hoechst staining
Recently Published Documents


TOTAL DOCUMENTS

85
(FIVE YEARS 28)

H-INDEX

17
(FIVE YEARS 3)

2021 ◽  
Vol 18 (1) ◽  
Author(s):  
Lei Yuan ◽  
Dongdong Zou ◽  
Xia Yang ◽  
Xin Chen ◽  
Youming Lu ◽  
...  

Abstract Background Communicating hydrocephalus (CH) is a common neurological disorder caused by a blockage of cerebrospinal fluid. In this study, we aimed to explore the potential molecular mechanism underlying CH development. Methods Quantitative proteomic analysis was performed to screen the differentially expressed proteins (DEPs) between patients with and without CH. A CH rat model was verified by Hoechst staining, and the co-localization of the target protein and neuron was detected using immunofluorescence staining. Loss-of-function experiments were performed to examine the effect of KLK6 on the synapse structure. Results A total of 11 DEPs were identified, and kallikrein 6 (KLK6) expression was found to be significantly upregulated in patients with CH compared with that in patients without CH. The CH rat model was successfully constructed, and KLK6 was found to be co-localized with neuronal nuclei in brain tissue. The expression level of IL-1β, TNF-α, and KLK6 in the CH group was higher than that in the control group. After knockdown of KLK6 expression using small-interfering RNA (siRNA), the expression levels of synapsin-1 and PSD95 in neuronal cells were increased, and the length, number, and structure of synapses were significantly improved. Following siRNA interference KLK6 expression, 5681 differentially expressed genes (DEGs) were identified in transcriptome profile. The upregulated DEGs of Appl2, Nav2, and Nrn1 may be involved in the recovery of synaptic structures after the interference of KLK6 expression. Conclusions Collectively, KLK6 participates in the development of CH and might provide a new target for CH treatment.


2021 ◽  
Vol 2021 ◽  
pp. 1-10
Author(s):  
Jun Zhang ◽  
Zhi-Nan Liu ◽  
Guo-Hua Deng

Here, through applying 2,6-bis(4′-carboxyl-phenyl)pyridine (H2L), a rigid ligand featuring both carboxylic acid and pyridine groups, a new coordination polymer containing Na(I) has been generated with the reaction between H2L ligand and NaNO3 in a water and DMF mixed solvent, and its chemical composition is [Na2L]n. Furthermore, the antiproliferative activity of Na(I) complex against the HXO-Rb44 retinoblastoma cells was detected with CCK-8 assay. Hoechst staining along with Annexin V-FITC/PI revealed that Na(I) complex induces the HXO-Rb44 retinoblastoma cells apoptosis. Flow cytometry analysis of reactive oxygen species (ROS) showed that Na(I) complex significantly increases the level of intracellular ROS. Importantly, western blot analysis revealed that Na(I) complex might induce apoptosis through inactivation of PI3K/AKT/mTOR pathway.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Zhao Liu ◽  
Jiuzheng Sun ◽  
Chuanzhi Li ◽  
Liyou Xu ◽  
Jun Liu

Abstract Background Histone modification plays essential roles in hepatocellular carcinoma (HCC) pathogenesis, but the regulatory mechanisms remain poorly understood. In this study, we aimed to analyze the roles of Megakaryoblastic leukemia 1 (MKL1) and its regulation of COMPASS (complex of proteins associated with Set1) in HCC cells. Methods MKL1 expression in clinical tissues and cell lines were detected by bioinformatics, qRT-PCR and western blot. MKL1 expression in HCC cells were silenced with siRNA, followed by cell proliferation evaluation via Edu staining and colony formation, migration and invasion using the Transwell system, and apoptosis by Hoechst staining. HCC cell tumorigenesis was assessed by cancer cell line-based xenograft model, combined with H&E staining and IHC assays. Results MKL1 expression was elevated in HCC cells and clinical tissues which was correlated with poor prognosis. MKL1 silencing significantly repressed proliferation, migration, invasion and colony formation but enhanced apoptosis in HepG2 and Huh-7 cells. MKL1 silencing also inhibited COMPASS components and p65 protein expression in HepG2 and Huh-7 cells. HepG2 cell tumorigenesis in nude mice was severely impaired by MKL1 knockdown, resulted into suppressed Ki67 expression and cell proliferation. Conclusion MKL1 promotes HCC pathogenesis by regulating hepatic cell proliferation, migration and apoptosis via the COMPASS complex and NF-κB signaling.


Molecules ◽  
2021 ◽  
Vol 26 (17) ◽  
pp. 5309
Author(s):  
Hyo Won Jung ◽  
Ji Hye Hwang

Anaplastic thyroid cancer (ATC) is one of the most fatal human malignancies. Ursi Fel (UF) is the bile of a brown bear that has been traditionally used for heat clearance and toxin relief in Korean and Chinese medicines. In this study, we determined the anticancer effects of a UF extract and its active compound, ursodeoxycholic acid (UDCA), in FRO human ATC cells. FRO cells were treated with UF extract and UDCA at different concentrations for various durations. Cell viability was measured using an MTT assay. Cell apoptosis was investigated by flow cytometric analysis following Annexin V and propidium iodide (PI) staining, and Hoechst staining was used to observe nuclear fragmentation. The expression of pro-apoptotic (Bax, caspase-3, cytochrome c, and PARP), anti-apoptotic (Bcl-2), and angiogenetic (TGF-β, VEGF, N-cadherin, and sirtuin-1) proteins and the phosphorylation of Akt and mechanistic target of rapamycin (mTOR) were determined by western blot analysis. Treatment with UF extract at 10, 25, and 50 μg/mL and UDCA at 25, 50, and 100 μM/mL significantly inhibited the growth of FRO cells in a dose-dependent manner. Flow cytometry and Hoechst staining revealed an increase in the apoptosis of FRO cells mediated by UF extract and UDCA in a dose-dependent manner. UF extract (25 and 50 μg) and UDCA (50 and 100 μM) significantly increased the expression of Bax, caspase-3, cytochrome c, and PARP and inhibited the expression of Bcl-2, TGF-β, VEGF, N-cadherin, and sirtuin-1 in FRO cells. Furthermore, UF extract and UDCA treatment stimulated Akt phosphorylation and inhibited mTOR phosphorylation in these cells. These results indicate that UF extract and UDCA exert anticancer properties in FRO cells by inducing apoptosis and inhibiting angiogenesis via regulating the Akt/mTOR signaling pathway.


2021 ◽  
Author(s):  
Mahsa Badrooh ◽  
Faezeh Shokrollahi ◽  
Shaghayegh Javan ◽  
Taraneh Ghasemipour ◽  
Samira Rezaei Mojdehi ◽  
...  

Abstract Seeking novel anticancer agents with minimal side effects against gastric cancer is vitally important. Copper, as an important trace element, takes roles in different physiologic pathways. Also, there is a higher demand for copper in cancer cells than normal ones. Copper complexes containing a therapeutic ligand could be promising candidates for gastric cancer chemotherapy. In this work, copper oxide nanoparticles were synthesized, functionalized with glutamic acid (CuO@Glu) and conjugated with thiosemicarbazone (Cuo@Glu/TSC NPs). The NPs were characterized and their antiproliferative potential against AGS cancer cells was investigated using MTT, flow cytometry, Hoechst staining, and caspase 3 activation assays. The FT-IR results showed the proper binding of TSC to CuO@Glu NPs and crystallinity of the prepared NPs was confirmed by the XRD pattern. The EDX analysis confirmed the presence of Cu, N, C, O, and S elements and lack of impurities. The Hydrodynamic size and zeta potential of the Cuo@Glu/TSC NPs were 710 nm and 27.5 mv, respectively. The NPs had spherical shape and were in a size range of 20-39 nm in diameter. This work revealed that CuO@Glu/TSC NPs efficiently inhibited the proliferation of AGS cells with significantly lower IC50 value (203 µg/mL) than normal HEK293 cells (IC50=435µg/mL). Flow cytometry and Hoechst staining obviously revealed apoptosis induction among CuO@Glu/TSC treated cells, and caspase-3 activity significantly increased by 1.4 folds. This study introduced CuO@Glu/TSC as an efficient anticancer against gastric cancer cells with lower toxicity toward normal cells which could be employed for cancer treatment after further characterization.


2021 ◽  
Vol 11 (8) ◽  
pp. 1560-1564
Author(s):  
Minhong Luo ◽  
Hao Liu ◽  
Caiju Mo ◽  
Qiang Wu ◽  
Zhenwei Chen ◽  
...  

This study investigated whether GLP-1 affects islet β cells in high glucose and the possible mechanism. INS-1 cells are separated into group A (treated with 5.6 mmol/L glucose), group B (group A+100 nmo1/L liraglutide), group C (cells were cultured in 16.7 mmol/L glucose) and group D (group C+100 nmo1/L liraglutide) followed by analysis of cell activity by CCK-8, apoptosis by Hoechst staining, INS level by ELISA, AMPK/mTOR signaling protein level by immunoblotting and RT-PCR. Compared with group A, cell proliferation was significantly increased in group B (P <0.05) and decreased in group C which had lower cell proliferation than group D (P <0.05). However, opposite results were obtained regarding cell apoptosis in four groups (P <0.05); group B showed higher INS level (145.36±8.55 pg/ml) and group C had lower level (80.14±5.36 pg/ml) than group A (105.23±7.78 pg/ml) (P <0.05). Meanwhile, AMPK and p-AMPK levels were significantly lower in group B and higher in group C than group A (P <0.05) with lower level in group D than group C (P <0.05). Whereas, opposite expression profile of p-mTOR and mTOR was found in these groups (P <0.05). High glucose inhibits INS-1 cell proliferation and promotes apoptosis. GLP-1 analogue inhibits INS-1 cell apoptosis in high glucose and accelerate proliferation possibly via regulation of AMPK/mTOR signaling pathway.


2021 ◽  
Vol 22 (14) ◽  
pp. 7586
Author(s):  
Dimitrios Agas ◽  
Reem Hanna ◽  
Stefano Benedicenti ◽  
Nicola De Angelis ◽  
Maria Giovanna Sabbieti ◽  
...  

Background: bone tissue regeneration remains a current challenge. A growing body of evidence shows that mitochondrial dysfunction impairs osteogenesis and that this organelle may be the target for new therapeutic options. Current literature illustrates that red and near-infrared light can affect the key cellular pathways of all life forms through interactions with photoacceptors within the cells’ mitochondria. The current study aims to provide an understanding of the mechanisms by which photobiomodulation (PBM) by 900-nm wavelengths can induce in vitro molecular changes in pre-osteoblasts. Methods: The PubMed, Scopus, Cochrane, and Scholar databases were used. The manuscripts included in the narrative review were selected according to inclusion and exclusion criteria. The new experimental set-up was based on irradiation with a 980-nm laser and a hand-piece with a standard Gaussian and flat-top beam profile. MC3T3-E1 pre-osteoblasts were irradiated at 0.75, 0.45, and 0.20 W in continuous-wave emission mode for 60 seconds (spot-size 1 cm2) and allowed to generate a power density of 0.75, 0.45, and 0.20 W/cm2 and a fluence of 45, 27, and 12 J/cm2, respectively. The frequency of irradiation was once, three times (alternate days), or five times (every day) per week for two consecutive weeks. Differentiation, proliferation, and cell viability and their markers were investigated by immunoblotting, immunolabelling, fluorescein-FragELTM-DNA, Hoechst staining, and metabolic activity assays. Results and conclusions: The 980-nm wavelength can photobiomodulate the pre-osteoblasts, regulating their metabolic schedule. The cellular signal activated by 45 J/cm2, 0.75 W and 0.75 W/cm2 consist of the PI3K/Akt/Bcl-2 pathway; differentiation markers were not affected, nor do other parameters seem to stimulate the cells. Our previous and present data consistently support the window effect of 980 nm, which has also been described in extracted mitochondria, through activation of signalling PI3K/Akt/Bcl-2 and cyclin family, while the Wnt and Smads 2/3-β-catenin pathway was induced by 55 J/cm2, 0.9 W and 0.9 W/cm2.


2021 ◽  
Author(s):  
Si Chen ◽  
Hanlin Wang ◽  
Tie Chen ◽  
Furong Deng ◽  
Hongbin Li ◽  
...  

Abstract Background: Gastric cancer is the most fatal digestive tract tumor. The current treatment of gastric cancer often causes adverse effects. Dihydroartemisinin (DHA), a first-line antimalarial drug, is a derivative of a compound from a well-known Chinese medicinal plant Artemisia annua. DHA demonstrates antitumor activities toward many different types of cancer while exerts no apparent adverse effects on normal cells, making it a promising lead compound for cancer treatment. DHA induces apoptosis in Gastric cancer cell line 7901 (SGC-7901). However, the exact mechanism of this antitumor activity remains not fully explored. Methods: A CCK-8 assay to detect cell viability with DHA treatment in gastric cancer SGC-7901. The colony formation was visualized by crystal violet staining. The DHA-treatment cells were stained by Annexin-V FITC/PI dye and then subject to cell flow cytometry. The apoptosis was further observed in the Hoechst staining assay. Real-time qPCR was conducted to detect apoptosis-related markers. Western blotting was conducted to detect the protein levels of the endoplasmic reticulum (ER) stress pathway-related proteins. KIRA6, an ER stress pathway inhibitor was applied to find out whether it could reverse the cell death. Results: DHA induced dose-dependent apoptosis in Gastric SGC-7901 cell with an IC50 of about 4 mg/ mL. It significantly increased the proportion of apoptotic cells in a dose-dependent pattern in the Annexin V/PI flow cytometry. Significantly higher percentages of cells with a more prominently stained nucleus were observed in the Hoechst staining assay. In qPCR assay, the mRNA level of Bcl-2 was significantly decreased while that of Bax was significantly increased in a dose-dependent manner after DHA treatment. In the western blot assay, increased Bax and Bim and decreased Caspase 9, Bcl-2 were observed. Consistently, the levels of pro-apoptotic proteins were increased while those of anti-apoptotic ones were decreased as shown in the Human Apoptosis Array assay after DHA treatment. DHA stimulated the expression of GRP78, ATF4, IRE1, CHOP, and phosphorylated c-Jun (p-c-Jun) as revealed in western blotting. The cell death caused by DHA treatment was reversed by KIRA6. Conclusions: DHA exerts its antitumor activity on SGC-7901 cells through the IRE1/c-Jun ER stress pathway.


2021 ◽  
Author(s):  
Lei Yuan ◽  
Dongdong Zou ◽  
Xia Yang ◽  
Xin Chen ◽  
Youming Lu ◽  
...  

Abstract Communicating hydrocephalus (CH) is a common neurological disorder caused by a blockage of cerebrospinal fluid. In this study, we aimed to explore the potential molecular mechanism underlying CH development. Quantitative proteomic analysis was performed to screen the differentially expressed proteins (DEPs) between patients with and without CH. A CH rat model was verified by Hoechst staining, and the co-localization of the target protein and neuron was detected using immunofluorescence staining. Loss-of-function experiments were performed to examine the effect of KLK6 on the synapse structure. A total of 11 DEPs were identified, and kallikrein 6 (KLK6) expression was found to be significantly upregulated in patients with CH compared with that in patients without CH. The CH rat model was successfully constructed, and KLK6 was found to be co-localized with neuronal nuclei in brain tissue. The expression level of KLK6 in the CH group was higher than that in the control group. After interference of KLK6 expression, the expression levels of synapsin-1 and PSD95 in neuronal cells were increased, and the length, number, and structure of synapses were significantly improved. The transcriptome profile (PRJNA719985) after interference of KLK6 expression was obtained, and 5,681 differentially expressed genes (DEGs) were identified. The upregulated DEGs of Appl2, Nav2, and Nrn1 may be involved in the recovery of synaptic structures after interference of KLK6 expression. Collectively, KLK6 participates in the development of CH and might provide a new target for CH treatment.


2021 ◽  
Author(s):  
Zhao Liu ◽  
Jiuzheng Sun ◽  
Chuanzhi Li ◽  
Liyou Xu ◽  
Jun Liu

Abstract Background Histone modification plays essential roles in hepatocellular carcinoma (HCC) pathogenesis, but the regulatory mechanisms remain poorly understood. In this study, we aimed to analyze the roles of Megakaryoblastic leukemia 1 (MKL1) and its regulation of COMPASS (complex of proteins associated with Set1) in HCC cells. Methods MKL1 expression in clinical tissues and cell lines were detected by bioinformatics, qRT-PCR and western blot. MKL1 expression in HCC cells were silenced with siRNA, followed by cell proliferation evaluation via Edu staining and colony formation, migration and invasion using the Transwell system, and apoptosis by Hoechst staining. HCC cell tumorigenesis was assessed by cancer cell line-based xenograft model, combined with H&E staining and IHC assays. Results MKL1 expression was elevated in HCC cells and clinical tissues which was correlated with poor prognosis. MKL1 silencing significantly repressed proliferation, migration, invasion and colony formation but enhanced apoptosis in HepG2 and Huh-7 cells. MKL1 silencing also inhibited COMPASS components and p65 protein expression in HepG2 and Huh-7 cells. HepG2 cell tumorigenesis in nude mice was severely impaired by MKL1 knockdown, resulted into suppressed Ki67 expression and cell proliferation. Conclusion MKL1 promotes HCC pathogenesis by regulating hepatic cell proliferation, migration and apoptosis via the COMPASS complex and NF-κB signaling.


Sign in / Sign up

Export Citation Format

Share Document