Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Pharmacol Res ; 182: 106332, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35779817

RESUMO

Currently, the reported source of extracellular vesicles (EVs) for the treatment of ischemic stroke(IS)is limited to mammals. Moreover, these EVs are restricted to clinical translation by the high cost of cell culture. In this respect, Lactobacillus plantarum culture is advantaged by low cost and high yield. However, it is poorly understood whether Lactobacillus plantarum-derived EVs (LEVs) are applicable for the treatment of IS. Here, our results demonstrated that LEVs reduced apoptosis in ischemic neuron both in vivo and in vitro. As revealed by high-throughput sequencing, miR-101a-3p expression was significantly elevated by LEV treatment in OGD/R-induced neurons, as confirmed in the tMCAO mice treated with LEVs. Mechanistically, c-Fos was directly targeted by miR-101a-3p. In addition, c-Fos determined ischemia-induced neuron apoptosis in vivo and in vitro through the TGF-ß1 pathway, miR-101a-3p inhibition aggravated ischemia-induced neuron apoptosis in vitro and in vivo, and miR-101a-3p overexpression produced the opposite results. Hsa-miR-101-3p was downregulated in the plasma of patients with IS but upregulated in the patients with neurological recovery after rt-PA intravenous thrombolysis. In conclusion, Our results demonstrated for the first time that LEVs might inhibit neuron apoptosis via the miR-101a-3p/c-Fos/TGF-ß axis, and has-miR-101-3p is a potential marker of neurological recovery in IS patients.


Assuntos
Lesões Encefálicas , Vesículas Extracelulares , Lactobacillus plantarum , MicroRNAs , Animais , Apoptose , Vesículas Extracelulares/metabolismo , Lactobacillus plantarum/genética , Lactobacillus plantarum/metabolismo , Mamíferos/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Fator de Crescimento Transformador beta
2.
Neurochem Res ; 46(5): 1119-1128, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33559830

RESUMO

This study aimed to explore the effects and function of microRNA-101a-3p (miR-101a-3p) in epilepsy. Rat model of pilocarpine-induced epilepsy was established and the seizure frequency was recorded. Expression of miR-101a-3p and c-Fos in hippocampus tissues of Rat models were detected by qRT-PCR and western blot. Besides, we established a hippocampal neuronal culture model of acquired epilepsy using Mg2+ free medium to evaluate the effects of miR-101a-3p and c-Fos in vitro. Cells were transfected with miR-101a-3p mimic, si-c-FOS, miR-101a-3p + c-FOS and its corresponding controls. MTT assay was used to detect cell viability upon transfection. Flow cytometry was performed to determine the apoptosis rate. Western blot was performed to measure the protein expression of apoptosis-related proteins (Bcl-2, Bax, and cleaved caspase 3), autophagy-related proteins (LC3 and Beclin1) and c-FOS. The targeting relationship between miR-101a-3p and c-FOS was predicted and verified by TargetScan software and dual-luciferase reporter assay. The role of miR-101a-3p was validated using epilepsy rat models in vivo. Another Rat models of pilocarpine-induced epilepsy with miR-NC or miR-101a-3p injection were established to evaluate the effect of miR-101a-3p overexpression on epilepsy in vivo. MiR-101a-3p was downregulated while c-FOS was increased in hippocampus tissues of Rat model of pilocarpine-induced epilepsy. Overexpression of miR-101a-3p or c-FOS depletion promoted cell viability, inhibited cell apoptosis and autophagy. C-FOS was a target of miR-101a-3p and miR-101a-3p negatively regulated c-FOS expression to function in epilepsy. Overexpression of miR-101a-3p attenuated pilocarpine-induced epilepsy in Rats in vivo. This study indicated that miR-101a-3p could attenuate pilocarpine-induced epilepsy by repressing c-Fos expression.


Assuntos
Epilepsia/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Apoptose/fisiologia , Autofagia/fisiologia , Sobrevivência Celular/fisiologia , Regulação para Baixo/fisiologia , Epilepsia/induzido quimicamente , Hipocampo/citologia , Hipocampo/metabolismo , Neurônios/metabolismo , Pilocarpina , Ratos Sprague-Dawley , Regulação para Cima/fisiologia
3.
J Oral Pathol Med ; 50(2): 236-243, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33187014

RESUMO

BACKGROUND: Temporomandibular joint osteoarthritis (TMJOA) is a degenerative disease that gradually affects the articular cartilage, synovium, and bone structure. To date, the molecular mechanism of TMJOA pathogenesis remains unclear. The aim of this study was to explore the biological function of the micro-ribonucleic acid 101a-3p (miR-101a-3p) and its role in TMJOA. METHODS: We detected the effect of interleukin-1ß (IL-1ß) on chondrocyte proliferation using Cell Counting Kit-8 (CCK-8) technology. Using quantitative polymerase chain reaction (qPCR), we detected transcription levels of miR-101a-3p in a rat model with TMJOA and inflamed chondrocytes, as well as in a group of normal rats. The effect of miR-101a-3p on apoptosis was examined in vitro using flow cytometry (FCM). We then analyzed the target of miR-101a-3p via bioinformatics and confirmed it using a luciferase reporter assay (LRA). RESULTS: We showed that IL-1ß could inhibit proliferation of chondrocytes. We found that miR-101a-3p levels were significantly lower in the rat inflammation model with TMJOA and inflamed chondrocytes than in the normal group. Additionally, miR-101a-3p substantially promoted apoptosis of chondrocytes, and both bioinformatic analyses and LRA found that this miRNA targeted the genes ubiquitin-conjugating enzyme 2D1 (UBE2D1) and Frizzled class receptor 4 (FZD4). CONCLUSION: Our results suggested that miR-101a-3p was involved in the pathogenesis of TMJOA and that its mechanism was probably interaction with its target genes UBE2D1 and FZD4.


Assuntos
Cartilagem Articular , MicroRNAs , Osteoartrite , Animais , Apoptose/genética , Condrócitos , Receptores Frizzled , MicroRNAs/genética , Osteoartrite/genética , Ratos , Articulação Temporomandibular
4.
J Cell Biochem ; 121(2): 1801-1810, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31633219

RESUMO

Valvulopathy is a familiar heart disease, which fearfully harms the health of the body. We studied the effects and mechanism of long noncoding RNA maternally expressed gene 3 (lncMEG3) on MVICs cell in inflammatory damage. Cell Counting Kit-8 and flow cytometry were respectively used to detect the effect of tumor necrosis factor α (TNF-α), MEG3 and microRNA (miR)-101a on cell viability and apoptosis. Moreover, MEG3 and miR-101a expression were changed by cell transfection and investigated by reverse transcription-quantitative polymerase chain reaction. Furthermore, Western blot was used to investigate the levels of Bax, pro-caspase-3, cleaved-caspase-3, pro-caspase-9, cleaved-caspase-9, interleukin (IL)-1ß, IL-6 and related-proteins of cell pathways. Otherwise, the levels of IL-1ß and IL-6 were also investigated by enzyme-linked immunosorbent assay kit. Reactive oxygen species (ROS) was examined by ROS assay. We found TNF-α caused inflammatory damage and upregulated MEG3. MEG3 was overexpressed and silenced in cells. Besides, MEG3 deteriorated inflammatory damage. Furthermore, MEG3 negatively regulated miR-101a and miR-101a mimic could reverse the effect of pc-MEG3. Besides, MEG3 enhanced the JNK and NF-κB pathways by downregulating miR-101a. In conclusion, MEG3 deteriorated cell inflammatory damage by downregulating miR-101a via JNK and NF-κB pathways.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/patologia , MicroRNAs/genética , Valva Mitral/patologia , RNA Longo não Codificante/genética , Fator de Necrose Tumoral alfa/farmacologia , Células Cultivadas , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Valva Mitral/metabolismo , Espécies Reativas de Oxigênio/metabolismo
5.
Mol Cell Biochem ; 470(1-2): 41-51, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32394311

RESUMO

Astragaloside IV (AS/IV) is one of the extracted components from the traditional Chinese medicine Astragalus which has been demonstrated to have potential capacity for anti-inflammation activity and for treating cardiovascular disease. Our purpose was to determine the function and underlying molecular mechanism of AS/IV in hypoxia/reoxygenation (H/R) injured in cardiomyocytes. Differentially expressed genes (DEGs) were screened using bioinformatic analysis, and the molecular targeting relationship was verified by the dual-luciferase report system. H/R injured cardiomyocytes were employed to explore the effect of AS/IV. QRT-PCR and Western blot analysis were applied to detect the expression of mRNA and proteins, respectively. Additionally, superoxide dismutase (SOD), lactic dehydrogenase (LDH) and MDA (malondialdehyde) levels were detected to determine the oxidative damage. Cell viability was assessed by CCK-8, and flow cytometry was used to evaluate cell apoptosis ratio. TGFBR1 and TLR2 were selected as DEGs. Additionally, AS/IV could enhance cell proliferation and upregulated miR-101a expression, which suppressed TGFBR1 and TLR2 expression in H/R injured cardiomyocytes. Moreover, the results of Western blot exhibited that the downstream genes (p-ERK and p-p38) in the MAPK signaling pathway were suppressed, which meant AS/IV could inhibit this pathway in H/R injured cardiomyocytes. Overall, this study demonstrated AS/IV could attenuate H/R injury in human cardiomyocytes via the miR-101a/TGFBR1/TLR2/MAPK signaling pathway axis, which means that it could serve as a possible alternate for H/R treatment.


Assuntos
Hipóxia Celular/efeitos dos fármacos , MicroRNAs/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Saponinas/farmacologia , Triterpenos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular , Biologia Computacional , Perfilação da Expressão Gênica , Humanos , Inflamação , L-Lactato Desidrogenase/metabolismo , Sistema de Sinalização das MAP Quinases , Malondialdeído/metabolismo , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Receptor 2 Toll-Like/metabolismo
6.
Clin Exp Pharmacol Physiol ; 46(3): 246-254, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30304543

RESUMO

Hypertensive nephropathy, clinically characterized by progressive renal fibrosis and inflammation, is a severe complication of hypertension. The objectives of this study were to investigate the roles of miR-101a in relieving angiotensin II (Ang II)-mediated hypertensive nephropathy and uncover the possible underlying mechanisms. A hypertensive mouse model was established via continuous 28-day AngII infusion. Systolic blood pressure (SBP), ratio of urine albumin to creatinine, blood urea nitrogen (BUN), serum creatinine (Scr) and glomerular filtration rate (GFR) were evaluated. Dual luciferase reporter assay was used to explore the target of miR-101a. mRNA levels of miR-101a, TGFßRI, fibrotic markers (Collagen I and α-SMA) and pro-inflammatory cytokines (IL-1ß and TNF-α) were determined by real-time PCR. Protein levels of TGFßRI, Collagen I, α-SMA, IL-1ß, TNF-α, t-p65, P-p65, t-Smad3, P-Smad3, t-IκBα and P-IκBα were detected by western blot. MiR-101a mimics significantly improved GFR and inhibited AngII-induced increase in the ratio of urine albumin to creatinine, BUN and Scr. MiR-101a mimics partially abolished AngII-induced increase in the mRNA and protein level of fibrotic markers by targeting TGFßRI and inhibiting TGFß/Smad3 pathway. Moreover, TGFßRI inhibitor galunisertib inhibited AngII-mediated renal injury in mice with hypertensive nephropathy. Additionally, miR-101a overexpression blocked AngII-induced up-regulation of pro-inflammatory markers via suppressing NF-κB pathway. MiR-101a exhibited protective effects against hypertensive nephropathy via inhibiting TGFß/Smad3 and NF-κB signalling pathways.


Assuntos
Angiotensina II/metabolismo , Hipertensão Renal/patologia , MicroRNAs/genética , NF-kappa B/metabolismo , Nefrite/patologia , Transdução de Sinais/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Modelos Animais de Doenças , Fibrose , Hipertensão Renal/genética , Hipertensão Renal/metabolismo , Hipertensão Renal/fisiopatologia , Rim/patologia , Rim/fisiopatologia , Masculino , Camundongos , Nefrite/genética , Nefrite/metabolismo , Nefrite/fisiopatologia
7.
Cell Mol Neurobiol ; 35(8): 1117-25, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25982511

RESUMO

The mechanism of sevoflurane preconditioning-induced neuroprotection is poorly understood. This study was aimed at identifying microRNAs (miRNAs) involved in the protective effect of sevoflurane preconditioning against hypoxic injury using the miRCURYTM LNA Array. The screened differentially expressed miRNAs were further validated using qRT-PCR. Finally, after transfection of miRNA (miR-101a or miR-34b) mimics or inhibitor, MTT and flow cytometry assays were used to evaluate cell survival and apoptosis in sevoflurane preconditioning. qRT-PCR confirmed the changes in expression of differentially expressed miRNAs that were screened by the microarray: down-regulation of rno-miR-101a, rno-miR-106b, and rno-miR-294 and up-regulation of rno-miR-883, rno-miR-16, and rno-miR-34b. MiR-101a and miR-34b were the most differentially expressed miRNAs. Sevoflurane preconditioning-inhibited apoptosis and preconditioning-enhanced cell viability of PC12 cells were significantly attenuated by transfection of miR-101a mimetic or miR-34b inhibitors, but were significantly enhanced by transfection of miR-34b mimetic. Therefore, a number of miRNAs, including miR-101a and miR-34b, might play important roles in the neuroprotection induced by sevoflurane preconditioning. Such miRNAs might provide novel targets for preventive and therapeutic strategies against cerebral ischemia-reperfusion injury.


Assuntos
Precondicionamento Isquêmico/métodos , Éteres Metílicos/farmacologia , MicroRNAs/biossíntese , Fármacos Neuroprotetores/farmacologia , Animais , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , MicroRNAs/antagonistas & inibidores , Células PC12 , Ratos , Sevoflurano
8.
Environ Pollut ; 343: 123153, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38103713

RESUMO

As a potential environmental obesogen, triclosan (TCS) carries inherent risks of inducing obesity and metabolic disorders. However, the underlying molecular mechanisms behind the lipid metabolism disorder induced by TCS have remained elusive. Through a fusion of transcriptomics and microRNA target prediction, we hypothesize that miR-101a as a responsive miRNA to TCS exposure in zebrafish, playing a central role in disturbing lipid homeostasis. As an evidence, TCS exposure triggers a reduction in miR-10a expression that accompanied by elevation of genes linked to regulation of lipid homeostasis. Through precision-controlled interventions involving miRNA expression modulation, we discovered that inhibition of miR-101a enhanced expression of its target genes implicated in lipid homeostasis, subsequently triggering excessive fat accumulation. Meanwhile, the overexpression of miR-101a acts as a protective mechanism, counteracting the lipid metabolism disorder induced by TCS in the larvae. Notably, the combination of short-chain fatty acids (SCFAs) emerged as a potential remedy to alleviate TCS-induced lipid accumulation partially by counteracting the decline in miR-101a expression induced by TCS. These revelations provide insight into a prospective molecular framework underlying TCS-triggered lipid metabolism disorders, thereby paving the way for pre-emptive strategies in combating the ramifications of TCS pollution.


