MicroRNAs (miRNAs) have been extensively studied over the decades and have been identified as potential molecular targets for cancer therapy. To date, many miRNAs have been found participating in the tumorigenesis of non-small cell lung cancer (NSCLC). The present study was designed to evaluate the functions of miR-125b-1-3p in NSCLC cells.MiR-125b-1-3p expression was detected in tissue samples from 21 NSCLC patients and in NSCLC cell lines using the real-time polymerase chain reaction. A549 cell lines were transfected with a miR-125b-1-3p mimic or miR-125b-1-3p antisense. Cell counting kit-8, wound healing, Matrigel invasion assays, and flow cytometry were used to assess the effects of these transfections on cell growth, migration, invasion, and apoptosis, respectively. Western blotting was used to detect apoptosis-related proteins, expression of S1PR1, and the phosphorylation status of STAT3. Significant differences between groups were estimated using Student's t-test or a one-way analysis of variance.MiR-125b-1-3p was downregulated in NSCLC samples and cell lines. Overexpression of miR-125b-1-3p inhibited NSCLC cell proliferation (37.8 ± 9.1%, t = 3.191, P = 0.013), migration (42.3 ± 6.7%, t = 6.321, P = 0.003), and invasion (57.6 ± 11.3%, t = 4.112, P = 0.001) and simultaneously induced more NSCLC cell apoptosis (2.76 ± 0.78 folds, t = 3.772, P = 0.001). MiR-125b-1-3p antisense resulted in completely opposite results. S1PR1 was found as the target gene of miR-125b-1-3p. Overexpression of miR-125b-1-3p inhibited S1PR1 protein expression (27.4 ± 6.1% of control, t = 4.083, P = 0.007). In addition, S1PR1 siRNA decreased STAT3 phosphorylation (16.4 ± 0.14% of control, t = 3.023, P = 0.015), as in cells overexpressing miR-125b-1-3p (16.7 ± 0.17% of control, t = 4.162, P = 0.026).Our results suggest that miR-125b-1-3p exerts antitumor functions in NSCLC cells by targeting S1PR1.miR-125-1-3p通过抑制S1PR1基因抑制非小细胞肺癌细胞摘要背景:微小RNA作为潜在的肿瘤治疗靶点目标,近年来引起了诸多研究者的关注。迄今为止,有诸多miRNA被发现在非小细胞肺癌(NSCLC)中起到重要作用。本次研究目的探索miR-125-1-3p在NSCLC中的作用及功能。 方法:MiR-125b-1-3p在21个NSCLC患者组织以及NSCLC细胞系中的表达水平采用RT-PCR进行检测。利用miR-125b-1-3p mimic 或者 miR-125b-1-3p antisense转染NSCLC A549细胞系用来过表达或沉默miR-125b-1-3p。CCK-8, 划痕实验,侵袭实验以及流式细胞分别用来检测miR-125b-1-3p对细胞增殖,迁移,侵入以及凋亡的影响。Western blotting用来检测凋亡相关蛋白,S1PR1蛋白以及STAT3蛋白的磷酸化状态。 结果:MiR-125b-1-3p在NSCLC组织以及细胞系中表达下调。过表达miR-125b-1-3p后可以抑制NSCLC细胞的增殖 (37.8 ± 9.1%, t = 3.191, P = 0.013),迁移 (42.3 ± 6.7%, t = 6.321, P = 0.003) 以及侵袭 (57.6 ± 11.3%, t = 4.112, P = 0.001)。与此同时,过表达miR-125b-1-3p可以同时诱导NSCLC细胞凋亡 (2.76 ± 0.78 fold, t = 3.772, P = 0.001)。抑制miR-125b-1-3p则取得相反的表型结果。S1PR1被发现是miR-125b-1-3p的调控基因。过表达miR-125b-1-3p能够抑制S1PR1蛋白的表达 (27.4 ± 6.1% of control, t = 4.083, P = 0.007)。此外过表达miR-125b-1-3p (16.7 ± 0.17% of control, t = 4.162, P = 0.026) 以及沉默S1PR1蛋白 (16.4 ± 0.14% of control, t = 3.023, P = 0.015) 后能够抑制STAT3蛋白的磷酸化。 结论:我们的发现揭示了miR-125b-1-3能够通过抑制S1PR1,从而在NSCLC中起到抑癌基因的作用。.
A method of establishing the geometrical model of leaf shape is used. The method starts with the binarization and edge detection of the digital image. According to the topology of the pixels in the edge, a linked list of Coordinates is created by the scanning algorithm. With the feature points detected from the linked list, the geometrical model of leaf shape is upbuilt by the rational cubic spline interpolation. Some experiment results show that this method is more simple and convenient and needs less data.
Abstract Background Mycobacterium chimaera infections subsequent to cardiac surgery are related to contaminated heater-cooler devices, with high mortality. Nevertheless, few studies have been reported in Asia. Case presentation We described the case of a 55-year-old man with Mycobacterium chimaera infection following cardiac surgery in the mainland of China. He was diagnosed with endocarditis caused by Mycobacterium chimaera subsequent to open heart surgery. Metagenomic next-generation sequencing (mNGS) and 16S rRNA gene PCR analysis were used to identify potential pathogens. The patient underwent redo valve replacement surgery and received combination therapy with azithromycin, ethambutol, linezolid, and amikacin. No signs of relapse were observed during the 11-month follow-up visit. Conclusions This is the first documented case of Mycobacterium chimaera infection following cardiac surgery in the mainland of China and the first documented transnational imported case worldwide. Moreover, mNGS is a novel diagnostic technology that can guide antimicrobial therapy prior to obtaining fluid/tissue culture results for Mycobacterium chimaera , providing a new approach for the detection of potential Mycobacterium chimaera infection.
The enhanced proliferation of mesenchymal stem cells (MSCs) can be helpful for the clinical translation of cell therapy. Low-level laser irradiation (LLLI) has been demonstrated as regulating MSC proliferation. MicroRNAs (miRNAs) are involved in various pathophysiologic processes in stem cells, but the role of miRNAs in the LLLI-based promotion of MSC proliferation remains unclear. We found that the proliferation level and cell cycle-associated genes in MSCs were increased after LLLI treatment in a time-dependent manner. Microarray assays revealed subsets of miRNAs to be differentially regulated, and these dynamic changes were confirmed by quantitative real-time polymerase chain reaction (qRT-PCR) after LLLI. miR-193 was the most highly up-regulated miRNA, and the change in it was related with the proliferation level. Gain-loss function experiments demonstrated that miR-193 could regulate the proliferation of MSCs, including human's and rat's, but could not affect the apoptosis and differentiation level. Blockade of miR-193 repressed the MSC proliferation induced by LLLI. By qRT-PCR, we found that miR-193, in particular, regulated cyclin-dependent kinase 2 (CDK2) expression. Bioinformatic analyses and luciferase reporter assays revealed that inhibitor of growth family, member 5 (ING5) could be the best target of miR-193 to functionally regulate proliferation and CDK2 activity, and the mRNA and protein level of ING5 was regulated by miR-193. Furthermore, the ING5 inhibited by small interfering RNA (siRNA) could up-regulate the proliferation of MSCs and the expression of CDK2. Taken together, these results strongly suggest that miR-193 plays a critical part in MSC proliferation in response to LLLI stimulation, which is potentially amenable to therapeutic manipulation for clinical application.
Abstract The purpose of this study was to investigate the fate of transplanted cells in the central zone of myocardial infarction (MI), and to clarify the relationship between the injection‐site impact and the efficacy of cell therapy. MI was created by coronary ligation in female rats. Three weeks later, 3‐million labelled male bone marrow mesenchymal stem cells (BMSCs) were directly injected into the border (BZC group) or central zone (CZC group) of MI area. As a control, culture medium was injected into the same sites. Cell survival was evaluated by quantitative real‐time polymerase chain reaction, and apoptosis was assayed with TUNEL and caspase‐3 staining. Four weeks after transplantation, heart function and cardiac morphometry were evaluated by echocardiography and Masson’s Trichrome staining, respectively. Angiogenesis and myogenesis were detected by immunofluorescence staining. After cell transplantation into the border or central zone, there was no cell migration between the different zones of MI. BMSCs in the CZC group exhibited no difference in apoptotic percentage, in the long‐term survival, when compared with those in the BZC group. However, they did effectively promote angiogenesis and cellular myogenic differentiation. Although cell delivery in the central zone of MI had no effect on the recovery of heart function compared with the BZC group, the retained BMSCs could still increase the scar thickness, and subsequently exhibit a trend in the reverse remodelling of ventricular dilation. Hence, we concluded that the central zone of MI should not be ignored during cell‐based therapy. Multiple site injection (border+central zone) is strongly recommended during the procedure of cell transplantation.
Abstract Background: The left atrial appendage (LAA) is the primary source of cardiogenic embolism in atrial fibrillation patients. Traditional LAA closure methods have shown suboptimal clinical outcomes. In recent years, some surgeons have proposed employing a pericardial slice for suturing the LAA. Methods: This study included patients with atrial fibrillation who underwent LAA closure during mitral valve surgery at our center between January 2019 and December 2021. There are two types of left atrial appendage closure surgical methods. The transesophageal echocardiogram performed 6 months after surgery will be used to verify the success of left atrial appendage closure. Left atrial appendage closure failure will be defined as reopening of the left atrial appendage or constant blood flow into the left atrial appendage. Result: Among the 104 patients included (mean age: 65.0 ± 7.58 years, male: 52.50%), a total of 7 patients were found to have reopened left atrial appendage, of which 5 patients belonged to the suture closure group and 2 patients (15.6% vs 2.8%, p=0.047) belonged to the pericardial patch exclusion group. Conclusions: The pericardial patch exclusion technique is a reliable technique to close the left atrial appendage in patients with atrial fibrillation, which can effectively reduce the possibility of reopening the left atrial appendage.
Axion-photon oscillation effect provides a possible explanation for the presence of very-high-energy (VHE) $\gamma$-ray signals from distant sources. In this work, we propose a model-dependent method to select possible sources that may give sufficient constraints on the axion parameters. We investigate such effect in the spectra of active galactic nuclei (AGN) B2 2234+28A and 3C 454.3 based on data obtained from Fermi Large Area Telescope (Fermi-LAT) and MAGIC U.L. We utilize the Markov Chain Monte Carlo method to fit the axion parameters, yielding a result of $g_{a\gamma}=3.05^{+0.51}_{-0.31} \times 10^{-11}$ GeV$^{-1}$ for the axion-photon coupling strength and $m_{a}=5.25^{+2.35}_{-2.65} \times 10^{-8} $ eV for the axion mass. We also perform 95\% confidence level (CL) constraints to set an upper limit for $g_{a\gamma}$.
The aim of the present study was to investigate whether miR‑203 can inhibit transforming growth factor‑β (TGF‑β)‑induced epithelial‑mesenchymal transition (EMT), and the migration and invasion ability of non‑small cell lung cancer (NSCLC) cells by targeting SMAD3. In the present study, the expression levels of miR‑203, SMAD3 mRNA and protein in NSCLC tissues were examined, as well as their corresponding paracancerous samples. The miR‑203 mimics and miR‑203 inhibitor were transfected into the H226 cell line. RT‑qPCR was used to assess the expression levels of E‑cadherin, Snail, N‑cadherin and vimentin mRNA, and western blotting was performed to detect the expression levels of p‑SMAD2, SMAD2, p‑SMAD3, SMAD3 and SMAD4. The cell migration and invasion abilities were detected by Transwell assays. The target site of SMAD3 was predicted by the combined action between miR‑203 and dual luciferase. The results revealed that the RNA levels of miR‑203, compared with paracancerous tissues, were decreased in NSCLC tissues, while SMAD3 mRNA and protein levels were upregulated, and miR‑203 inhibited SMAD3 expression. Induction of TGF‑β led to decreased E‑cadherin mRNA levels, upregulation of Snail, N‑cadherin and vimentin mRNA levels (P<0.05), and significant increase in cell migration and invasion, whereas transfection of miR‑203 mimics reversed the aforementioned results (P<0.05). Conversely, miR‑203 inhibitor could further aggravate the aforementioned results (P<0.05). Western blot results revealed that transfection of miR‑203 mimics significantly reduced the protein expression of SMAD3 and p‑SMAD3 (P<0.05). Furthermore, the results of the Dual‑Luciferase assay revealed that miR‑203 inhibited SMAD3 expression by interacting with specific regions of its 3'‑UTR. Overall, a novel mechanism is revealed, in which, miR‑203 can inhibit SMAD3 by interacting with specific regions of the 3'‑UTR of SMAD3, thereby restraining TGF‑β‑induced EMT progression and migration and invasion of NSCLC cells.