Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Stem Cells ; 42(5): 460-474, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38381592

RESUMO

Cell therapy based on mesenchymal stem cells (MSCs) alleviate muscle atrophy caused by diabetes and aging; however, the impact of human umbilical cord mesenchymal stem cells on muscle atrophy following nerve injury and the underlying mechanisms remain unclear. In this study, we evaluated the therapeutic efficacy of human umbilical cord MSCs (hucMSCs) and hucMSC-derived exosomes (hucMSC-EXOs) for muscle atrophy following nerve injury and identified the underlying molecular mechanisms. Sciatic nerve crush injury in rats and the induction of myotubes in L6 cells were used to determine the ameliorating effect of hucMSCs and hucMSC-EXOs on muscle atrophy. Q-PCR and Western blot analyses were used to measure the expression of muscle-specific ubiquitin ligases Fbxo32 (Atrogin1, MAFbx) and Trim63 (MuRF-1). Dual-luciferase reporter gene experiments were conducted to validate the direct binding of miRNAs to their target genes. Local injection of hucMSCs and hucMSC-EXOs mitigated atrophy in the rat gastrocnemius muscle following sciatic nerve crush injury. In vitro, hucMSC-EXOs alleviated atrophy in L6 myotubes. Mechanistic analysis indicated the upregulation of miR-23b-3p levels in L6 myotubes following hucMSC-EXOs treatment. MiR-23b-3p significantly inhibited the expression of its target genes, Fbxo32 and Trim63, and suppressed myotube atrophy. Notably, an miR-23b-3p inhibitor reversed the inhibitory effect of miR-23b-3p on myotube atrophy in vitro. These results suggest that hucMSCs and their exosomes alleviate muscle atrophy following nerve injury. MiR-23b-3p in exosomes secreted by hucMSCs contributes to this mechanism by inhibiting the muscle-specific ubiquitination ligases Fbxo32 and Trim63.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Atrofia Muscular , Traumatismos dos Nervos Periféricos , Ubiquitina-Proteína Ligases , Exossomos/metabolismo , Animais , Atrofia Muscular/patologia , Atrofia Muscular/metabolismo , Atrofia Muscular/terapia , Atrofia Muscular/genética , Humanos , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Células-Tronco Mesenquimais/metabolismo , Ratos , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/patologia , Traumatismos dos Nervos Periféricos/terapia , Ratos Sprague-Dawley , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Cordão Umbilical/citologia , Músculo Esquelético/patologia , Músculo Esquelético/metabolismo , Masculino , Proteínas com Motivo Tripartido/metabolismo , Proteínas com Motivo Tripartido/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia
2.
Biochemistry (Mosc) ; 89(1): 97-115, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38467548

RESUMO

The overall survival of patients with the advanced and recurrent gastric cancer (GC) remains unfavorable. In particular, this is due to cancer spreading and resistance to chemotherapy associated with the epithelial-mesenchymal transition (EMT) of tumor cells. EMT can be identified by the transcriptome profiling of GC for EMT markers. Indeed, analysis of the TCGA and GTEx databases (n = 408) and a cohort of GC patients (n = 43) revealed that expression of the CDH2 gene was significantly decreased in the tumors vs. non-tumor tissues and correlated with the overall survival of GC patients. Expression of the EMT-promoting transcription factors SNAIL and ZEB1 was significantly increased in GC. These data suggest that targeting the EMT might be an attractive therapeutic approach for patients with GC. Previously, we demonstrated a potent anti-cancer activity of the olive leaf extract (OLE). However, its effect on the EMT regulation in GC remained unknown. Here, we showed that OLE efficiently potentiated the inhibitory effect of the chemotherapeutic agents 5-fluorouracil (5-FU) and cisplatin (Cis) on the EMT and their pro-apoptotic activity, as was demonstrated by changes in the expression of the EMT markers (E- and N-cadherins, vimentin, claudin-1) in GC cells treated with the aforementioned chemotherapeutic agents in the presence of OLE. Thus, culturing GC cells with 5-FU + OLE or Cis + OLE attenuated the invasive properties of cancer cells. Importantly, upregulation of expression of the apoptotic markers (PARP cleaved form) and increase in the number of cells undergoing apoptosis (annexin V-positive) were observed for GC cells treated with a combination of OLE and 5-FU or Cis. Collectively, our data illustrate that OLE efficiently interferes with the EMT in GC cells and potentiates the pro-apoptotic activity of certain chemotherapeutic agents used for GC therapy.


Assuntos
Olea , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Olea/metabolismo , Transição Epitelial-Mesenquimal , Fluoruracila/farmacologia , Cisplatino/farmacologia , Linhagem Celular Tumoral , Extratos Vegetais/farmacologia , Caderinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Movimento Celular
3.
Anim Biotechnol ; 35(1): 2346808, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38739483

RESUMO

This study aimed to evaluate the effect of miR-23b-3p on growth hormone (GH) in pituitary cells of Yanbian yellow cattle. The mRNA and protein levels of GH and miR-23b-3p target genes were measured by real time fluorescence quantitative PCR (qPCR) and Western blot, respectively. The target relationship of miR-23b-3p was validated by double luciferase reporter gene system. The results showed that GH mRNA and protein levels in pituitary cells of Yanbian yellow cattle were significantly lower in the miR-23b-3p-mi group than in the NC group (P<0.01), while GH mRNA and protein levels were higher in the miR-23b-3p-in group than in the iNC group (P<0.05). The result of bioinformatics analysis and double luciferase reporter gene system validation proved that miR-23b-3p targeted 3'UTR of pituitary specific transcription factor 1 (POU1F1). POU1F1 mRNA and protein levels were lower miR-23b-3p-mi group than in the NC group (P<0.01), while POU1F1 mRNA and protein levels were higher in the miR-23b-3p-in group than in the iNC group (P<0.01). These results demonstrated that miR-23b-3p could regulate GH expression in pituitary cells by regulating POU1F1 gene.


Assuntos
Hormônio do Crescimento , MicroRNAs , Fator de Transcrição Pit-1 , Animais , Bovinos/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo , Hipófise/metabolismo , Regulação da Expressão Gênica , Regiões 3' não Traduzidas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Int J Med Sci ; 20(7): 958-968, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37324192

RESUMO

The treatment of bone loss due to periodontitis has posed a great challenge for physicians for decades. Therefore, it is of extraordinary significance to identify an effective regeneration scheme for alveolar bone. This study aimed to investigate long non-coding RNA (lncRNA) small nucleolar RNA host gene 5 (SNHG5) whether sponges microRNA-23b-3p (miR-23b-3p) to achieve the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs). Results revealed that the expression of SNHG5 was upregulated whereas that of miR-23b-3p was downregulated in osteogenic hPDLSCs. Alizarin red staining assays and qRT-PCR demonstrated that SNHG5 silencing or miR-23b-3p overexpression inhibits hPDLSCs osteogenic differentiation and vice versa. In addition, miR-23b-3p partially abolished the promotive effect of SNHG5 on osteogenic differentiation of hPDLSCs. Dual luciferase report and RNA pulldown assay verified that miR-23b-3p is a regulatory target of SNHG5 and that Runx2 is a gene target of miR-23b-3p. In brief, the results demonstrate that SNHG5 promotes the osteogenic differentiation of hPDLSCs by regulating the miR-23b-3p/Runx2 axis. Our study provides novel mechanistic insights into the critical role of lncRNA SNHG5 as a miR-23b-3p sponge to regulate Runx2 expression in hPDLSCs and may serve as a potential therapeutics target for periodontitis.


Assuntos
MicroRNAs , Periodontite , RNA Longo não Codificante , Humanos , Diferenciação Celular/genética , Células Cultivadas , MicroRNAs/genética , MicroRNAs/metabolismo , Osteogênese/genética , Ligamento Periodontal , Periodontite/genética , Periodontite/metabolismo , RNA Longo não Codificante/metabolismo , Células-Tronco
5.
Exp Eye Res ; 222: 109179, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35810770

RESUMO

Age-related cataract (ARC) is a common eye disease that occurs mostly in the elderly. Emerging evidence suggests that circular RNA (circRNA) plays an important role in disease development. However, there are few reports about the role of circRNA in cataract. Here, we investigated the function of circ_0060,144 in ARC. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to measure the expression of circ_0060,144, miR-23b-3p, and homeodomain interacting protein kinase 3 (HIPK3) mRNA. CCK-8 and flow cytometry analysis of cell proliferation and apoptosis. Western blot was performed to measure protein-associated proliferation and apoptosis. ELISA was used to detect cellular MDA and GSH-Px levels. Dual-luciferase reporter, RNA immunoprecipitation (RIP), and RNA pull-down assays were used to investigate the association between miR-23b-3p and circ_0060,144 or HIPK3. Circ_0060,144 and HIPK3 mRNA expression were decreased in ARC tissues, and miR-23b-3p was increased. Circ_0060,144 overexpression promoted proliferation and inhibited apoptosis of SRA01/04 cells. And proliferation-related and apoptosis-related proteins also confirmed this conclusion. In addition, circ_0060,144 overexpression reduced MDA level and increased GSH-Px level. In terms of mechanism, circ_0060,144 inhibited HIPK3 expression via sponging miR-23b-3p. Circ_0060,144 promoted ARC development via regulation of miR-23b-3p/HIPK3 axis.


Assuntos
Catarata , Peptídeos e Proteínas de Sinalização Intracelular , MicroRNAs , Proteínas Serina-Treonina Quinases , RNA Circular , Idoso , Apoptose , Catarata/genética , Proliferação de Células , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , RNA Circular/genética , RNA Mensageiro
6.
Clin Exp Nephrol ; 26(5): 385-397, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34997887

RESUMO

BACKGROUND: Diabetic nephropathy (DN), a diabetic complication, is the leading cause of end-stage renal disease. KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1), a long non-coding RNA, has been unmasked to participate in the pathogenesis of DN. However, the specific mechanism by which KCNQ1OT1 regulates podocyte injury remains unclear. METHODS: Relative expression of KCNQ1OT1 was measured with quantitative real-time polymerase chain reaction (qRT-PCR). The levels of inflammatory cytokines were analyzed by enzyme linked immunosorbent assay (ELISA). The viability, proliferation, and apoptosis of high glucose (HG)-treated podocyte were analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. Protein levels were analyzed by western blotting. The regulatory mechanism of KCNQ1OT1 was surveyed by bioinformatics analysis, dual-luciferase reporter, and RNA immunoprecipitation (RIP) assays. RESULTS: We observed an apparent upregulation in KCNQ1OT1 expression in serums of DN patients and HG-treated podocytes. Furthermore, KCNQ1OT1 downregulation alleviated HG-induced inflammation, proliferation repression, and apoptosis in podocytes. Notably, KCNQ1OT1 was identified as a miR-23b-3p sponge, and miR-23b-3p directly targeted Semaphorin-3A (Sema3A). Moreover, miR-23b-3p silencing reversed KCNQ1OT1 knockdown-mediated effects on inflammation, proliferation, and apoptosis of HG-induced podocytes. Also, Sema3A overexpression reversed the effects of miR-23b-3p mimic on inflammation, proliferation, and apoptosis of HG-induced podocytes. Importantly, KCNQ1OT1 regulated Sema3A expression by sponging miR-23b-3p. CONCLUSIONS: HG-induced KCNQ1OT1 promoted inflammation, proliferation repression, and apoptosis of podocytes via increasing Sema3A expression through sponging miR-23b-3p. This study provided evidence to support the involvement of KCNQ1OT1 in the pathogenesis of DN.


Assuntos
Nefropatias Diabéticas , MicroRNAs , Podócitos , Apoptose , Nefropatias Diabéticas/patologia , Feminino , Glucose/metabolismo , Glucose/toxicidade , Humanos , Inflamação , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Podócitos/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia
7.
Ren Fail ; 44(1): 1961-1975, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36350669

RESUMO

BACKGROUND: Given the reported effects of nuclear paraspeckle assembly transcript 1 (NEAT1) on kidney injury, a study is worth formulating to investigate whether and how NEAT1 impacts podocytes. MATERIALS AND METHODS: A mouse podocyte injury model was established using the adriamycin (ADR)-induced mouse podocyte cell line (MPC5). The target relationships between NEAT1 and microRNA (miR)-23b-3p and between miR-23b-3p and Bcl-2 interacting protein 3 like (BNIP3L) were verified by dual-luciferase reporter assay and RNA immunoprecipitation assay. After ADR-induced MPC5 cells were transfected with NEAT1 overexpression plasmid (oe-NEAT1) or shNEAT1, the viability and apoptosis of MPC5 cells were evaluated by Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. The expressions of MPC5, miR-23b-3p, BNIP3L and the factors related to podocyte injury, apoptosis and epithelial-mesenchymal transition were determined using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. RESULTS: NEAT1 was high-expressed in ADR-induced cell model. After transfection with oe-NEAT1, the expression of NEAT1, the levels of marker (Desmin) and apoptosis were promoted, while the viability and the levels of podocyte injury markers (WT1, Nephrin) were inhibited in ADR-induced cells. However, shNEAT1 generated the effects opposite to oe-NEAT1. Besides, miR-23b-3p competitively bound to NEAT1 and targeted BNIP3L. MiR-23b-3p inhibitor reversed the effect of shNEAT1, while its effect could be further offset by shBNIP3L. Furthermore, miR-23b-3p inhibitor affected mouse podocyte injury through downregulating Bcl-2 and E-cadherin levels and upregulating Cleaved-caspase-3, Bax, N-cadherin, Vimentin and Snail levels, but shBNIP3L did oppositely. CONCLUSION: NEAT1 promotes the podocyte injury via targeting miR-23b-3p/BNIP3L axis.


Assuntos
MicroRNAs , Podócitos , RNA Longo não Codificante , Animais , Camundongos , Apoptose/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Paraspeckles , Podócitos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
8.
Int J Mol Sci ; 23(21)2022 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-36362337

RESUMO

In cervical cancer (CC), miR-23b-3p, miR-124-3p, and miR-218-5p have been found to act as tumor suppressors by regulating cellular processes related to progression and metastasis. The objective of the present review is to provide an update on the experimental evidence about the role of miR-23b-3p, miR-124-3p, and miR-218-5p in the regulation of CC progression. Additionally, we present the results of a bioinformatic analysis that suggest that these miRNAs have a somewhat redundant role in the same cellular processes that may result in a synergistic effect to promote CC progression. The results indicate that specific and common target genes for miR-23b-3p, miR-124-3p, and miR-218-5p regulate proliferation, migration, apoptosis, and angiogenesis, all processes that are related to CC maintenance and progression. Furthermore, several target genes may regulate cancer-related signaling pathways. We found that a total of 271 proteins encoded by the target mRNAs of miR-23b-3p, miR-124-3p, or miR-218-5p interact to regulate the cellular processes previously mentioned, and some of these proteins are regulated by HPV-16 E7. Taken together, information analysis indicates that miR-23b-3p, miR-124-3p, and miR-218-5p may potentiate their effects to modulate the cellular processes related to the progression and maintenance of CC with and without HPV-16 involvement.


Assuntos
MicroRNAs , Neoplasias do Colo do Útero , Feminino , Humanos , Neoplasias do Colo do Útero/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Transdução de Sinais , Apoptose/genética , Atenção , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica
9.
J Biol Chem ; 295(41): 14140-14152, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32690611

RESUMO

Transcriptional repressor zinc finger and BTB domain containing 1 (ZBTB1) is required for DNA repair. Because DNA repair defects often underlie genome instability and tumorigenesis, we determined to study the role of ZBTB1 in cancer. In this study, we found that ZBTB1 is down-regulated in breast cancer and this down-regulation is associated with poor outcome of breast cancer patients. ZBTB1 suppresses breast cancer cell proliferation and tumor growth. The majority of breast cancers are estrogen receptor (ER) positive and selective estrogen receptor modulators such as tamoxifen have been widely used in the treatment of these patients. Unfortunately, many patients develop resistance to endocrine therapy. Tamoxifen-resistant cancer cells often exhibit higher HER2 expression and an increase of glycolysis. Our data revealed that ZBTB1 plays a critical role in tamoxifen resistance in vitro and in vivo To see if ZBTB1 regulates HER2 expression, we tested the recruitments of ZBTB1 on HER2 regulatory sequences. We observed that over-expressed ZBTB1 occupies the estrogen receptor α (ERα)-binding site of the HER2 intron in tamoxifen-resistant cells, suppressing tamoxifen-induced transcription. In an effort to identify potential microRNAs (miRNAs) regulating ZBTB1, we found that miR-23b-3p directly targets ZBTB1. MiR-23b-3p regulates HER2 expression and tamoxifen resistance via targeting ZBTB1. Finally, we found that miR-23b-3p/ZBTB1 regulates aerobic glycolysis in tamoxifen-resistant cells. Together, our data demonstrate that ZBTB1 is a tumor suppressor in breast cancer cells and that targeting the miR-23b-3p/ZBTB1 may serve as a potential therapeutic approach for the treatment of tamoxifen resistant breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Receptor ErbB-2/biossíntese , Proteínas Repressoras/metabolismo , Tamoxifeno/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Glicólise/genética , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Receptor ErbB-2/genética , Proteínas Repressoras/genética , Proteínas Supressoras de Tumor/genética
10.
J Pharmacol Sci ; 145(1): 69-78, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33357782

RESUMO

Postmenopausal osteoporosis (PMOP) is one of the most common metabolic bone diseases in postmenopausal women. Increasing evidence has indicated that microRNAs (miRNAs) play vital regulatory roles during osteoporosis progression. This study aimed to investigate the potential function of miR-23b-3p in the osteogenic differentiation of human bone marrow mesenchymal stem cells (hMSCs). PMOP was induced in mice by bilateral ovariectomy. X-ray absorptiometry was applied to detect BMD and BMC in PMOP mice. Luciferase reporter assay and RIP assay were utilized to investigate the relationship between miR-23b-3p and MRC2. We found the upregulation of miR-23b-3p in bone tissues of PMOP mice. Silencing of miR-23b-3p relieved PMOP in mice. Moreover, miR-23b-3p knockdown facilitated the osteogenic differentiation of hMSCs by increasing the expression of Runx2, OCN, Osterix and promoting ALP activity. Mechanistically, MRC2 is a downstream target gene of miR-23b-3p. MRC2 knockdown significantly rescued the promoting effect of lenti-miR-23b-3p inhibitor on osteogenic differentiation of hMSCs. Furthermore, miR-23b-3p targeted MRC2 to inhibit the Wnt/ß-catenin pathway during the osteogenic differentiation of hMSCs. In summary, inhibition of miR-23b-3p alleviates PMOP by targeting MRC2 to inhibit the Wnt/ß-catenin signaling, which may provide a novel molecular insight for osteoporosis therapy.


Assuntos
Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/fisiologia , MicroRNAs/genética , MicroRNAs/fisiologia , Osteoporose Pós-Menopausa/genética , Osteoporose Pós-Menopausa/terapia , Receptores de Superfície Celular/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Marcação de Genes , Humanos , Camundongos , Terapia de Alvo Molecular , Osteogênese/genética , Ovariectomia
11.
BMC Endocr Disord ; 21(1): 57, 2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33781239

RESUMO

BACKGROUND: The role of miR-23b-3p in insulin resistance (IR) remained poorly understood. METHODS: After acacetin injection, obesity-induced IR model was constructed with or without miR-23b-3p upregulation and Neuraminidase 1 (NEU1) overexpression in mice. Body weight, serum metabolite and fat percent of the mice were measured. Tests on oral glucose and insulin tolerance were performed, and inflammatory cytokines C-reactive protein (CRP), Interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein 1 (MCP1) levels were quantified with enzyme-linked immunosorbent assay (ELISA). The binding sites between miR-23b-3p and NEU1 were predicted by TargetScan, and verified using dual-luciferase reporter assay. Relative expressions were detected with quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. Proportion of Treg and Th17 cells in total CD4+ T cells was detected with flow cytometry. RESULTS: MiR-23b-3p offset the effects of acacetin on body weight, fat percent, inflammatory cytokines levels and expressions of markers of regulatory T cells (Treg cells) and T helper 17 cells (Th17 cells), NEU1 and miR-23b-3p. NEU1 was a target of miR-23b-3p, and overexpressed NEU1 reversed the effects of upregulated miR-23b-3p on reducing Treg cells but increased body weight, fat percent and inflammatory cytokines levels, percentage of Th17 cells, and upregulated NEU1 expression. CONCLUSION: Upregulation of miR-23b-3p offset the effects of acacetin on obesity-induced IR through regulating Treg/Th17 cell balance via targeting NEU1.The present findings provide a possible prevention strategy for obesity-induced IR.


Assuntos
Resistência à Insulina , MicroRNAs/metabolismo , Neuraminidase/metabolismo , Obesidade/metabolismo , Células Th17 , Animais , Flavonas , Masculino , Camundongos Endogâmicos C57BL , Obesidade/imunologia
12.
Endocr J ; 68(5): 519-529, 2021 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-33408314

RESUMO

Long noncoding RNAs (lncRNAs) have been reported to play critical role in the development of diabetic nephropathy (DN). However, the effects and mechanism of plasmacytoma variant translocation 1 (PVT1) remain poorly understood. The expression of PVT1, miR-23b-3p, early growth response factor 1 (EGR1), Fibronectin (FN), Collagen IV (Col IV), alpha smooth muscle actin (α-SMA), E-cadherin, and vimentin, transforming growth factor (TGF)-ß1 was examined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was assessed by Cell Counting-8 (CCK-8) assay. Western blot assay was conducted to measure the protein levels of FN, Col IV, E-cadherin, α-SMA, vimentin, TGF-ß1, and EGR1. The interaction between miR-23b-3p and PVT1 or EGR1 was predicted by starBase or TargetScan and confirmed by the dual luciferase reporter assay. The oxidative stress factors were analyzed by corresponding kits. We found that the expression of PVT1 and EGR1 was increased and miR-23b-3p was decreased in serum samples of DN patients and HG-induced HRMCs. Knockdown of PVT1 significantly inhibited HG-induced proliferation, extracellular matrix (ECM) accumulation, epithelial-mesenchymal transition (EMT), and oxidative stress in HRMCs, while these effects were abated by inhibiting miR-23b-3p. In addition, EGR1 was confirmed as downstream target of miR-23b-3p and miR-23b-3p could specially bind to PVT1. Besides, downregulation of PVT1 inhibited the progression of DN partially via upregulating miR-23b-3p and downregulating EGR1. In conclusion, our results suggested that PVT1 knockdown suppressed DN progression though functioning as ceRNA of miR-23b-3p to regulate EGR1 expression in vitro, providing potential value for the treatment of DN.


Assuntos
Proliferação de Células/efeitos dos fármacos , Nefropatias Diabéticas/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Glucose/farmacologia , Células Mesangiais/efeitos dos fármacos , RNA Longo não Codificante/metabolismo , Linhagem Celular , Nefropatias Diabéticas/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Células Mesangiais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Longo não Codificante/genética
13.
Environ Toxicol ; 36(8): 1576-1590, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33913242

RESUMO

The purpose of this article was to probe the mechanism underlying long noncoding RNA (lncRNA)-LINC00184 in cholangiocarcinoma development and to investigate the effects of LINC00184 on cholangiocarcinoma. We used bioinformatics to analyze the expression of LINC00184, microRNA (miR)-23b-3p and ANXA2 in cholangiocarcinoma tissues. The levels of LINC00184, miR-23b-3p, and ANXA2 were detected by qRT-PCR. Cell proliferation was tested by CCK8. Transwell assay was used to detect cell invasion and migration. The target connection between LINC00184, miR-23b-3p, or ANXA2 was probed by luciferase reporter assay. RNA pull-down method was employed to test the relationship among LINC00184/miR-23b-3p/ANXA2 in cholangiocarcinoma cells. The Pearson correlation coefficient analyzed was applied to analyze the correlation among LINC00184, miR-23b-3p, and ANXA2. LC-MS/M analysis was used to explore whether the changes of adenine metabolism was affected by LINC00184 in cholangiocarcinoma cells. We discovered that LINC00184 expression was heightened in cholangiocarcinoma patients and cells. Knockdown of LINC00184 repressed cell proliferation, invasion, migration and adenine metabolism in cholangiocarcinoma cells. miR-23b-3p was regarded as a target of LINC00184 and its depletion perversed the inhibitive influence of LINC00184 silencing on cholangiocarcinoma cells. ANXA2 was a target of miR-23b-3p and was negatively modulated by miR-23b-3p. Moreover, ANXA2 was positively modulated by LINC00184 via sponging miR-23b-3p. In short, silencing of LINC00184 suppressed cell proliferation, invasion and migration through over-expression of miR-23b-3p and reducing of ANXA2 in cholangiocarcinoma cells. These findings contribute to understanding the influences of LINC00184, miR-23b-3p, and ANXA2 on cholangiocarcinoma and provide basis for cholangiocarcinoma treatment.


Assuntos
Anexina A2 , Neoplasias dos Ductos Biliares , Colangiocarcinoma , MicroRNAs , RNA Longo não Codificante , Adenina , Ductos Biliares Intra-Hepáticos , Movimento Celular , Proliferação de Células , Humanos
14.
Environ Toxicol ; 36(11): 2322-2332, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34418280

RESUMO

Long noncoding RNAs (lncRNAs) are known to be the important regulators in cancer progression. However, the role of lncRNA FAM66C (FAM66C) is yet to be investigated in intrahepatic cholangiocarcinoma (ICC). This study aimed to investigate the effects and related mechanisms of FAM66C in ICC. Human ICC tissues and cell lines were collected. The expression levels of FAM66C, hsa-miR-23b-3p (miR-23b-3p), and KCND2 were detected by qRT-RCR. The transfection experiments were employed to measure the effect of FAM66C on cell viabilities, migration, and invasion in ICC cells by CCK-8, transwell assays. Glycolysis was investigated by glucose consumption, lactate production and ATP levels. The dual-luciferase reporter and RNA pull down assays were conducted as a means of confirming the interactions between FAM66C, miR-23b-3p, and KCND2. Furthermore, the levels of the EMT-associated proteins (KCND2, GLUT1, PKM2, and LDHA) in ICC cells were detected by western blot. FAM66C was increased in ICC tissues and cells, increased cell viability, glycolysis, migration and invasion, and decreased apoptosis were shown in FAM66C overexpressing cells. Mechanistic analyses revealed that FAM66C regulated the downstream target gene KCND2 by sponging miR-23b-3p. FAM66C effect on ICC was further validated in murine xenograft assays. FAM66C knockdown cells gave rise to tumors that were smaller in size, consistent with the role of FAM66C as a promoter of in vivo tumor growth. These data revealed that FAM66C was able to drive ICC tumor progression and glycolytic activity via the miR-23b-3p/KCND2 axis, indicating FAM66C may be a viable target for treating ICC.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , MicroRNAs , RNA Longo não Codificante , Animais , Neoplasias dos Ductos Biliares/genética , Ductos Biliares Intra-Hepáticos/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Colangiocarcinoma/genética , Regulação Neoplásica da Expressão Gênica , Glicólise/genética , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Canais de Potássio Shal
15.
J Cell Mol Med ; 24(15): 8368-8378, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32558131

RESUMO

Ischemia/reperfusion (I/R)-mediated acute myocardial infarction (AMI) is a major pathological factor implicated in the progression of ischemic heart disease (IHD). Long non-coding RNA plays an important role in regulating the occurrence and development of cardiovascular disease. The aim of this study was to investigate the regulating role of LINC00261 in hypoxia/reoxygenation (H/R)-induced cardiomyocyte apoptosis. The relative expression of LINC00261, miR-23b-3p and NRF2 were determined in rats I/R myocardial tissues and H/R-induced cardiomyocytes. The rat model and cell model of LINC00261 overexpression were established to investigate the biological function of LINC00261 on H9C2 cell. The interaction between LINC00261, miR-23b-3p, NRF2 and FOXO3a was identified using bioinformatics analysis, luciferase reporter assay, RNA immunoprecipitation (RIP) assay, chromatin immunoprecipitation (CHIP) assay and qRT-PCR. The expression of LINC00261 was significantly down-regulated in myocardial tissues and H9C2 cell. Overexpression of LINC00261 improves cardiac function and reduces myocardium apoptosis. Interestingly, transcription factor FOXO3a was found to promote LINC00261 transcription. Moreover, LINC00261 was confirmed as a spong of miR23b-3p and thereby positively regulates NRF2 expression in cardiomyocytes. Our findings reveal a novel role for LINC00261 in regulating H/R cardiomyocyte apoptosis and the potency of the LINC00261/miR-23b-3p/NRF2 axis as a therapeutic target for the treatment of MIRI.


Assuntos
Proteína Forkhead Box O3/genética , Hipóxia/genética , MicroRNAs/genética , Miócitos Cardíacos/patologia , Fator 2 Relacionado a NF-E2/genética , RNA Longo não Codificante/genética , Animais , Apoptose/genética , Linhagem Celular , Regulação para Baixo/genética , Hipóxia/patologia , Infarto do Miocárdio/genética , Isquemia Miocárdica/genética , Miocárdio/patologia , Ratos , Transcrição Gênica/genética
16.
J Gene Med ; 22(11): e3247, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32602607

RESUMO

BACKGROUND: Long non-coding RNA (lncRNA) small nucleolar RNA host gene 17 (SNHG17) is a carcinogenic lncRNA in diverse cancers. The expression pattern and mechanisms of SNHG17 in glioma still await verification. METHODS: Paired glioma samples were enrolled. SNHG17, miR-23b-3p, and zinc-fingers and homeoboxes 1 (ZHX1) mRNA expression were examined by a quantitative real-time polymerase chain reaction (qRT-PCR). SNHG17 short hairpin RNA (shRNA) and miR-23b-3p mimics were transfected into LN229 and U251 cell lines to repress SNHG17 and up-regulate miR-23b-3p expression, respectively. Proliferation, migration and invasion of LN229 and U251 cells were probed by a cell counting kit-8 assay and a Transwell assay. Bioinformatics prediction, dual-luciferase reporter assay, RNA immunoprecipitation assay, qRT-PCR and western blotting were applied to determine the regulatory relationships among SNHG17, miR-23b-3p and ZHX1. RESULTS: SNHG17 expression was markedly raised in glioma tissues, which was positively correlated with ZHX1 expression and negatively associated with the expression of miR-23b-3p. After transfection of SNHG17 shRNAs into glioma cells, the proliferation, migration and invasion of cancer cells was markedly restrained. miR-23b-3p mimics the function of SHNG17 knockdown. Furthermore, miR-23b-3p was shown to be negatively modulated by SNHG17, and ZHX1 was identified as a target of miR-23b-3p. CONCLUSIONS: SNHG17 is a "competing endogenous RNA" with respect to modulating ZHX1 expression by adsorbing miR-23b-3p and thereby promoting glioma progression.


Assuntos
Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Proteínas de Homeodomínio/metabolismo , RNA Longo não Codificante/genética , Fatores de Transcrição/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Glioma/genética , Glioma/metabolismo , Glioma/cirurgia , Proteínas de Homeodomínio/genética , Humanos , MicroRNAs/genética , Invasividade Neoplásica , Prognóstico , Fatores de Transcrição/genética , Células Tumorais Cultivadas
17.
Biochem Genet ; 57(4): 507-521, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30697640

RESUMO

Non-alcoholic fatty liver disease is one of the main causes of chronic liver disease and therefore is currently considered a major public health problem. Sirtuin 1 (SIRT1) is an NAD-dependent deacetylase enzyme that contributes in the regulation of metabolic processes and protects against lipid accumulation in hepatocytes. Its expression is potentially regulated by microRNAs which attach to the 3' untranslated region (3'-UTR) of their target mRNA. HepG2 cells were incubated by glucose to induce lipid accumulation and were subsequently transfected with mir-23b mimic and inhibitor. Real-time PCR was used for measuring the expression of mir-23b and SIRT1 mRNA. Cell survival assay and intracellular triglyceride measurement were performed using colorimetric methods. Determination of SIRT1 protein level and activity were done by western blot and fluorometric analysis, respectively. The interaction of miR-23b with 3'-UTR of SIRT1 mRNA was confirmed by dual luciferase. miR-23b mimic inhibited gene and protein expression of SIRT1, while the inhibitor of miR-23b significantly elevated the expression levels of SIRT1 mRNA and protein. The results showed that the 3'-UTR of SIRT1 mRNA is a direct target for miR-23b. The intracellular triglyceride level was increased following the inhibition of SIRT1 in transfected HepG2 cell by miR-23b mimic. Cell viability was decreased in response to miR-23b upregulation compared to control cells. miR-23b reduces the expression and activity of SIRT1 and therefore may be a causative factor in the enhancement of lipid accumulation in HepG2 cells.


Assuntos
Hepatócitos/metabolismo , Metabolismo dos Lipídeos/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Regiões 3' não Traduzidas , Sobrevivência Celular/genética , Regulação para Baixo , Células HEK293 , Células Hep G2 , Humanos , Modelos Biológicos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sirtuína 1/metabolismo , Triglicerídeos/metabolismo , Regulação para Cima
18.
Cell Physiol Biochem ; 50(3): 823-840, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30355917

RESUMO

BACKGROUND/AIMS: Accumulating evidence strongly suggests that microRNAs (miRNAs) modulate the expression of known tumor suppressor genes and oncogenes. In the present study, we found that the proliferation and invasion ability of pancreatic ductal adenocarcinoma (PDAC) cells were significantly suppressed by the overexpression of miR-23b-3p. In addition, there are miR-23b-3p binding sites in annexin A2 (ANXA2). Here, we investigated whether miR-23b-3p had an impact on the progression and metastasis of PDAC by targeting ANXA2. METHODS: Cell proliferation, migration, and invasion, and cell cycle assays were performed to explore the effect of miR-23b-3p on various malignant phenotypes of pancreatic cancer cells. The size of tumors was observed following miR-23b-3p overexpression in an in vivo chick chorioallantoic membrane assay. Dual-luciferase reporter, quantitative real-time PCR, western blot, and immunohistochemical analyses were used to validate the relationship between miR-23b-3p and ANXA2 in vitro. RESULTS: We observed that miR-23b-3p could bind specifically to the 3' untranslated region of ANXA2 and inhibit its expression. MiR-23b-3p overexpression downregulated the expression of ANXA2 mRNA in PDAC cells and limited the size of tumors or even prevented tumor formation. In addition, there was a negative correlation between miR-23b-3p expression and ANXA2 protein expression in clinical specimens. CONCLUSION: MiR-23b-3p inhibits the development and progression of PDAC by regulating ANXA2 directly.


Assuntos
Anexina A2/metabolismo , Carcinoma Ductal Pancreático/patologia , MicroRNAs/metabolismo , Neoplasias Pancreáticas/patologia , Regiões 3' não Traduzidas , Adulto , Animais , Anexina A2/genética , Antagomirs/metabolismo , Sequência de Bases , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Galinhas , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/patologia , Feminino , Humanos , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Pessoa de Meia-Idade , Neovascularização Patológica/patologia , Neoplasias Pancreáticas/metabolismo , Alinhamento de Sequência
19.
Cell Biol Int ; 42(3): 313-323, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29064597

RESUMO

High concentrations of plasma lipoprotein(a) [Lp(a)] have been inferred to be an independent risk factor for cardiovascular and cerebrovascular diseases, such as coronary artery diseases, restenosis, and stroke. Apolipoprotein(a) [apo(a)] is one of the most important components of Lp(a) and contributes greatly to the increased concentration of plasma Lp(a). As a critical positive transacting factor of apo(a) gene, Ets1 has been proven as a target gene of several miRNAs, such as miR-193b, miR-125b-5p, miR-200b, miR-1, and miR-499. In this study, a series of experiments on miRNAs and relative miRNAs inhibitor delivered HepG2 cells were conducted, and two miRNAs that downregulate the apo(a) by targeting the 3'-UTR of Ets1 were identified. Results showed that apo(a) and Ets1 were differentially expressed in SMMC7721 and HepG2 cell lines. Meanwhile, apo(a) and Ets1 were inversely correlated with several hepatic endogenous miRNAs, such as miR-125b-5p, miR-23b-3p, miR-26a-5p, and miR-423-5p, which were predicted to bind to Ets1. Results show that miR-125b-5p and miR-23b-3p mimics could inhibit the synthesis of apo(a) by directly targeting Ets1 in HepG2, thereby reducing the plasma Lp (a) concentration.


Assuntos
Apolipoproteínas A/biossíntese , MicroRNAs/metabolismo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Regiões 3' não Traduzidas , Apolipoproteínas A/genética , Apolipoproteínas A/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Regulação para Baixo , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroRNAs/genética , Proteína Proto-Oncogênica c-ets-1/genética
20.
Diabetologia ; 59(3): 644-54, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26687158

RESUMO

AIMS/HYPOTHESIS: The mechanisms underlying the cellular metabolic memory induced by high glucose remain unclear. Here, we sought to determine the effects of microRNAs (miRNAs) on metabolic memory in diabetic retinopathy. METHODS: The miRNA microarray was used to examine human retinal endothelial cells (HRECs) following exposure to normal glucose (N) or high glucose (H) for 1 week or transient H for 2 days followed by N for another 5 days (H→N). Levels of sirtuin 1 (SIRT1) and acetylated-nuclear factor κB (Ac-NF-κB) were examined following transfection with miR-23b-3p inhibitor or with SIRT1 small interfering (si)RNA in the H→N group, and the apoptotic HRECs were determined by flow cytometry. Retinal tissues from diabetic rats were similarly studied following intravitreal injection of miR-23b-3p inhibitor. Chromatin immunoprecipitation (ChIP) analysis was performed to detect binding of NF-κB p65 to the potential binding site of the miR-23b-27b-24-1 gene promoter in HRECs. RESULTS: High glucose increased miR-23b-3p expression, even after the return to normal glucose. Luciferase assays identified SIRT1 as a target mRNA of miR-23b-3p. Reduced miR-23b-3p expression inhibited Ac-NF-κB expression by rescuing SIRT1 expression and also relieved the effect of metabolic memory induced by high glucose in HRECs. The results were confirmed in the retina using a diabetic rat model of metabolic memory. High glucose facilitated the recruitment of NF-κB p65 and promoted transcription of the miR-23b-27b-24-1 gene, which can be suppressed by decreasing miR-23b-3p expression. CONCLUSIONS/INTERPRETATION: These studies identify a novel mechanism whereby miR-23b-3p regulates high-glucose-induced cellular metabolic memory in diabetic retinopathy through a SIRT1-dependent signalling pathway.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/metabolismo , Glucose/metabolismo , MicroRNAs/fisiologia , Sirtuína 1/metabolismo , Animais , Linhagem Celular , Retinopatia Diabética/genética , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Sirtuína 1/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa