeding osteogenic differentiation. Notably, IGF2BP1 was identified as a direct binding partner of lncRNA MALAT1, highlighting its pivotal role within this intricate network. Moreover, we determined the optimal IGF2BP1 concentration (50 ng/ml) as a potent enhancer of osteogenesis, effectively countering the inhibition induced by si-MALAT1. Furthermore, in vivo experiments utilizing rat calvarial defects provided compelling evidence, solidifying lncRNA MALAT1's crucial role in bone formation. CONCLUSIONS: Our study reveals the regulatory network involving lncRNA MALAT1, miR-124-3p, and IGF2BP1 in PDLSCs' osteogenic differentiation. CLINICAL RELEVANCE: These findings enhance our understanding of lncRNA-mediated osteogenesis, offering potential therapeutic implications for periodontal tissue regeneration and the treatment of bone defects.


Assuntos
MicroRNAs , RNA Longo não Codificante , Ratos , Animais , Osteogênese/fisiologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Ligamento Periodontal , Hibridização in Situ Fluorescente , MicroRNAs/genética , MicroRNAs/metabolismo , Diferenciação Celular/genética , Células-Tronco , Células Cultivadas
5.
Neurochem Res ; 48(11): 3457-3471, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37470906

RESUMO

The objective of this research was to investigate the role of lncRNA MALAT1 and HSP90 in the regulation of neuronal necroptosis in mice with cerebral ischemia-reperfusion (CIR). We used male C57BL/6J mice to establish a middle cerebral artery occlusion (MCAO) model and conducted in vitro experiments using the HT-22 mouse hippocampal neuron cell line. The cellular localization of NeuN and MLKL, as well as the expression levels of neuronal necroptosis factors, MALAT1, and HSP90 were analyzed. Cell viability and necroptosis were assessed, and we also investigated the relationship between MALAT1 and HSP90. The results showed that MALAT1 expression increased after MCAO and oxygen-glucose deprivation/re-oxygenation (OGD/R) treatment in both cerebral tissues and cells compared with the control group. The levels of neuronal necroptosis factors and the co-localization of NeuN and MLKL were also increased in MCAO mice compared with the Sham group. MALAT1 was found to interact with HSP90, and inhibition of HSP90 expression led to decreased phosphorylation levels of neuronal necroptosis factors. Inhibition of MALAT1 expression resulted in decreased co-localization levels of NeuN and MLKL, decreased phosphorylation levels of neuronal necroptosis factors, and reduced necroptosis rate in cerebral tissues. Furthermore, inhibiting MALAT1 expression also led to a shorter half-life of HSP90, increased ubiquitination level, and decreased phosphorylation levels of neuronal necroptosis factors in cells. In conclusion, this study demonstrated that lncRNA MALAT1 promotes neuronal necroptosis in CIR mice by stabilizing HSP90.


Assuntos
Isquemia Encefálica , Proteínas de Choque Térmico HSP90 , RNA Longo não Codificante , Traumatismo por Reperfusão , Animais , Masculino , Camundongos , Apoptose/genética , Isquemia Encefálica/metabolismo , Glucose/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Necroptose , Neurônios/metabolismo , Reperfusão , Traumatismo por Reperfusão/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo
6.
J Nanobiotechnology ; 21(1): 451, 2023 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-38012570

RESUMO

Spinal cord injury (SCI) is a traumatic condition of the central nervous system that causes paralysis of the limbs. Micro electric fields (EF) have been implicated in a novel therapeutic approach for nerve injury repair and regeneration, but the effects of human umbilical cord mesenchymal stem cell-derived small extracellular vesicles that are induced by micro electric fields (EF-sEVs) stimulation on SCI remain unknown. The aim of the present study was to investigate whether EF-sEVs have therapeutic effects a rat model of SCI. EF-sEVs and normally conditioned human umbilical cord mesenchymal stem cells-derived small extracellular vesicles (CON-sEVs) were collected and injected intralesionally into SCI model rats to evaluate the therapeutic effects. We detect the expression of candidate long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (lncRNA-MALAT1) in EF-sEVs and CON-sEVs. The targets and downstream effectors of lncRNA-MALAT1 were investigated using luciferase reporter assays. Using both in vivo and in vitro experiments, we demonstrated that EF-sEVs increased autophagy and decreased apoptosis after SCI, which promoted the recovery of motor function. We further confirmed that the neuroprotective effects of EF-sEVs in vitro and in vivo correlated with the presence of encapsulated lncRNA-MALAT1 in sEVs. lncRNA-MALAT1 targeted miR-22-3p via sponging, reducing miR-22-3p's suppressive effects on its target, SIRT1, and this translated into AMPK phosphorylation and increased levels of the antiapoptotic protein Bcl-2. Collectively, the present study identified that the lncRNA-MALAT1 in EF-sEVs plays a neuroprotective role via the miRNA-22-3p/SIRT1/AMPK axis and offers a fresh perspective and a potential therapeutic approach using sEVs to improve SCI.