Assuntos
Transtornos do Metabolismo dos Lipídeos , MicroRNAs , Triclosan , Animais , Triclosan/toxicidade , Triclosan/metabolismo , Peixe-Zebra/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Ácidos Graxos Voláteis/metabolismo
9.
Aging (Albany NY) ; 16(10): 8732-8746, 2024 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-38775730

RESUMO

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disease characterized by the loss of dopaminergic neurons in substantia nigra pars compacta (SNpc). This study focuses on deciphering the role of microRNA (miR)-101a-3p in the neuronal injury of PD and its regulatory mechanism. METHODS: We constructed a mouse model of PD by intraperitoneal injection of 1-methyl 4-phenyl 1, 2, 3, 6-tetrahydropyridine hydrochloride (MPTP), and used 1-methyl-4-phenylpyridinium (MPP+) to treat Neuro-2a cells to construct an in-vitro PD model. Neurological dysfunction in mice was evaluated by swimming test and traction test. qRT-PCR was utilized to examine miR-101a-3p expression and ROCK2 expression in mouse brain tissues and Neuro-2a cells. Western blot was conducted to detect the expression of α-synuclein protein and ROCK2 in mouse brain tissues and Neuro-2a cells. The targeting relationship between miR-101a-3p and ROCK2 was determined by dual-luciferase reporter gene assay. The apoptosis of neuro-2a cells was assessed by flow cytometry. RESULTS: Low miR-101a-3p expression and high ROCK2 expression were found in the brain tissues of PD mice and MPP+-treated Neuro-2a cells; PD mice showed decreased neurological disorders, and apoptosis of Neuro-2a cells was increased after MPP+ treatment, both of which were accompanied by increased accumulation of α-synuclein protein. After miR-101a-3p was overexpressed, the neurological function of PD mice was improved, and the apoptosis of Neuro-2a cells induced by MPP+ was alleviated, and the accumulation of α-synuclein protein was reduced; ROCK2 overexpression counteracted the protective effect of miR-101a-3p. Additionally, ROCK2 was identified as the direct target of miR-101a-3p. CONCLUSION: MiR-101a-3p can reduce neuronal apoptosis and neurological deficit in PD mice by inhibiting ROCK2 expression, suggesting that miR-101a-3p is a promising therapeutic target for PD.


Assuntos
Modelos Animais de Doenças , MicroRNAs , Quinases Associadas a rho , Animais , MicroRNAs/metabolismo , MicroRNAs/genética , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/genética , Camundongos , Masculino , Camundongos Endogâmicos C57BL , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Linhagem Celular Tumoral , Apoptose/genética , 1-Metil-4-fenilpiridínio/toxicidade
10.
Food Chem Toxicol ; 159: 112733, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34856318

RESUMO

Benzo[a]pyrene (BaP) can cause hepatorenal toxicity. Secoisolariciresinol diglucoside (SDG), a polyphenolic compound present in flaxseed, has shown a variety of biological activities including antioxidant, anti-inflammatory, anti-apoptotic effects. This study aimed to investigate the protective effects and working mechanisms of SDG against BaP-induced hepatorenal injury. Forty male mice were administrated daily (via gastric gavage; 4 weeks) with 0.9% saline (control), BaP (75 mg/kg body weight (b.w.)), SDG (100 mg/kg b.w.), SDG (100 mg/kg b.w.)+BaP (75 mg/kg b.w.). Results showed that the mice treated with SDG + BaP had significantly (P < 0.05) higher body weight, lower organ-to-body weight ratio, alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP) activities, and less levels of serum creatinine (CRE) and blood urea nitrogen (BUN) than those treated with BaP alone. SDG administration alleviated BaP-induced oxidative damages, inflammation and apoptosis. Furthermore, it significantly (P < 0.05) downregulated phosphor-p38 (p-p38) and phosphor-extracellular regulated protein kinases (p-ERK) levels, upregulated mitogen-activated protein kinase phosphatase-1 (MKP-1) level, and suppressed miR-101a expression compared with BaP alone group. Taken together, these results showed for the first time that SDG has protective effects against BaP-induced liver and kidney toxicity in mice through regulating oxidative stress, inflammation and apoptosis via miR-101a/MKP-1-mediated p38 and ERK pathway.


Assuntos
Injúria Renal Aguda/metabolismo , Benzo(a)pireno/toxicidade , Butileno Glicóis/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Glucosídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Fosfatase 1 de Especificidade Dupla/genética , Fosfatase 1 de Especificidade Dupla/metabolismo , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo
11.
Sci China Life Sci ; 64(10): 1612-1623, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33521860

RESUMO

Skeletal muscle differentiation is a highly coordinated process that involves many cellular signaling pathways and microRNAs (miRNAs). A group of muscle-specific miRNAs has been reported to promote myogenesis by suppressing key signaling pathways for cell growth. However, the functional role and regulatory mechanism of most non-muscle-specific miRNAs with stage-specific changes during differentiation are largely unclear. Here, we describe the functional characterization of miR-101a/b, a pair of non-muscle-specific miRNAs that show the largest change among a group of transiently upregulated miRNAs during myogenesis in C2C12 cells. The overexpression of miR-101a/b inhibits myoblast differentiation by suppressing the p38/MAPK, Interferon Gamma, and Wnt pathways and enhancing the C/EBP pathway. Mef2a, a key protein in the p38/MAPK pathway, was identified as a direct target of miR-101a/b. Interestingly, we found that the long non-coding RNA (lncRNA) Malat1, which promotes muscle differentiation, interacts with miR-101a/b, and this interaction competes with Mef2a mRNA to relieve the inhibition of the p38/MAPK pathway during myogenesis. These results uncovered a "braking" role in differentiation of transiently upregulated miRNAs and provided new insights into the competing endogenous RNA (ceRNA) regulatory mechanism in myoblast differentiation and myogenesis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Desenvolvimento Muscular/genética , Animais , Diferenciação Celular , Linhagem Celular , Sistema de Sinalização das MAP Quinases , Fatores de Transcrição MEF2/genética , Camundongos , Mioblastos/citologia , RNA Longo não Codificante/genética , Regulação para Cima
12.
J Biochem ; 168(4): 365-374, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32413114

RESUMO

This study aimed to investigate the mechanism of mangiferin on regulating endoplasmic reticulum (ER) stress in acute liver injury. The mouse model of acute liver injury was established by injection of LPS/D-GalN. The primary mouse hepatocytes were stimulated with LPS to induce the in vitro model. The effect of miR-20a/101a on the luciferase activity of Nrf2 3'-UTR was assessed by luciferase reporter assay. Mangiferin improved the liver function, inhibited the oxidative stress and ER stress and down-regulated the expressions of miR-20a and miR-101a in LPS/D-GalN-induced mice and LPS-induced hepatocytes. The knockdown of miR-20a and miR-101a co-operatively alleviated ER stress of LPS-induced hepatocytes. miR-20a and miR-101a both targeted Nrf2 and the over-expression of miR-20a or miR-101a decreased Nrf2 protein level, while their silences increased Nrf2 protein level. The silence of miR-20a and miR-101a promoted Nrf2 expression and inhibited the ER stress in LPS-induced hepatocytes, while the knockdown of Nrf2 reversed these effects. The over-expression of miR-20a and miR-101a eliminated the effects of mangiferin on Nrf2 protein level and ER stress in LPS-induced hepatocytes and Nrf2 over-expression altered these trends. Our findings suggest that mangiferin alleviates ER stress in acute liver injury by regulating the miR-20a/miR-101a-Nrf2 axis.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , MicroRNAs/genética , MicroRNAs/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Xantonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/patologia , Modelos Animais de Doenças , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
13.
Aging (Albany NY) ; 12(8): 7232-7247, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32315985

RESUMO

The purpose of this study was to reveal the hypothesis that lncRNA X inactive specific transcript (XIST) can participate in the regulation of cardiomyocyte apoptosis in neonatal mice cardiomyocytes (NMCMs) and myocardial infarction (MI) through targeting miR-101a-3p. NMCMs were isolated from neonatal C57BL/6 mice and anoxia was induced in hypoxic chamber. MTT assay and flow cytometry were used to determine proliferation and apoptosis respectively. The target relationship among XIST, miR-101a-3p and FOS was revealed by bioinformatic analysis, luciferase reporter assay, pull-down assay and RNA immunoprecipitation assay. The expression of XIST, miR-101a-3p, FOS and apoptosis-related proteins was determined by qRT-PCR or western blot. MI model was constructed to reveal the role of XIST. We found that XIST was up-regulated in NMCMs under anoxia condition. Moreover, XIST increased FOS expression by sponging miR-101a-3p in anoxia cells. Silencing XIST expression improved cell viability and suppressed apoptosis in vitro and inhibited myocardial infarction by reducing the level of c-FOS and apoptosis-related proteins in vivo. Our findings suggest that XIST is involved in MI, modulation of its level can be used as a new strategy or potential target in the treatment of myocardial infarction.


Assuntos
MicroRNAs/genética , Infarto do Miocárdio/genética , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , RNA Longo não Codificante/genética , Regulação para Cima , Animais , Animais Recém-Nascidos , Apoptose , Western Blotting , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Longo não Codificante/metabolismo , Transdução de Sinais
14.
Hum Exp Toxicol ; 39(12): 1628-1638, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32633566

RESUMO

Acute kidney injury (AKI) is an independent risk factor for chronic kidney disease (CKD). However, the role and mechanism of microRNA (miRNA, miR) in AKI-CKD transition are elusive. In this study, a murine model of renal ischemia/reperfusion was established to investigate the repairing effect and mechanism of miR-101a-3p on renal injury. The pathological damage of renal tissue was observed by hematoxylin and eosin and Masson staining. The levels of miR-101, profibrotic cytokines, and epithelial-mesenchymal transition (EMT) markers were analyzed using Western blotting, real-time polymerase chain reaction, and/or immunofluorescence. MiR-101 overexpression caused the downregulation of α-smooth muscle actin, collagen-1, and vimentin, as well as upregulation of E-cadherin, thereby alleviating the degree of renal tissue damage. MiR-101 overexpression mitigated hypoxic HK-2 cell damage. Collagen, type X, alpha 1 and transforming growth factor ß receptor 1 levels were downregulated in hypoxic cells transfected with miR-101 mimic. Our study indicates that miR-101 is an anti-EMT miRNA, which provides a novel therapeutic strategy for AKI-CKD transition.


Assuntos
Injúria Renal Aguda/genética , Transição Epitelial-Mesenquimal , MicroRNAs , Insuficiência Renal Crônica/genética , Injúria Renal Aguda/patologia , Animais , Linhagem Celular , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Colágeno Tipo XII/genética , Fibrose , Humanos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Insuficiência Renal Crônica/patologia , Regulação para Cima
15.
Mol Med Rep ; 21(1): 89-96, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31746349

RESUMO

Hypoxia/reoxygenation (H/R) is used as an in vivo model of ischemia/reperfusion injury, and myocardial ischemia can lead to heart disease. Therefore, it is necessary to prevent myocardial H/R injury to avoid the risk of heart disease. The aim of the present study was to investigate whether inhibiting microRNA (miR)­101a­3p attenuated H9C2 cell H/R injury, apoptosis mechanisms and key target proteins. Cell viability and apoptosis were determined by Cell Counting Kit­8 assays and flow cytometry using a cell apoptosis kit, respectively. The contents of creatine kinase (CK) and lactate dehydrogenase (LDH) were detected using colorimetric assays. Dual luciferase assays were carried out to determine if miR­101a­3p inhibited Janus kinase (JAK)2. Western blot analysis and reverse transcription­quantitative PCR were used to determine proteins levels and mRNAs expression. It was found that the inhibition of miR­101a­3p increased the growth of H9C2 cells and decreased H9C2 cell apoptosis during H/R injury. The inhibition of miR­101a­3p reduced the amounts of CK and LDH in H/R model H9C2 cells. The inhibition of miR­101a­3p lowered the levels of Bax, interleukin­6 and tumor necrosis factor­α, but raised the levels of phosphorylated (p)­STAT3 and p­JAK2 in H9C2 cells subjected to H/R injury treatment. miR­101a­3p mimic was found to inhibit H9C2 cell viability, raise p­JAK2 level and slightly increase p­STAT3 during H/R injury. AG490 induced H9C2 cell apoptosis, and decreased the levels of p­JAK2 and p­STAT3 during H/R injury. The data indicated that inhibiting miR­101a­3p reduced H/R damage in H9C2 cells and decreased apoptosis via Bax/Bcl­2 signaling during H/R injury. In addition, it was suggested that the inhibition of miR­101a­3p decreased H/R injury in H9C2 cell by regulating the JAK2/STAT3 signaling pathway.


Assuntos
Janus Quinase 2/metabolismo , MicroRNAs/metabolismo , Mioblastos Cardíacos/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Mioblastos Cardíacos/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Ratos
16.
Exp Ther Med ; 14(1): 289-297, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28672928

RESUMO

MicroRNAs (miRNAs or miRs) and the target genes before and after warm acupuncture at the genetic level were assessed, and the cytokines and neurotransmitters related to insomnia were studied. Male Sprague-Dawley rats were used to create PCPA insomnia rat models and randomly divided into the normal, model, warm acupuncture, and drug groups. The Dinghui Acupoint, Heyi Acupoint, and Xin Acupoint were inserted in the Mongolian medicine warm acupuncture group. The differential expression profile of microRNA in the brain tissue of the insomnia rats was determined before and after Mongolian medicine warm acupuncture for establishment of miR-101a mimics and inhibitor. qPCR was used to detect the expression level of miR-101a. Western blotting was used to detect the expression level of PAX8. The rats receiving Mongolian medicine warm acupuncture had 141 miRNAs with differential expression compared with the normal rats. The expression level of miR-101a in the cells of the hippocampus of the insomnia rats transfected with miR-101a mimics increased significantly at 72 h (P<0.05). The activity of the neuronal cells transfected with miR-101a inhibitor increased significantly at 72 h (P<0.05). The western blotting result indicated that the expression of the PAX8 protein in the neuronal cells of the insomnia model rats was inhibited and downregulated significantly at 72 h after addition of miR-101a mimics compared with that in the scramble added group (P<0.01). The levels of the interleukins IL-1, IL-2, and IL-6 and the tumor necrosis factor-α in the hypothalamus, hippocampus, and prefrontal cortex decreased significantly compared with those in the blank control group (P<0.05). The levels of noradrenaline, dopamine, and glutamic decreased significantly following warm acupuncture or western medicine treatment (P<0.05). In conclusion, this study demonstrates that the upregulation of miR-101a in the rats treated with warm acupuncture is directly associated with PAX8 regulation.

17.
Gene ; 572(2): 198-204, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26160440

RESUMO

MicroRNAs are a class of small, non-coding RNAs that participate in the regulation of diverse biological processes including skeletal muscle development. However, the underlying molecular mechanisms are not fully understood, particularly in goat. Here, we identified goat miR-101a as a novel myogenic microRNA mediating myogenic differentiation. The expression of miR-101a was enriched in goat skeletal muscles and up-regulated during satellite cell differentiation. After transfection with a miR-101a mimic and culturing in differentiation medium, satellite cell differentiation was promoted, accompanied by a significant increase in mRNA expression of the myogenic marker, MyoG, and decreased expression of MyoD. In contrast, blocking the function of miR-101a with a 2'-O-methylated antisense oligonucleotide inhibitor repressed satellite cell differentiation. However, both gain- and loss-of-function studies demonstrated that miR-101a had no significant influence on satellite cell proliferation. Therefore, our results provide a new insight on miR-101a in the regulation of goat skeletal muscle satellite cell proliferation and differentiation.


Assuntos
Cabras/genética , MicroRNAs/genética , Músculo Esquelético/fisiologia , Proteína MyoD/genética , Miogenina/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Feminino , Oligonucleotídeos Antissenso/farmacologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa