scholarly journals Exosomal let-7f-5p Derived from Mineralized Osteoblasts Promotes the Angiogenesis of Endothelial Cells via DUSP1 / Erk1/2 Signaling Pathway

Author(s):  
Yiqun He ◽  
Hailong Li ◽  
Zuochong Yu ◽  
Linli Li ◽  
Xujun Chen ◽  
...  

Abstract Background: Angiogenesis is essential for the tissue engineering bone formation, and osteoblasts (OBs) has been proved to play an important role in angiogenesis via various pro-angiogenic factors. However, whether the mineralized osteoblast derived exosomes (MOB-Exos) and containing let-7f-5p can promote the angiogenesis of endothelial cells (ECs) is still unknown.Methods: MOB-Exos, let-7f-5p mimicked MOB-Exos (miR mimic group) and let-7f-5p inhibited MOB-Exos (miR inhibitor group) were respectively harvested from mineralized osteoblasts (MOBs) and then co-cultured with bEnd.3. Besides, the Erk1/2 signaling pathway in ECs in miR mimic group was inhibited. Subsequently, CCK-8 assays, wound healing assays, transwell migration assays and tube formation assays were performed to detect the angiogenic capability of ECs. Dual luciferase reporter assays were conducted to verify the target genes of exosomal let-7f-5p. Results: The results showed that MOB-Exos could significantly promote the angiogenesis of ECs, which could be enhanced by mimicking exosomal let-7f-5p, and attenuated by inhibiting exosomal let-7f-5p. And the angiogenic capability of ECs was partly impaired after inhibiting the Erk1/2 signaling pathway despite co-cultured with let-7f-5p mimicked MOB-Exos. Moreover, let-7f-5p suppressed the luciferase activity of wide-type DUSP1, while mutation of DUSP1 abrogated the repressive ability of let-7f-5p. Conclusion: Based the results, our study concluded that exosomal let-7f-5p derived from MOBs could promote the angiogenesis of ECs via activating DUSP1/Erk1/2 signaling pathway, which might be a promising target for tissue engineering bone formation.

2020 ◽  
Author(s):  
Qiong Wang ◽  
Guanwen Wang ◽  
Lianjie Niu ◽  
Shaorong Zhao ◽  
Jianjun Li ◽  
...  

Abstract Background: Hepatocellular carcinoma (HCC), the most common primary liver cancer, rely on the formation of new blood vessel for growth and frequent intrahepatic and extrahepatic metastasis. Therefore, it is important to explore the underlying molecular mechanisms of tumor angiogenesis of HCC. Recently, microRNAs have been shown to modulate angiogenic processes by modulating the expression of critical angiogenic factors. However, the potential roles of tumor-derived exosomal microRNAs in regulating tumor angiogenesis remain to be elucidated. Methods: MiRNome sequencing was performed to uncover the miRNAs that are dysregulated in HCC patient serum-derived exosomes. Expression levels of miR-1290 in tissues and cells were determined by quantitative real-time PCR. The effect of mir-1290 on proliferation was evaluated by CCK-8 assay. The angiogenic ability of cells were determined by transwell, wound-healing, tube formation and matrigel plug assays. SMMC-7721 xenograft tumor model was established in NOD-SCID nude mice using miR-1290 and NC antagomirs to determin the angiogenic effect of mir-1290 in vivo. Target protein expression was determined by western blotting. Dual luciferase reporter assay was performed to confirm the action of miR-1290 on downstream target genes including SMEK1. Results are reported as means ± S.D. and differences were tested for significance using 2-sided Student’s t-test.Results: In this study, our miRNome sequencing demonstrated that miR-1290 was overexpressed in HCC patient serum-derived exosomes, and we found that delivery of miR-1290 into human endothelial cells enhanced their angiogenic ability. Our results further revealed that SMEK1 is a direct target of miR-1290 in endothelial cells. MiR-1290 exerted its pro-angiogenic function, at least in part, by alleviating the inhibition of VEGFR2 phosphorylation done by SMEK1. Conclusions: Collectively, our findings provide evidence that miR-1290 is overexpressed in HCC and promotes tumor angiogenesis via exosomal secretion, implicating its potential role as a therapeutic target for HCC.


2021 ◽  
Author(s):  
Tian Rong Zhang ◽  
WeiQiang Huang

Abstract Background Angiogenesis is an important factor in promoting vascular repair and a valuable process in the treatment of cardiovascular diseases. Circular RNAs (circRNAs) are widely expressed in eukaryotic cells and play an important role in the regulation of endothelial cells (ECs). In our study, bioinformatics analysis and real-time fluorescent PCR detection revealed that circRNA 0010928 (circ-0010928) is differentially expressed in human cardiac microvascular endothelial cells (HCMECs). Material & Methods We evaluated the role of circ-0010928 in HCMECs. Then, we can verify the function of circ-0010928 in HCMECs by cell counting kit-8 (CCK8), scratch test, transwell experiment, tube forming experiment, flow cytometry. Use dual luciferase experiment to detect the binding relationship between circ-0010928, miR-921 and LSM14A. Results Overexpression of circ-0010928 inhibited the proliferation, migration and tube formation of HCMECs under hypoxic conditions and promoted their apoptosis. In addition, dual luciferase reporter assays confirmed that circ-0010928 acted as a sponge of miR-921 and LSM14A as a downstream target gene of miR-921. Silencing miR-921 could also inhibit the proliferation, migration and tube formation of HCMECs and negatively regulate angiogenesis. Conclusion CircRNA-0010928 may inhibit the function of miRNA-921by combining with miRNA-921, and then miRNA-921 plays a role in regulating LSM14A, thereby regulating the state of angiogenesis.


2021 ◽  
Vol 12 ◽  
Author(s):  
Haijun Zhao ◽  
Yanhui He

Background: Diabetic retinopathy (DR) is the most important manifestation of diabetic microangiopathy. MicroRNAs (miRNAs), members of non-coding RNAs, have been frequently reported to regulate various diseases including DR. MiR-124-3p is involved in DR based on bioinformatics. The current study aimed to investigate the role of miR-124-3p in high glucose (HG)-treated human retinal microvascular endothelial cells (HRMECs), an in vitro model of DR.Methods: Bioinformatics analysis was applied to reveal the targets downstream miR-124-3p. A series of assays including CCK-8, luciferase reporter, western blot, and tube formation assays were used to explore the function and mechanism of miR-124-3p in HG-stimulated HRMECs.Results: We found out that miR-124-3p was downregulated in HG-stimulated HRMECs. Functionally, miR-124-3p overexpression restrained the HG-induced cell injury of HRMECs. Mechanistically, we predicted 5 potential target mRNAs of miR-124-3p. G3BP stress granule assembly factor 2 (G3BP2) was validated to bind with miR-124-3p. Rescue assays showed that miR-124-3p suppressed cell injury of HG-stimulated HRMECs through G3BP2. In addition, miR-124-3p regulated the p38MAPK signaling pathway by G3BP2, and G3BP2 promoted injury of HG-treated HRMECs through the activation of the p38MAPK signaling pathway.Conclusion: MiR-124-3p suppressed the dysfunctions of HG-treated HRMECs by targeting G3BP2 and activating the p38MAPK signaling. This new discovery provided a potential biomarker for DR treatment.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Xuewu You ◽  
Wenxiong Sun ◽  
Ying Wang ◽  
Xiaoli Liu ◽  
Aihong Wang ◽  
...  

Abstract Background Angiogenesis provides essential nutrients and oxygen for tumor growth and has become the main mechanism of tumor invasion and metastasis. Exosomes are nanoscale membrane vesicles containing proteins, lipids, mRNA and microRNA (miRNA), which mediate intercellular communication and play an important role in tumor progression. Accumulated evidence indicates that tumor-derived exosomal miRNAs participate in the tumor microenvironment and promote angiogenesis. Methods Bioinformatic target prediction and dual luciferase reporter assays were performed to identify the binding site between miR-663b and the 3′-UTR of vinculin (VCL). VCL overexpression lentivirus and miR-663b overexpression/inhibition lentivirus were used to create a VCL overexpression model and miR-663b overexpression/inhibition model in-vitro. Immunohistochemistry (IHC) assays and western blot assays were used to detect protein expression. Exosome-cell cocultures, wound healing assays, tube formation assays and transwell assays were used to measure the migration and tube formation ability of vascular endothelial cells [human umbilical vein endothelial cells (HUVECs)]. siRNA targeted VCL was used to knockdown VCL. Results In the present study, we found that miR-663b was elevated in cervical cancer tissue and exosomes. miR-663b could bind the 3′-UTR of VCL and inhibit its expression. VCL is downregulated in cervical cancer, and decreased VCL has a negative correlation with a high level of miR-663b. Further studies demonstrated that exosomes secreted by cervical cancer cells can deliver miR-663b to HUVECs and inhibit the expression of VCL, thereby promoting angiogenesis and tumor growth. Conclusions miR-663b derived from cancer cell exosomes acts as a driving factor for angiogenesis and a potential target of antiangiogenic therapy in cervical cancer. Our findings illustrated a new signaling pathway, including exosomes, miRNAs and target genes, which provides potential targets for antiangiogenic therapy.


2020 ◽  
Author(s):  
Tian Wang ◽  
Juan Liao ◽  
Zijie Yang ◽  
Weitao Shen ◽  
Zhi kui Gao ◽  
...  

Abstract Background Angiogenesis, a pivotal component in the tumor microenvironment (TME), boosts tumor growth and metastasis. Cancer-derived exosomes, which have been widely reported to play a crucial role in the establishment of TME can be effective angiogenic modulators. We aim to investigate the contribution of microRNA-21 (miR-21) for angiogenesis which was packaged in cancer-derived exosomes in esophageal squamous cell carcinoma (ESCC). Methods The co-cultivation model was constructed to mimic the tumor microenvironment based at a physical level to explore the effects of cancer-derived exosomes on angiogenesis of human umbilical vein endothelial cells (HUVECs). EdU assay, transwell assay and tube formation assay formation experiments were conducted for the evaluation of HUVECs proliferation, migration, and angiogenesis, respectively. In addition, Dual-luciferase reporter (DLR) assay was performed to validate the relationship between miR-21 and its target gene PTEN. Similarly, miR-21 inhibitors and LY294002 was applied to evaluate the regulation of miR-21 via pro-angiogenesis in recipient HUVECs by PTEN/Akt signaling pathway. Results After 24 h co-cultivation with EC9706 cells, miR-21 levels in recipient HUVECs was raised. The results from EdU assay, transwell assay and blood vessel formation experiment showed that exosomes which were secreted from EC9706 cells (EC9706-Exo) delivered miR-21 stimulated proliferation, migration and tube formation of HUVECs. DLR assay indicated that miR-21 could directly bind to the 3'-untranslated region (UTR) of PTEN genes, real-time PCR and western blot analysis for PTEN showed it was inhibited by EC9706-Exo shuttled miR-21. Meanwhile, phospho-Akt (p-Akt) (Ser473), one of the downstream genes of PTEN, was significantly increased in recipient HUVECs compared to the control group, while inhibiting miR-21 and PI3K/Akt pathway respectively both led to a sharp decrease in p-Akt levels, suggesting that exosomal miR-21 promote angiogenesis via activating PTEN/Akt signaling pathway. Conclusion Exosomal miR-21 acts as a driver of pro-angiogenesis by activating PTEN/Akt signaling pathway, it might serve as a blood-based biomarker for ESCC metastasis. Suppressing the expression or blocking the transmission of these exosome-derived miR-21 might be a novel antiangiogenic therapeutic strategy for ESCC.


2021 ◽  
Vol 18 ◽  
Author(s):  
Juxuan Ruan ◽  
Lei Wang ◽  
Jiheng Dai ◽  
Jing Li ◽  
Ning Wang ◽  
...  

Objective: Angiogenesis led by brain microvascular endothelial cells (BMECs) contributes to the remission of brain injury after brain ischemia reperfusion. In this study, we investigated the effects of hydroxysafflor yellow A(HSYA) on angiogenesis of BMECs injured by OGD/R via SIRT1-HIF-1α-VEGFA signaling pathway. Methods: The OGD/R model of BMECs was established in vitro by OGD for 2h and reoxygenation for 24h. At first, the concentrations of vascular endothelial growth factor (VEGF), Angiopoietin (ang) and platelet-derived growth factor (PDGF) in supernatant were detected by ELISA, and the proteins expression of VEGFA, Ang-2 and PDGFB in BMECs were tested by western blot; the proliferation, adhesion, migration (scratch healing and transwell) and tube formation experiment of BMECs; the expression of CD31 and CD34 were tested by immunofluorescence staining. The levels of sirtuin1(SIRT1), hypoxia-inducible factor-1α (HIF-1α), VEGFA mRNA and protein were tested. Results: HSYA up-regulated the levels of VEGF, Ang and PDGF in the supernatant of BMECs under OGD/R, and the protein expression of VEGFA, Ang-2 and PDGFB were increased; HSYA could significantly alleviate the decrease of cell proliferation, adhesion, migration and tube formation ability of BMECs during OGD/R; HSYA enhanced the fluorescence intensity of CD31 and CD34 of BMECs during OGD/R; HSYA remarkably up-regulated the expression of SIRT1, HIF-1α, VEGFA mRNA and protein after OGD/R, and these increase decreased after SIRT1 was inhibited. Conclusion: SIRT1-HIF-1α-VEGFA signaling pathway is involved in HSYA improves angiogenesis of BMECs injured by OGD/R.


2019 ◽  
Vol 86 (4) ◽  
pp. 425-431 ◽  
Author(s):  
Zhi Chen ◽  
Jingpeng Zhou ◽  
Xiaolong Wang ◽  
Yang Zhang ◽  
Xubin Lu ◽  
...  

AbstractWe established a mastitis model using exogenous infection of the mammary gland of Chinese Holstein cows with Staphylococcus aureus and extracted total RNA from S. aureus-infected and healthy mammary quarters. Differential expression of genes due to mastitis was evaluated using Affymetrix technology and results revealed a total of 1230 differentially expressed mRNAs. A subset of affected genes was verified via Q-PCR and pathway analysis. In addition, Solexa high-throughput sequencing technology was used to analyze profiles of miRNA in infected and healthy quarters. These analyses revealed a total of 52 differentially expressed miRNAs. A subset of those results was verified via Q-PCR. Bioinformatics techniques were used to predict and analyze the correlations among differentially expressed miRNA and mRNA. Results revealed a total of 329 pairs of negatively associated miRNA/mRNA, with 31 upregulated pairs of mRNA and 298 downregulated pairs of mRNA. Differential expression of miR-15a and interleukin-1 receptor-associated kinase-like 2 (IRAK2), were evaluated by western blot and luciferase reporter assays. We conclude that miR-15a and miR-15a target genes (IRAK2) constitute potential miRNA–mRNA regulatory pairs for use as biomarkers to predict a mastitis response.


2015 ◽  
Vol 117 (suppl_1) ◽  
Author(s):  
Qi Sun ◽  
Dongcao Lv ◽  
Qiulian Zhou ◽  
Yihua Bei ◽  
Junjie Xiao

MicroRNAs (miRNAs, miRs), endogenous small non-coding RNA, have been shown to act as essential regulators in angiogenesis which plays important roles in improving blood flow and cardiac function following myocardial infarction. The current study investigated the potential of miR-4260 in endothelial cell function and angiogenesis using human umbilical vein endothelial cells (HUVEC). Our data demonstrated that overexpression of miR-4260 was associated with increased proliferation and migration of HUVEC using EdU incorporation assay (17.25%±1.31 vs 25.78%±1.24 in nc-mimics vs miR-4260 mimics, respectively) and wound healing assay, respectively. While downregulation of miR-4260 inhibited the proliferation (17.90%±1.37 vs 10.66%±1.41 in nc-inhibitor vs miR-4260 inhibitor, respectively) and migration of HUVEC. Furthermore, we found that miR-4260 mimics increased (129.75±3.68 vs 147±3.13 in nc-mimics vs miR-4260 mimics, respectively), while miR-4260 inhibitor decreased the tube formation of HUVECs in vitro (123.25±2.17 vs 92±4.45 in nc-inhibitor vs miR-4260 inhibitor expression, respectively). Our data indicate that miR-4260 contributes to the proliferation, migration and tube formation of endothelial cells, and might be essential regulators for angiogenesis. Further study is needed to investigate the underlying mechanism that mediates the role of miR-4260 in angiogenesis by identifying its putative downstream target genes.


2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Qianwen Shao ◽  
Jing Xu ◽  
Rong Deng ◽  
Wei Wei ◽  
Bing Zhou ◽  
...  

Abstract Background Small nucleolar RNA host gene 6 (SNHG6) regulates diverse biological processes in cancers. Potential function of SNHG6 in human colon and rectal adenocarcinoma (CRC) was evaluated. Methods Quantitative real-time polymerase chain reaction, MTT assays, Colony formation assays, Transwell assay, Western Blotting and Luciferase reporter assays were performed to measure the biological functions and potential molecular mechanisms of SNHG6 in CRC. Results SNHG6 was over-expressed in CRC, and high expression of s SNHG6 were associated with short survival times. We then identified miR-101-3p as an inhibitory target of SNHG6. Knockdown of SNHG6 significantly decreased miR-101-3p expression. Moreover, silenced SNHG6 obviously inhibited CRC cell growth, weakened cell invasion capacity and blocked the Wnt/β-catenin signaling pathway. Conclusion SNHG6 could regulate the progression of CRC via modulating the expression levels of miR-101-3p and the activity of Wnt/β-catenin signaling.


Cells ◽  
2020 ◽  
Vol 9 (5) ◽  
pp. 1173 ◽  
Author(s):  
Mailin Gan ◽  
Shunhua Zhang ◽  
Yuan Fan ◽  
Ya Tan ◽  
Zhixian Guo ◽  
...  

Cardiac hypertrophy is a common pathological condition and an independent risk factor that triggers cardiovascular morbidity. As an important epigenetic regulator, miRNA is widely involved in many biological processes. In this study, miRNAs expressed in rat hearts that underwent isoprenaline-induced cardiac hypertrophy were identified using high-throughput sequencing, and functional verification of typical miRNAs was performed using rat primary cardiomyocytes. A total of 623 miRNAs were identified, of which 33 were specifically expressed in cardiac hypertrophy rats. The enriched pathways of target genes of differentially expressed miRNAs included the FoxO signaling pathway, dopaminergic synapse, Wnt signaling pathway, MAPK (mitogen-activated protein kinase) signaling pathway, and Hippo signaling pathway. Subsequently, miR-144 was the most differentially expressed miRNA and was subsequently selected for in vitro validation. Inhibition of miR-144 expression in primary myocardial cells caused up-regulation of cardiac hypertrophy markers atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). The dual luciferase reporter system showed that ANP may be a target gene of miR-144. Long non-coding RNA myocardial infarction associated transcript (LncMIAT) is closely related to heart disease, and here, we were the first to discover that LncMIAT may act as an miR-144 sponge in isoproterenol-induced cardiac hypertrophy. Taken together, these results enriched the understanding of miRNA in regulating cardiac hypertrophy and provided a reference for preventing and treating cardiac hypertrophy.


Sign in / Sign up

Export Citation Format

Share Document