Assuntos
Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Traumatismos da Medula Espinal , Ratos , Humanos , Animais , RNA Longo não Codificante/metabolismo , Proteínas Quinases Ativadas por AMP , Sirtuína 1/genética , Sirtuína 1/metabolismo , Apoptose , Traumatismos da Medula Espinal/metabolismo , MicroRNAs/metabolismo , Autofagia
7.
Oral Dis ; 29(5): 2012-2026, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35467063

RESUMO

OBJECTIVES: Methyltransferase-like 14 (METTL14) plays an epigenetic role in various cancer through N6-methyladenosine (m6A) modification. This study sought to analyze the mechanism of METTL14 in oral squamous cell carcinoma (OSCC) cell proliferation. METHODS: Expression levels of METTL14, lncRNA metastasis associated with lung adenocarcinoma transcript 1 (lncRNA MALAT1), microRNA (miR)-224-5p, and histone lysine demethylase 2A (KDM2A) in OSCC tissues (N = 40), and cell lines (FaDu, SCC-25, CAL-27, and SCC-15) were detected. Cell viability and colony formation capacity were assessed. m6A level, stability, and subcellular localization of lncRNA MALAT1 were determined. Nude mouse xenograft tumor assay was performed to confirm the role of METTL14 in vivo. RESULTS: METTL14 and lncRNA MALAT1 were upregulated, and miR-224-5p was downregulated in OSCC tissues and cells. Silencing METTL14 repressed OSCC cell viability and colony formation. Overexpression of MALAT1 and KDM2A or miR-224-5p downregulation reversed the inhibition of silencing METTL14 on OSCC cell proliferation. METTL14 induced m6A modification of MALAT1 to upregulate MALAT1. MALAT1 is comparatively bound to miR-224-5p to promote KDM2A transcription. In vivo, METTL14 promoted tumor growth via regulating MALAT1/miR-224-5p/ KDM2A. CONCLUSIONS: Overall, our findings verified the therapeutic role of silencing METTL14 in OSCC treatment through the MALAT1/miR-224-5p/KDM2A axis.


Assuntos
Carcinoma de Células Escamosas , Proteínas F-Box , Neoplasias de Cabeça e Pescoço , MicroRNAs , Neoplasias Bucais , RNA Longo não Codificante , Camundongos , Animais , Humanos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias de Cabeça e Pescoço/genética , Regulação Neoplásica da Expressão Gênica , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Metiltransferases/genética
8.
Int J Neurosci ; 133(7): 740-753, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34461809

RESUMO

PURPOSE: To investigate the regulation and related mechanisms of MALAT1 in cerebral ischemia- reperfusion (CI/R) injury. MATERIALS AND METHODS: 72 mice were divided into sham group (n=24), MCAO group (n=24), MCAO+pcDNA-NC group (n=12) and MCAO+MALAT1 group (n=12). At 12 h, 24 h and 48 h after reperfusion, 6 mice were randomly selected from the sham group and the MCAO group to detect the expression of MALAT1, miR-142-3p and SIRT1 in brain tissue. All mice were scored for neurobehavioral after 48 h of reperfusion. After the completion of the scoring, 6 mice were randomly selected from each group and brain tissue was obtained for TTC analysis. The remaining mice of each group were kept on the Morris water maze test after 3 days of feeding. TTC staining and cerebral infarct volume determination. The infarct size of each brain slice was calculated using Image J image analysis software. OGD/R model PC12 cells were prepared according to simulating CI/R injury in vitro. MALAT1 was cloned into the pcDNA3.1 to construct a MALAT1 overexpression vector with the empty vector NC as a control. Plasmid or oligonuceotides were transfected into PC12 cells. The content of TNF-α, IL-1ß, IL-6, the content of reactive oxygen species (ROS), malondialdehyde (MDA) in brain tissue was detected. The activity of superoxide dismutase (SOD), catalase (CAT) activity was measured. RESULTS: MALAT1 was down-regulated in a time-dependent manner in CI/R-damaged mouse cerebral cortex and OGD/R-induced PC12 cells, accompanied by an increase in the expression of miR-142-3p and a decrease in sirtuin 1 (SIRT1) expression. Overexpression of MALAT1 inhibited OGD/R-induced cell necrosis and apoptosis and promoted cell proliferation. Overexpression of MALAT1 reduced the levels of TNF-α, IL-6, IL-1ß, ROS and MDA and increased the activities of SOD and CAT in OGD/R-injured PC12 cells. MALAT1 negatively regulated the expression of miR-142-3p, and SIRT1 was a target gene of miR-142-3p. The expression of SIRT1 induced by MALAT1 overexpression was obviously abolished by the introduction of miR-142-3p mimic. MALAT1 overexpression can exert its role by regulating the miR-142-3p/SIRT1 axis. Besides, overexpression of MALAT1 improved cerebral infarction, neurological impairment and cognitive dysfunction in CI/R mice. CONCLUSION: MALAT1 mediates SIRT1 expression by acting as a ceRNA of miR-142-3p to improve CI/R injury.Abbreviations: CAT: catalase; CI/R: cerebral ischemia-reperfusion; IL-1ß: interleukin-1ß; IL-6: interleukin-6; lncRNA: long-chain non-coding RNA; MALAT1: metastasis-associated lung adenocarcinoma transcript1; MCAO: middle cerebral artery occlusion; MDA: malondialdehyde; OGD/R: oxygen-glucose deprivation and reoxygenation; ROS: reactive oxygen species; SIRT1: sirtuin 1; SOD: superoxide dismutase; TNF-α: tumour necrosis factor-alpha.


Assuntos
Isquemia Encefálica , Disfunção Cognitiva , MicroRNAs , RNA Longo não Codificante , Traumatismo por Reperfusão , Ratos , Camundongos , Animais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima , Catalase/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fator de Necrose Tumoral alfa/genética , Interleucina-6/metabolismo , Traumatismo por Reperfusão/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Disfunção Cognitiva/genética , Apoptose
9.
Molecules ; 28(5)2023 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-36903369

RESUMO

LncRNAs are a group of non-coding RNA transcripts with lengths of over 200 nucleotides and can interact with DNA, RNA, and proteins to regulate gene expression of malignant tumors in human tissues. LncRNAs participate in vital processes, such as chromosomal nuclear transport in the cancerous site of human tissue, activation, and the regulation of proto-oncogenes, the differentiation of immune cells, and the regulation of the cellular immune system. The lncRNA metastasis-associated lung cancer transcript 1 (MALAT1) is reportedly involved in the occurrence and development of many cancers and serves as a biomarker and therapeutic target. These findings highlight its promising role in cancer treatment. In this article, we comprehensively summarized the structure and functions of lncRNA, notably the discoveries of lncRNA-MALAT1 in different cancers, the action mechanisms, and the ongoing research on new drug development. We believe our review would serve as a basis for further research on the pathological mechanism of lncRNA-MALAT1 in cancer and provide evidence and novel insights into its application in clinical diagnoses and treatments.


Assuntos
Neoplasias , RNA Longo não Codificante , Humanos , Linhagem Celular Tumoral , RNA Longo não Codificante/genética , Neoplasias/patologia
10.
J Gene Med ; 24(6): e3409, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35030644

RESUMO

BACKGROUND: Accumulating evidence demonstrates that long non-coding RNAs (lncRNAs) are associated with the development of osteoporosis. The present study aimed to investigate the effect of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) on osteogenic differentiation in osteoporosis. METHODS: MALAT1 levels were detected by a real-time polymerase chain reaction (RT-qPCR). Moreover, the levels of osteogenic differentiation-related factors (Bmp4, Col1a1 and Spp1) were measured by a RT-qPCR and western blotting. Alkaline phosphatase (ALP) activity was detected using an ALP staining assay. RESULTS: MALAT1 levels were downregulated in hindlimb unloading mice and simulated in microgravity (MG) treated MC3T3-E1 cells. Moreover, MG treatment induced the downregulation of the expression of ALP, BMP4, Col1a1 and Spp1, whereas overexpression of MALAT1 abolished the downregulation. MG also inhibited ALP activity, whereas MALAT1 reversed the effect. Furthermore, miR-217 was identified as a target of MALAT1, and AKT3 was verified as a target of miR-217. Overexpression of miR-217 rescued the promotion of osteogenic differentiation induced by MALAT1 in MG treated cells. Knockdown of AKT3 abolished the facilitation of osteogenic differentiation induced by downregulation of miR-217. CONCLUSIONS: MALAT1 promotes osteogenic differentiation through regulating miR-217/AKT3 axis, suggesting that MALAT1 is a potential target with respect to alleviating osteoporosis.


Assuntos
MicroRNAs/antagonistas & inibidores , Osteoporose , RNA Longo não Codificante/metabolismo , Animais , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Osteogênese , Osteoporose/patologia , Osteoporose/terapia , RNA Longo não Codificante/genética
11.
BMC Cardiovasc Disord ; 22(1): 152, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35392816

RESUMO

BACKGROUND: Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) participates in the occurrence and development of cardiovascular and cerebrovascular diseases such as stroke and coronary heart disease by regulating inflammatory reactions, programmed cell death, and other pathological processes. Previous studies revealed that the MALAT1 gene polymorphism was associated with cardiac and cerebrovascular diseases. However, the prognostic role of the MALAT1 polymorphism in major adverse cardiac and cerebrovascular events (MACCEs) remains unknown. Therefore, this study intends to explore the association between the MALAT1 rs3200401 polymorphism and MACCEs. METHOD: We enrolled 617 myocardial infarction (MI) patients and 1125 control participants who attended the First Affiliated Hospital of Xinjiang Medical University from January 2010 to 2018. SNPscan™ typing assays were used to detect the MALAT1 rs3200401 genotype. During the follow-up, MACCEs were recorded. Kaplan-Meier curves and univariate and multivariate Cox survival analyses were used to explore the correlation between MALAT1 gene polymorphisms and the occurrence of MACCEs. RESULTS: Among the total participants and MI patients, the frequencies of the T allele (total Participants 19.5% vs. 15.3%, P = 0.047, MI patients 20.7% vs. 14.1%, P = 0.014) and CT + TT genotypes (total Participants 37.4% vs. 28.1%, P = 0.013, MI patients 39.5% vs. 25.8%, P = 0.003) were significantly higher in subjects with MACCEs than in subjects without MACCEs. However, in control participants, the frequencies of the T allele (16.6% vs. 16.0%, P = 0.860) and CT + TT genotypes (31.4% vs. 29.3%, P = 0.760) were not higher in subjects with MACCEs than in subjects without MACCEs. In addition, among the total participants and MI patients, the Kaplan-Meier curve analysis indicated that the subjects with rs3200401 CT + TT genotypes had a higher incidence of MACCEs than CC genotype carriers (P = 0.015, P = 0.001). Nevertheless, similar results were not observed in the control participants (P = 0.790). Multivariate Cox regression indicated that compared with patients with the CC genotype, patients with CT + TT genotypes had a 1.554-fold increase in MACCE risk (hazard ratio: 1.554, 95% confidence interval: 1.060-2.277, P = 0.024). CONCLUSIONS: The MALAT1 rs3200401 CT + TT genotypes could be a risk factor for MACCEs in MI patients, suggesting that the MALAT1 gene may become a biomarker for poor prognosis in MI patients.


Assuntos
Infarto do Miocárdio , RNA Longo não Codificante , Humanos , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/genética , Polimorfismo de Nucleotídeo Único , Prognóstico , RNA Longo não Codificante/genética
12.
J Clin Lab Anal ; 36(2): e24215, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35028972

RESUMO

BACKGROUND: Primary open-angle glaucoma (POAG) is the commonest form of glaucoma which is estimated to cause bilaterally blind within 11.1 million people by 2020. Therefore, the primary objectives of this study were to investigate the clinical significance of single-nucleotide polymorphisms (SNPs) in the lncRNAs MALAT1 and ANRIL in a Chinese Han POAG cohort. METHODS: Three hundred and forty-six glaucoma patients and 263 healthy controls were recruited, and totally 14 SNPs in MALAT1 and ANRIL were genotyped between the two populations. RESULTS: The MALAT1 SNPs rs619586 (A>G), rs3200401 (C>T), and rs664589 (C>G) were associated with POAG risk, and the ANRIL SNPs rs2383207 (A>G), rs564398 (A>G), rs2157719 (A>G), rs7865618 (G>A), and rs4977574 (A>G) were associated with POAG (p < 0.05). The MALAT1 haplotypes ACG and ATC, comprised rs619586, rs3200401, and rs664589, increased POAG risk, and the ANRIL haplotype AAGAA, made up of rs2383207, rs7865618, rs4977574, rs564398, and rs2157719, show a significantly increased risk of POAG. In addition, rs619586 (A>G) of MALAT1 and rs564398/rs2157719 of ANRIL were associated with a smaller vertical cup-to-disc ratio, while rs619586 of MALAT1 and rs2383207/rs4977574 of ANRIL were associated with higher intraocular pressure in the POAG population. CONCLUSION: Single-nucleotide polymorphisms and haplotypes in ANRIL and MALAT1 were associated with POAG onset in our study population, which provide more possibilities to POAG diagnosis and treatment.


Assuntos
Glaucoma de Ângulo Aberto/genética , Haplótipos , Polimorfismo de Nucleotídeo Único , RNA Longo não Codificante , Idoso , Estudos de Casos e Controles , China , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade
13.
World J Surg Oncol ; 20(1): 203, 2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35706002

RESUMO

BACKGROUND: This study aimed to explore the specific pathogenesis of lncRNA MALAT1 promoting the invasion and metastasis of hepatocellular carcinoma (HCC) through peripheral blood vessels by regulating the expression of miRNA-613 molecule. METHODS: The data of 60 HCC metastatic patients and 60 HCC non-metastatic patients detected by the contrast-enhanced ultrasound (CEUS) in the Second Affiliated Hospital of Qiqihar Medical College from January 2020 to June 2021 were collected, as well as postoperatively retained HCC tissues and paired paracancer tissues (5 cm laterally from the edge of the cancer area), to study the changes of microangiogenesis in HCC tissues with CEUS. The correlation between CEUS grading and lncRNA MALAT1 in patients with HCC was analyzed through Pearson correlation analysis, lncRNA MALAT1 and miRNA-613 in HCC tissues of patients with HCC were detected by qRT-PCR, followed by the bioinformatic analysis for the relationship between lncRNA MALAT1 and miRNA-613. The Log-growing human HCC cell strain, HepG2, was selected for experiments. Adenovirus transfection knocked down lncRNA MALAT1 in HCC cells, which was divided into two groups (inhibitor-NC group and lncR-inhibitor group), followed by knocking down miRNA-613 on the basis of knocking down lncRNA MALAT1, which was divided into three groups (inhibitor-NC group, lncR-inhibitor groups, and lncR/miR613-inhibitor group). The expression of miRNA-613 and lncRNA MALAT1 in each group was detected by qRT-PCR. The migration and invasiveness of cells in each group were detected by Transwell assay. RESULTS: CEUS of HCC and Pearson correlation analysis showed that CEUS grading and lncRNA MALAT1 were positively correlated in patients with HCC. In HCC tissues of patients with HCC, lncRNA MALAT1 expressed high and miRNA-613 expressed low. The results of bioinformatic analysis showed the targeting of lncRNA MALAT1 and miRNA-613. Knocking down lncRNA MALAT1 could increase miRNA-613 expression significantly, and reduce the migration of HCC cells. Inhibiting miRNA-613 based on knocking down lncRNA MALAT1 could increase the survival and migration of HCC cells. CONCLUSIONS: lncRNA MALAT1 can promote HCC metastasis through the peripheral vascular infiltration by inhibiting the level of MiRNA-613, which can, therefore, be used as a potential target for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Metástase Neoplásica , RNA Longo não Codificante/genética
14.
Cancer Cell Int ; 21(1): 70, 2021 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-33482814

RESUMO

BACKGROUND: Gastric cancer is one of the most prevalent and deadly malignancies without efficient treatment option. This study aimed to investigate the effect of hydrogen gas on the behavior of gastric cancer cells. METHODS: Gastric cancer cell lines MGC-803 and BGC-823 were treated with or without H2 /O2 gas mixture (66.7%:33.3% v/v). Proliferation and migration were assessed by MTT and scratch wound healing assays respectively. The expression of lncRNA MALAT1, miR-124-3p, and EZH2 was analyzed by real-time quantitative PCR and/or western blot. Tumor growth was estimated using xenograft mouse model. RESULTS: H2 gas significantly inhibited gastric tumor growth in vivo and the proliferation, migration, and lncRNA MALAT1 and EZH2 expression of gastric cancer cells while upregulated miR-124-3p expression. LncRNA MALAT1 overexpression abolished all the aforementioned effects of H2. LncRNA MALAT1 and miR-124-3p reciprocally inhibited the expression of each other. MiR-124-3p mimics abrogated lncRNA MALAT1 promoted EZH2 expression and gastric cancer cell proliferation and migration. CONCLUSIONS: These data demonstrated that H2 might be developed as a therapeutics of gastric cancer and lncRNA MALAT1/miR-124-3p/EZH2 axis could be a target for intervention.

15.
FASEB J ; 34(4): 5282-5298, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32067273

RESUMO

Melatonin is a hormone produced by the pineal gland, and it has extensive beneficial effects on various tissue and organs; however, whether melatonin has any effect on cardiac fibrosis in the pathogenesis of diabetic cardiomyopathy (DCM) is still unknown. Herein, we found that melatonin administration significantly ameliorated cardiac dysfunction and reduced collagen production by inhibiting TGF-ß1/Smads signaling and NLRP3 inflammasome activation, as manifested by downregulating the expression of TGF-ß1, p-Smad2, p-Smad3, NLRP3, ASC, cleaved caspase-1, mature IL-1ß, and IL-18 in the heart of melatonin-treated mice with diabetes mellitus (DM). Similar beneficial effects of melatonin were consistently observed in high glucose (HG)-treated cardiac fibroblasts (CFs). Moreover, we also found that lncRNA MALAT1 (lncR-MALAT1) was increased along with concomitant decrease in microRNA-141 (miR-141) in DM mice and HG-treated CFs. Furthermore, we established NLRP3 and TGF-ß1 as target genes of miR-141 and lncR-MALAT1 as an endogenous sponge or ceRNA to limit the functional availability of miR-141. Finally, we observed that knockdown of miR-141 abrogated anti-fibrosis action of melatonin in HG-treated CFs. Our findings indicate that melatonin produces an antifibrotic effect via inhibiting lncR-MALAT1/miR-141-mediated NLRP3 inflammasome activation and TGF-ß1/Smads signaling, and it might be considered a potential agent for the treatment of DCM.


Assuntos
Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/patologia , Fibrose/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Cardiopatias/tratamento farmacológico , Melatonina/farmacologia , Animais , Antioxidantes/farmacologia , Cardiomiopatias Diabéticas/epidemiologia , Fibrose/etiologia , Fibrose/metabolismo , Fibrose/patologia , Cardiopatias/etiologia , Cardiopatias/metabolismo , Cardiopatias/patologia , Inflamassomos/antagonistas & inibidores , Inflamassomos/genética , Inflamassomos/metabolismo , Masculino , Camundongos , MicroRNAs/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , RNA Longo não Codificante/genética , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
16.
FASEB J ; 34(1): 66-81, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914639

RESUMO

Angiogenesis is critical for the development, progression, and metastasis of hepatocellular carcinoma (HCC), but the roles of miR-3064-5p in HCC angiogenesis are still unknown. In this study, the roles of miR-3064-5p in HCC angiogenesis were studied in 192 HCC patients, xenograft mouse models, and HCC cell lines. The results showed that miR-3064-5p expression was significantly decreased in HCC tissues and cells, and downregulated miR-3064-5p was associated with upregulated angiogenic potential of HCC. MiR-3064-5p inhibited proangiogenic VEGFA and angiogenin expressions but induced antiangiogenic endostatin and MMP12 expressions, finally leading to suppression of HCC angiogenesis, as shown by the decline in intratumoral microvessel density (MVD). Moreover, miR-3064-5p was inversely correlated with lncRNA MALAT1 and FOXA1. FOXA1 bound to and interacted with CD24 and then regulated Src phosphorylation. MiR-3064-5p played an antiangiogenic role by inhibiting the FOXA1/CD24/Src pathway, whereas oncogenic MALAT1 functioned as a competing endogenous RNA (ceRNA) by sponging miR-3064-5p to alleviate the suppressive effect on the FOXA1 pathway. HCC patients with high miR-3064-5p, low MALAT1, or low FOXA1 expression had a better prognosis with longer overall survival and recurrence-free survival. In univariate and multivariate analyses, miR-3064-5p was identified as the independent prognostic predicator for HCC progression and patient survival. Taken together, miR-3064-5p exerts an antiangiogenic role by targeting the FOXA1/CD24/Src pathway but oncogenic lncRNA MALAT1 acts as a ceRNA to sponge miR-3064-5p. MiR-3064-5p is of great clinical significance and is a novel prognostic indicator and an attractive therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular/irrigação sanguínea , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Neoplasias Hepáticas/irrigação sanguínea , MicroRNAs/metabolismo , Neovascularização Patológica/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Antígeno CD24/genética , Antígeno CD24/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Fator 3-alfa Nuclear de Hepatócito/genética , Humanos , Neoplasias Hepáticas/patologia , Camundongos , MicroRNAs/genética , Neoplasias Experimentais , RNA Longo não Codificante/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
17.
Int J Immunogenet ; 48(5): 419-428, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34291564

RESUMO

With this investigation, we investigated on the contribution of lncRNA MALAT1 to inflammation disorder in Parkinson's Disease (PD). Serum samples were gathered from sporadic PD patients and healthy controls, and single nucleotide polymorphisms (SNPs) of MALAT1, including rs11227209, rs3200401, rs4102217, rs591291, rs619586 and rs664589, were identified. Serum level of MALAT1 was quantified using RT-PCR, and IL-1ß, IL-6, TNF-α and IFN-γ levels in serum were measured with ELISA kits. Inflammation cell models were established by treating PC12 cells with LPS, and cytokine production of pcDNA3.1-MALAT1/si-MALAT1-transfected PC12 cells was evaluated. The results showed that PD patients with high serum level of MALAT1 were associated with lower MMSE score and higher serum levels of IL-1ß, IL-6, TNF-α and IFN-γ than patients carrying low serum level of MALAT1 (p < .05). Mutant alleles of SNPs in MALAT1, including rs3200401 (C>T) and rs4102217 (G>C), tended to elevate PD susceptibility and facilitate cytokine production, as compared with their wild alleles (p < .05). And LPS-exposed PC12 cells secreted larger amounts of inflammation cytokines in the pcDNA3.1-MALAT1 group than in the Mock group (p < .05). In conclusion, MALAT1 participated in modifying inflammation disorder underlying PD aetiology, suggesting that it might be a promising therapeutic target for PD.


Assuntos
Doença de Parkinson , RNA Longo não Codificante/metabolismo , Predisposição Genética para Doença , Humanos , Inflamação/genética , Doença de Parkinson/genética , Polimorfismo de Nucleotídeo Único , RNA Longo não Codificante/genética
18.
J Clin Lab Anal ; 35(8): e23881, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34240756

RESUMO

BACKGROUND: Acute kidney injury (AKI) was characterized by loss of renal function, associated with chronic kidney disease, end-stage renal disease, and length of hospital stay. Long non-coding RNAs (lncRNAs) participated in AKI development and progression. Here, we aimed to investigate the roles and mechanisms of lncRNA MALAT1 in AKI. METHODS: AKI serum samples were obtained from 129 AKI patients. ROC analysis was conducted to confirm the diagnostic value of MALAT1 in differentiating AKI from healthy volunteers. After hypoxic treatment on HK-2 cells, the expressions of inflammatory cytokines, MALAT1, miR-204, APOL1, p65, and p-p65, were measured by RT-qPCR and Western blot assays. The targeted relationship between miR-204 and MALAT1 or miR-204 and APOL1 was determined by luciferase reporter assay and RNA pull-down analysis. After transfection, CCK-8, flow cytometry, and TUNEL staining assays were performed to evaluate the effects of MALAT1 and miR-204 on AKI progression. RESULTS: From the results, lncRNA MALAT1 was strongly elevated in serum samples from AKI patients, with the high sensitivity and specificity concerning differentiating AKI patients from healthy controls. In vitro, we established the AKI cell model after hypoxic treatment. After experiencing hypoxia, we found significantly increased MALAT1, IL-1ß, IL-6, and TNF-α expressions along with decreased miR-204 level. Moreover, the targeted relationship between MALAT1 and miR-204 was confirmed. Silencing of MALAT1 could reverse hypoxia-triggered promotion of HK-2 cell apoptosis. Meanwhile, the increase of IL-1ß, IL-6, and TNF-α after hypoxia treatment could be repressed by MALAT1 knockdown as well. After co-transfection with MALAT1 silencing and miR-204 inhibition, we found that miR-204 could counteract the effects of MALAT1 on HK-2 cell progression and inflammation after under hypoxic conditions. Finally, NF-κB signaling was inactivated while APOL1 expression was increased in HK-2 cells after hypoxia treatment, and lncRNA MALAT1 inhibition reactivated NF-κB signaling while suppressed APOL1 expression by sponging miR-204. CONCLUSIONS: Collectively, these results illustrated that knockdown of lncRNA MALAT1 could ameliorate AKI progression and inflammation by targeting miR-204 through APOL1/NF-κB signaling.


Assuntos
Injúria Renal Aguda/genética , Apolipoproteína L1/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/sangue , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Adulto , Apolipoproteína L1/metabolismo , Estudos de Casos e Controles , Hipóxia Celular/genética , Linhagem Celular , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Testes de Função Renal , Masculino , MicroRNAs/antagonistas & inibidores , Pessoa de Meia-Idade , NF-kappa B/genética , NF-kappa B/metabolismo
19.
Ren Fail ; 43(1): 1288-1297, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34503385

RESUMO

Long non-coding RNAs (LncRNAs) participate in the regulation of chronic kidney disease (CKD), and acute kidney injury (AKI) is identified as an important risk factor for CKD. This study investigated the involvement of a novel LncRNA MALAT1 in regulating lipopolysaccharide (LPS)-induced cell pyroptosis and inflammation in the human renal tubular epithelial HK-2 cells. Here, the HK-2 cells were subjected to LPS (2 µg/mL) treatment to establish cellular AKI models in vitro, and we validated that LPS triggered NLRP3-mediated pyroptotic cell death, promoted cell apoptosis and inflammation-associated cytokines secretion to induce HK-2 cell injury. Then, a novel LncRNA MALAT1/miRNA (miRNA)-135b-5p axis was verified to rescue cell viability in LPS treated HK-2 cells by targeting NLRP3. Mechanistically, miRNA-135b-5p bound to LncRNA MALAT1, and LncRNA MALAT1 positively regulated NLRP3 through acting as RNA sponger for miRNA-135b-5p. Further gain- and loss-of-function experiments evidenced that both LncRNA MALAT1 ablation and miRNA-135b-5p overexpression reversed LPS-induced cell pyroptosis, apoptosis, and inflammation in the HK-2 cells, and the protective effects of LncRNA MALAT1 knock-down on LPS-treated HK-2 cells were abrogated by silencing miRNA-135b-5p. In general, our study firstly investigated the role of the LncRNA MALAT1/ miRNA-135b-5p/NLRP3 signaling cascade in regulating LPS-induced inflammatory death in HK-2 cells.


Assuntos
Injúria Renal Aguda/metabolismo , MicroRNAs/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/efeitos dos fármacos , RNA Longo não Codificante/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Lipopolissacarídeos/toxicidade , MicroRNAs/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , RNA Longo não Codificante/genética , Transdução de Sinais
20.
Cent Eur J Immunol ; 46(3): 295-304, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34764801

RESUMO

INTRODUCTION: Resveratrol plays a protective role against sepsis development, and the long noncoding RNA (lncRNA) MALAT1 is an inflammation-relevant biomarker. This investigation attempted to reveal whether resveratrol attenuated inflammation of sepsis-induced acute kidney injury (AKI) by regulating MALAT1. MATERIAL AND METHODS: In total 120 rats were divided into a control group (n = 20), a Sham group (n = 20), a sepsis group (n = 40) and a resveratrol group (n = 40), and serum levels of inflammatory cytokines and AKI biomarkers were determined. An equal number of rats under identical treatments were, additionally, tracked for their survival, and the serum level of lncRNA MALAT1 was measured by RT-PCR. Moreover, septic cell models were constructed by treating HK-2 cells with lipopolysaccharide (LPS), and tumor necrosis factor α (TNF-α), interleukin (IL)-1ß, IL-6 levels released by the cells were determined with ELISA. RESULTS: Resveratrol treatment significantly brought down serum levels of inflammatory cytokines (i.e. TNF-α, IL-1ß and IL-6), kidney function indicators (i.e. Scr, blood urea nitrogen [BUN] and Scys C), AKI biomarkers (i.e. NGAL and KIM-1) and MALAT1 in cecal ligation and puncture (CLP)-induced septic model rats (all p < 0.05), and the life span of septic rats was elongated by resveratrol treatment (p < 0.05). Viability and cytokine release of LPS-treated HK2 cells were rescued by resveratrol (p < 0.05), which was accompanied by a marked fall of MALAT1 expression (p < 0.05). In addition, si-MALAT1 diminished viability and suppressed cytokine release of HK2 cells, while pcDNA3.1-MALAT1 hindered the impact of resveratrol on the inflammatory response of HK2 cells (p < 0.05). Ultimately, miR-205, a protective molecule in sepsis-relevant AKI, was down-regulated by resveratrol and si-MALAT1 (p < 0.05). CONCLUSIONS: Resveratrol relieved sepsis-induced AKI by restraining the lncRNA MALAT1/miR-205 axis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA