27 research outputs found

    Targeting oncogenic miR-335 inhibits growth and invasion of malignant astrocytoma cells

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Astrocytomas are the most common and aggressive brain tumors characterized by their highly invasive growth. Gain of chromosome 7 with a hot spot at 7q32 appears to be the most prominent aberration in astrocytoma. Previously reports have shown that microRNA-335 (miR-335) resided on chromosome 7q32 is deregulated in many cancers; however, the biological function of miR-335 in astrocytoma has yet to be elucidated.</p> <p>Results</p> <p>We report that miR-335 acts as a tumor promoter in conferring tumorigenic features such as growth and invasion on malignant astrocytoma. The miR-335 level is highly elevated in C6 astrocytoma cells and human malignant astrocytomas. Ectopic expression of miR-335 in C6 cells dramatically enhances cell viability, colony-forming ability and invasiveness. Conversely, delivery of antagonist specific for miR-335 (antagomir-335) to C6 cells results in growth arrest, cell apoptosis, invasion repression and marked regression of astrocytoma xenografts. Further investigation reveals that miR-335 targets disheveled-associated activator of morphogenesis 1(Daam1) at posttranscriptional level. Moreover, silencing of endogenous Daam1 (siDaam1) could mimic the oncogenic effects of miR-335 and reverse the growth arrest, proapoptotic and invasion repression effects induced by antagomir-335. Notably, the oncogenic effects of miR-335 and siDAAM1 together with anti-tumor effects of antagomir-335 are also confirmed in human astrocytoma U87-MG cells.</p> <p>Conclusion</p> <p>These findings suggest an oncogenic role of miR-335 and shed new lights on the therapy of malignant astrocytomas by targeting miR-335.</p

    Treatment for ischemic stroke: From thrombolysis to thrombectomy and remaining challenges

    No full text
    Stroke is a leading cause of death and long-term disabilities. Despite decades of extensive efforts in search of brain injury mechanisms and therapeutic interventions, pharmacological treatment is limited to the use of thrombolytic agent tissue plasminogen activator, which has limited therapeutic time window and potential side effect of intracranial hemorrhage. Over the past few years, endovascular thrombectomy with stent-retriever devices combined with advanced imaging modalities has transformed the standard of stroke care, offering an opportunity to improve the outcome in selected patients as late as 24 h after the onset of stroke. This mini-review summarizes the advancement in the treatment of ischemic stroke, from thrombolysis to thrombectomy and remaining challenges in the field

    Role of TRPM7 Channels in Hyperglycemia-Mediated Injury of Vascular Endothelial Cells

    Get PDF
    <div><p>This study investigated the change of transient receptor potential melastatin 7 (TRPM7) expression by high glucose and its role in hyperglycemia induced injury of vascular endothelial cells. Human umbilical vein endothelial cells (HUVECs) were incubated in the presence or absence of high concentrations of D-glucose (HG) for 72h. RT-PCR, Real-time PCR, Western blotting, Immunofluorescence staining and whole-cell patch-clamp recordings showed that TRPM7 mRNA, TRPM7 protein expression and TRPM7-like currents were increased in HUVECs following exposure to HG. In contrast to D-glucose, exposure of HUVECs to high concentrations of L-glucose had no effect. HG increased reactive oxygen species (ROS) generation, cytotoxicity and decreased endothelial nitric oxide synthase protein expression, which could be attenuated by knockdown of TRPM7 with TRPM7 siRNA. The protective effect of silencing TRPM7 against HG induced endothelial injury was abolished by U0126, an inhibitor of the extracellular signal-regulated kinase signaling pathway. These observations suggest that TRPM7 channels play an important role in hyperglycemia-induced injury of vascular endothelial cells.</p> </div

    Effect of TRPM7 siRNA on viability and cytotoxicity in HG treated HUVECs.

    No full text
    <p>The cells were preincubated with TRPM7 siRNA or control siRNA for 48h, and then stimulated with HG for 72h. (A) Confluent field by light microscopy. <i>Scale bar, 100 µM</i>. (B) Cell viability was assessed by MTT assay. (C) Cytotoxicity was assessed by LDH release assay. **<i>p</i><0.01 vs. control; <sup>##</sup><i>p</i><0.01 vs. control siRNA. n=5 for MTT and LDH assay.</p

    Effect of TRPM7 siRNA on MAPK pathway in HG treated HUVECs.

    No full text
    <p>The cells were preincubated with TRPM7 siRNA or control siRNA for 48h, and then stimulated with HG for 72h. (A) Representative immunoblots showing phospho-ERK1/2, phospho-p38MAPK and phospho-JNK protein expression level. (B) The corresponding bar showing the relative optical density of phospho-ERK1/2 protein normalized to total ERK1/2. **<i>p</i><0.01 vs. control; <sup>##</sup><i>p</i><0.01 vs. control siRNA. n=5 for immunoblotting (C) The corresponding bar graphs showing the relative optical density of phospho-p38MAPK protein normalized to beta-actin. (D) The corresponding bar graphs showing the relative expression of phospho-JNK protein normalized to beta-actin. n=4.</p

    Effect of TRPM7 siRNA on TRPM7 mRNA and protein expression in high D-glucose (HG, 30 mM) treated HUVECs.

    No full text
    <p>The cells were preincubated with TRPM7 siRNA or control siRNA for 48h, and then stimulated with HG for 72h. (A) TRPM7 protein level measured by immunofluorescence. <i>Scale bar, 100 µm</i>. (B) TRPM7 mRNA level detected by RT-PCR. (C) TRPM7 mRNA level normalized to GAPDH. (D) Representative immunoblots showing the level of TRPM7 protein expression. (E) The corresponding bar graphs showing the relative expression of TRPM7 protein normalized to beta-actin. **<i>p</i><0.01 vs. control; <sup>#</sup><i>p</i><0.05 vs. control siRNA, <sup>##</sup><i>P</i><0.01 vs. control siRNA. n=4 for RT-PCR and immunoblotting. </p

    Effect of HG on TRPM7 protein expression in HUVECs.

    No full text
    <p>(A) Representative immunoblots showing TRPM7 protein expression in HUVECs with or without HG (30 mM) for 72h. (B) The corresponding bar graphs showing relative expression of TRPM7 protein normalized to beta-actin. (C) Representative TRPM7-like currents recorded in HUVECs cultured in control or HG (30 mM) for 72h. (D) TRPM7-like current density. **<i>p</i><0.01 vs. HG; *<i>p</i><0.05 vs. control. n=4 for immunoblotting and 17-18 cells patched for current recording. </p

    Effect of U0126 on viability and phospho-ERK1/2 expression in HG treated HUVECs.

    No full text
    <p>The cells were preincubated with U0126 (10 μM) for 18h, then treated with TRPM7 siRNA for 48h, and finally stimulated with HG for 72h. (A) Cell viability was assessed by MTT assay. (B) Representative immunoblots showing phospho-ERK1/2 expression level. (C) The corresponding bar graphs showing the relative expression of phospho-ERK1/2 protein normalized to beta-actin. **<i>p</i><0.01 vs. control; <sup>##</sup><i>p</i><0.01 vs. control siRNA. n=5 for immunoblotting.</p

    Effect of TRPM7 siRNA on eNOS protein expression, NO and ROS generation in HG treated HUVECs.

    No full text
    <p>The cells were preincubated with TRPM7 siRNA or control siRNA for 48h, and then stimulated with HG for 72h. (A) Representative immunoblots showing eNOS protein expression level. (B) The corresponding bar graphs showing the relative expression of eNOS protein normalized to beta-actin. (C) The production of NO was determined by measurement of nitrite, a stable product of NO. (D) The production of intracellular ROS was assessed by the oxidation of 2’,7’-dichlorofluorescin diacetate to fluorescent 2’,7’-dichlorofluorescein. **<i>p</i><0.01 vs. control; <sup>##</sup><i>p</i><0.01 vs. control siRNA. n=3 for immunoblotting , 5 for ROS generation assay and 6 for NO measurement.</p

    Silencing TRPM7 in Mouse Cortical Astrocytes Impairs Cell Proliferation and Migration via ERK and JNK Signaling Pathways

    No full text
    <div><p>Transient receptor potential melastatin 7 (TRPM7), a non-selective cation channel, is highly expressed expressed in the brain and plays a critical role in ischemic neuronal death. Astrocyte, the most abundant cell type in central nervous system (CNS), exerts many essential functions in the physiological and pathological conditions. Here we investigated the expression and functions of the TRPM7 channel in mouse cortical astrocytes. Using reverse transcription (RT)-PCR, immunostaining, western blot and patch clamp recording, we showed that functional TRPM7 channel is expressed in cultured mouse cortical astrocytes. Knocking down TRPM7 with specific siRNA impairs the proliferation and migration of astrocytes by 40.2% ± 3.9% and 40.1% ± 11.5%, respectively. Consistently, inhibition of TRPM7 with 2-aminoethoxydiphenyl borate (2-APB) also decreases astrocyte proliferation and migration by 46.1% ± 2.5% and 64.2% ± 2.4%. MAPKs and Akt signaling pathways have been shown to be implicated in TRPM7-mediated responses including cell proliferation and migration. Our data show that suppression of TRPM7 in astrocytes reduces the phosphorylation of extracellular signal-regulated kinases (ERK) and c-Jun N-terminal kinases (JNK), but not p38 mitogen-activated protein kinase and Akt. In addition, TRPM7, as a cation channel, has been involved in the Ca<sup>2+</sup> and Mg<sup>2+</sup> homeostasis in several types of cells. In our study, we found that silencing TRPM7 decreases the intracellular basal Mg<sup>2+</sup> concentration without affecting Ca<sup>2+</sup> concentration in astrocytes. However, an addition of Mg<sup>2+</sup> to the growth medium could not rescue the impaired proliferation of astrocytes. Together, our data suggest that TRPM7 channel may play a critical role in the proliferation and migration of astrocytes via the ERK and JNK pathways.</p></div
    corecore