Knockout of CDK2 induced early apoptosis in A375 cells. (A) Apoptosis of A375 cells infected with lentiviruses sgCDK2-108 or sgCDK2-NC was determined by staining with Annexin V-Light 650 and propidium iodide through flow cytometry at 72 h. (B) The percentages of early, late and total apoptotic cells are displayed as histograms. The data are expressed the mean ± standard deviation of three independent experiments. * P<0.05 vs. sgCDK2-NC control. CDK, cyclin-dependent kinase; sg, single-guide; ns, not significant.

Knockout of CDK2 induced early apoptosis in A375 cells. (A) Apoptosis of A375 cells infected with lentiviruses sgCDK2-108 or sgCDK2-NC was determined by staining with Annexin V-Light 650 and propidium iodide through flow cytometry at 72 h. (B) The percentages of early, late and total apoptotic cells are displayed as histograms. The data are expressed the mean ± standard deviation of three independent experiments. * P<0.05 vs. sgCDK2-NC control. CDK, cyclin-dependent kinase; sg, single-guide; ns, not significant.

Source publication
Article
Full-text available
Cutaneous melanoma is one of the most common malignant skin tumors, with a continuously increasing incidence. Cyclin-dependent kinase (CDK) 2 is a key regulator of G1-S transition and modulation of G2 progression; however, its role in cancer is a matter of debate. In the present study, a lentivirus expressing single-guide RNA (sgRNA) was constructe...

Contexts in source publication

Context 1
... was then analyzed in A375 cells infected with lentiviruses sgCDK2-108 or sgCDK2-NC. Compared with the sgCDK2-NC group, early apoptosis of A375 cells infected with lentivirus sgCDK2-108 was observed (Fig. 3A). The rate of early apoptosis in A375 cells infected with lentivirus sgCDK2-108 was 11.76%, and the rate of total apoptosis reached 12.47% (P<0.05; Fig. 3B), suggesting that knockout of CDK2 induces early apoptosis in A375 ...
Context 2
... analyzed in A375 cells infected with lentiviruses sgCDK2-108 or sgCDK2-NC. Compared with the sgCDK2-NC group, early apoptosis of A375 cells infected with lentivirus sgCDK2-108 was observed (Fig. 3A). The rate of early apoptosis in A375 cells infected with lentivirus sgCDK2-108 was 11.76%, and the rate of total apoptosis reached 12.47% (P<0.05; Fig. 3B), suggesting that knockout of CDK2 induces early apoptosis in A375 ...

Citations

... Interestingly, it was reported that Sn nanocomposites are substantially more cytotoxic than gold (Au) complexes and they revealed exceptional antiproliferative characteristics in many cancer types. These nanocomposites arrested the cell cycle at the G0/G1 phase [13] with subsequent cell apoptosis as apoptosis occurs as a result of G0/G1 phase arrest [45]. ...
Article
Full-text available
This study aims to explore the efficacy of Copper/Tin (CuS/SnS) nanocomposites loaded into exosomes against skin cancer A431 cell line. CuS/SnS nanocomposites (S1, S2, S3) were synthesized and characterized, then loaded into exosomes (Exo) (S1‐Exo, S2‐Exo and S3‐Exo) and characterized. After that, the loaded samples were investigated in vitro against A431 using cytotoxicity, apoptosis, and cell cycle assays. CuS/SnS nanocomposites were indexed to hexagonal CuS structure and orthorhombic α‐SnS phase and showed nano‐rode shape. The exosomes loaded with nanocomposites were regular and rounded within the size of 120 nm, with no signs of broken exosomes or leakage of their contents. The cytotoxicity assay indicated the enhanced cytotoxic of S1‐Exo versus the free nano‐form S1 on A431. Interestingly, S1‐Exo recorded 1.109 times more than DOX in its anti‐skin cancer capacity. Moreover, S1‐Exo recorded 40.2% for early apoptosis and 22.1% for late apoptosis. Furthermore, it displayed impact in arresting the cancer cell cycle at G0/G1 phase and reducing G2/M phase. Noteworthy, loaded nanocomposites were safe against normal HSF skin cells. In conclusion, the loaded CuS/SnS nanocomposites into the exosomes could be of great potential as anti‐skin cancer candidates through induction of apoptosis and promotion of the cell cycle arrest at G0/G1 phase.
... Based on the results of the reverse docking and enzyme activity tests, we analyzed the expression of c-KIT downstream proteins using a Western blot assay with GAPDH serving as an internal control. Moreover, the downstream effectors of c-KIT-driven malignancies are the MAPK and PI3K pathways [28][29][30]. In this study, HCT116 cells were treated with various concentrations (0, 1.25 μM, 2.5 μM, and 5 μM) of T4 for 48 h to evaluate its biological effects on downstream targets of c-KIT by Western blot analysis. ...
Article
Full-text available
A series of novel echinatin derivatives with 1,3,4-oxadiazole moieties were designed and synthesized. Most of the newly synthesized compounds exhibited moderate antiproliferative activity against the four cancer cell lines. Notably, Compound T4 demonstrated the most potent activity, with IC50 values ranging from 1.71 µM to 8.60 µM against the four cancer cell lines. Cell colony formation and wound healing assays demonstrated that T4 significantly inhibited cell proliferation and inhibited migration. We discovered that T4 exhibited moderate binding affinity with the c-KIT protein through reverse docking. The results were effectively validated through subsequent molecular docking and c-KIT enzyme activity assays. In addition, Western blot analysis revealed that T4 inhibits the phosphorylation of downstream proteins of c-KIT. The results provide valuable inspiration for exploring novel insights into the design of echinatin-related hybrids as well as their potential application as c-KIT inhibitors to enhance the efficacy of candidates.
... Cyclin-dependent kinase 4 (CDK4) regulates the cell cycle and interacts with the MYC gene in this process. It participates in the transition from the G1 to the S phase of the cell cycle [39]. Overexpression of the MYC gene can result in the upregulation of CDK4 expression, thereby facilitating cell cycle progression and cellular division [40]. ...
Preprint
Full-text available
Background: The MYC gene is one of the regulatory and proto-oncogenic genes overexpressed in most prostate cancers (PCa). Studies have shown that abnormal expression of microRNAs is involved in the onset and development of many different types of human cancer, including prostate cancer. Methods and result: In this study, we first evaluated targeting the effect of miR-377 on MYC by luciferase assay. Real-time PCR was used to determine whether miR-377 could decrease the MYC mRNA in transfected PCa cell lines (PC-3 and DU145). Also, the expression of BCL-2/Bax, PTEN, and CDK4 mRNA levels were measured due to MYC degression. Also, the effects of miR-377 on apoptosis cells, proliferation, cell cycle, and wound healing were analyzed. We showed that miR-377 targets MYC mRNA by luciferase reporter assay. A significant reduction in MYC mRNA level was detected following miR-377 transfection in PC-3 and DU145 cell lines. Also, we demonstrated the decrease of BCL-2 and CDK4 and an increase in Bax, and PTEN in prostate cancer cell lines, following the reduction of MYC. Furthermore, we showed that the higher levels of miR-377 in PCa cell lines induced apoptosis, reduced proliferation, and migration, and stopped the cell cycle. Conclusion: All these data reveal that miR-377 functions as an MYC inhibitor in PCa and may serve as a potential therapeutic target for treating this cancer.
... CDK2 is a core cell cycle regulator during cell division [41]. Knockout of CDK2 blocked melanoma cells at G0/G1 phase [42], while other studies found that cinobufagin could induce cell cycle arrest at G2/M phase by inhibiting CDK2 [43]. And DC-K2IN212, a selective inhibitor of CDK2, could also block melanoma cells at S and G2/M phases [44]. ...
Article
Full-text available
Cisplatin (CDDP) is the first-line drug in the clinical treatment of esophageal squamous cell carcinoma (ESCC), which has severe nephrotoxicity. Diosmetin (DIOS) can protect kidney from oxidative damage, however, its function in ESCC is unknown. This study aims to explore the effect and mechanism of DIOS on ESCC and its combined effect with CDDP. Herein, we found that DIOS significantly inhibited the progression of ESCC in vitro and in vivo. Furthermore, the anti-tumor effect of DIOS was not statistically different from that of CDDP. Mechanically, transcriptomics revealed that DIOS inhibited the E2F2/RRM2 signaling pathway. The transcriptional regulation of RRM2 by E2F2 was verified by luciferase assay. Moreover, docking model, CETSA, pull-down assay and CDK2 inhibitor assay confirmed that DIOS directly targeted CDK2, leading to significant suppression of ESCC. Additionally, the patient-derived xenografts (PDX) model showed that the combination of DIOS and CDDP significantly inhibited the growth of ESCC. Importantly, the combined treatment with DIOS and CDDP significantly reduced the mRNA expression levels of kidney injury biomarkers KIM-1 and NGAL in renal tissue, as well as the levels of blood urea nitrogen, serum creatinine and blood uric acid compared to the single treatment with CDDP. In conclusion, DIOS could be an effective drug and a potential chemotherapeutic adjuvant for ESCC treatment. Furthermore, DIOS could reduce the nephrotoxicity of CDDP to some extent.
... Furthermore, they play a crucial role in the pathogenesis of cancers [22,23]. CDK1 interacts with cyclin B1, allowing the progression of the cell cycle through the G2 phase, while CDK2 is responsible for G1/S and S/G2 transition [23][24][25][26][27][28]. Moreover, CDK2 is involved in controlling proliferation, cell differentiation, adaptive immune response, and apoptosis. ...
Article
Full-text available
The current study continues the evaluation of the anticancer potential of three de novo synthesized pyrazolo[4,3-e]tetrazolo[1,5-b][1,2,4]triazine sulfonamides—MM129, MM130, and MM131—against human cancer cells of HeLa, HCT 116, PC-3, and BxPC-3 lines. The pro-apoptotic activity of the investigated sulfonamides was shown by observations of changes in the mitochondrial transmembrane potential of the tested cells, externalization of phosphatidylserine on the cellular membrane surface, and cell morphology in microscopic imaging. The computational studies have shown that MM129 exhibited the lowest binding energy values when docked against CDK enzymes. In addition, the highest stability was shown for complexes formed between MM129 and CDK5/8 enzymes. All examined compounds induced cell cycle arrest in the G0/G1 phase in the BxPC-3 and PC-3 cells and simultaneously caused the accumulation of cells in the S phase in the HCT 116 cells. In addition, the increase in the subG1 fraction was observed in PC-3 and HeLa cells. The application of a fluorescent H2DCFDA probe revealed the high pro-oxidative properties of the tested triazine derivatives, especially MM131. In conclusion, the obtained results suggest that MM129, MM130, and MM131 exhibited strong pro-apoptotic properties towards investigated cells, mainly against the HeLa and HCT 116 cell lines, and high pro-oxidative potential as well. Moreover, it is suggested that the anticancer activity of the tested compounds may be associated with their ability to inhibit CDK enzymes activities.
... The phosphorylation level of Rb (pRb) was decreased in a concentration-dependent manner by a 24 h HPF treatment, whereas the total Rb protein level was unchanged ( Figure 3B). Cyclin A2 is highly expressed in the S-phase when it is associated with CDK2 or CDK1 [44]. In addition to cyclin D1, cyclin A2 expression level was decreased after 24 h of treatment ( Figure 3B). ...
Article
Full-text available
Hyperforin (HPF), the main component responsible for the antidepressant action of Hypericum perforatum, displays additional beneficial properties including anti-inflammatory, antimicrobic, and antitumor activities. Among its antitumor effects, HPF activity on melanoma is poorly documented. Melanoma, especially BRAF-mutated melanoma, is still a high-mortality tumor type and the currently available therapies do not provide solutions. We investigated HPF’s antimelanoma effectiveness in A375, FO1 and SK-Mel-28 human BRAF-mutated cell lines. Cell viability assays documented that all melanoma cells were affected by low HPF concentrations (EC50% 2–4 µM) in a time-dependent manner. A Br-deoxy-uridine incorporation assay attested a significant reduction of cell proliferation accompanied by decreased expression of cyclin D1 and A2, CDK4 and of the Rb protein phosphorylation, as assessed by immunoblots. In addition, the expression of P21/waf1 and the activated form of P53 were increased in A375 and SK-Mel-28 cells. Furthermore, HPF exerts cytotoxic effects. Apoptosis is induced 24 h after HPF administration, documented by an increase of cleaved-PARP1 and a decrease of both Bcl2 and Bcl-xL expression levels. Autophagy is induced, attested by an augmented LC3B expression and augmentation of the activated form of AMPK. Moreover, HPF lowers GPX4 enzyme expression, suggesting ferroptosis induction. HPF has been reported to activate the TRPC6 Ca++ channel and/or Ca++ and Zn++ release from mitochondria stores, increasing cytosolic Ca++ and Zn++ concentrations. Our data highlighted that HPF affects many cell-signaling pathways, including signaling induced by Ca++, such as FRA1, pcJun and pCREB, the expression or activity of which are increased shortly after treatment. However, the blockage of the TRPC6 Ca++ channel or the use of Ca++ and Zn++ chelators do not hinder HPF cytostatic/cytotoxic activity, suggesting that damages induced in melanoma cells may pass through other pathways. Remarkably, 24 h after HPF treatment, the expression of activated forms of the transcription factors NF-κB P65 subunit and STAT3 are significantly lowered. Several cytosolic (PGM2, LDHA and pPKM2) and mitochondrial (UQCRC1, COX4 and ATP5B) enzymes are downregulated by HPF treatment, suggesting a generalized reduction of vital functions in melanoma cells. In line with these results is the recognized ability of HPF to affect mitochondrial membrane potential by acting as a protonophore. Finally, HPF can hinder both melanoma cell migration and colony formation in soft agar. In conclusion, we provide evidence of the pleiotropic antitumor effects induced by HPF in melanoma cells.
... [15] Previous studies have found that with the upregulation of CKS2, the mRNA of CDK2/4/6 proteins inhibited by the P21 protein gradually increased, [14] and CDK2 was involved in the development of melanoma by regulating the G1/S and G2 phases of the cell cycle. [16] Similarly, melanocyte loss due to the abnormal expression of CDK2 was observed in vitiligo patients, but the exact mechanism is unclear. [17] Akt phosphorylation is closely associated with cell proliferation and apoptosis. ...
Article
Full-text available
Previous studies have attempted to elucidate the molecular mechanism of vitiligo; however, its pathogenesis remains unclear. This study aimed to explore biomarkers related to vitiligo through bioinformatic analysis. The microarray datasets GSE53146 and GSE65127 were downloaded from the Gene Expression Omnibus database. Firstly, differentially expressed genes (DEGs) in GSE53146 were screened, and then an enrichment analysis was performed. Secondly, the protein-protein interaction (PPI) network of DEGs was constructed using the STRING database, and the key genes were screened using the MCODE plugin in Cytoscape and verified using GSE65127. Finally, quantiseq was used to evaluate immune cell infiltration in vitiligo, then to observe the correlation between biomarkers and immune cells. In total, 544 DEGs were identified, including 342 upregulated and 202 downregulated genes. Gene Ontology (GO) enrichment showed that DEGs were related to inflammatory and immune responses, and Kyoto Encyclopedia of Genes and Genomes enrichment showed that DEGs were involved in many autoimmune diseases. In the PPI network, 7 key genes, CENPN, CKS2, PLK4, RRM2, TPX2, CCNA2, and CDC45 were identified by MCODE cluster and verified using the GSE65127 dataset. With an area under the curve (AUC) > 0.8 as the standard, 2 genes were screened, namely CKS2 and RRM2. Further immune infiltration analysis showed that M2 macrophages were involved in the pathogenesis of vitiligo, whereas CKS2 and RRM2 were both related to M2 macrophages. This study shows that CKS2 and RRM2 have potential as biomarkers of vitiligo and provides a theoretical basis for a better understanding of the pathogenesis of vitiligo.
... In consistence with the aforementioned explanation, interpretation of the RTPCR data demonstrated that Er 2 O 3 -NPs induced apoptosis of Hep-G2 cells resulted from the noticed concurrent upregulation of the apoptotic (p53 and Bax) genes and downregulation of the anti-apoptotic (Bcl2) gene expression levels. Moreover, arresting of cells at the G0/G1 phase triggers apoptosis 25 ...
Article
Full-text available
The remarkable physical and chemical characteristics of noble metal nanoparticles, such as high surface-to-volume ratio, broad optical properties, ease of assembly, surfactant and functional chemistry, have increased scientific interest in using erbium oxide nanoparticles (Er2O3-NPs) and other noble metal nanostructures in cancer treatment. However, the therapeutic effect of Er2O3-NPs on hepatic cancer cells has not been studied. Therefore, the current study was conducted to estimate the therapeutic potential of Er2O3-NPs on human hepatocellular carcinoma (Hep-G2) cells. Exposure to Er2O3-NPs for 72 h inhibited growth and caused death of Hep-G2 cells in a concentration dependent manner. High DNA damage and extra-production of intracellular reactive oxygen species (ROS) were induced by Er2O3-NPs in Hep-G2 cells. As determined by flow cytometry, Er2O3-NPs arrested Hep-G2 cell cycle at the G0/G1 phase and markedly increased the number of Hep-G2 cells in the apoptotic and necrotic phases. Moreover, Er2O3-NPs caused simultaneous marked increases in expression levels of apoptotic (p53 and Bax) genes and decreased level of anti-apoptotic Bcl2 gene expression level in Hep-G2 cells. Thus it is concluded that Er2O3-NPs inhibit proliferation and trigger apoptosis of Hep-G2 cells through the extra ROS generation causing high DNA damage induction and alterations of apoptotic genes. Thus it is recommended that further in vitro and in vivo studies be carried out to study the possibility of using Er2O3-NPs in the treatment of cancer.
... Correspondingly, the expressions of cyclin A and CDK2 exhibited upward trends, which were consistent with the result of the cell cycle arrested in the S phase. Additionally, CDK2 is the regulator of the G1-S transition [42]. Moreover, the formation of the cyclin D-CDK4/6 complex also engages in the transition of G1 to S phase [43]. ...
... Co spondingly, the expressions of cyclin A and CDK2 exhibited upward trends, which w consistent with the result of the cell cycle arrested in the S phase. Additionally, CDK the regulator of the G1-S transition [42]. Moreover, the formation of the cyclin D-CDK complex also engages in the transition of G1 to S phase [43]. ...
Article
Full-text available
The phenolic profiles, antioxidant activity, antiproliferative property and the underlying molecular mechanisms of cell apoptosis of Rhodiola rosea free phenolic (RFE) were analyzed in this work. Overall, Rhodiola rosea rhizome phenolic extract (RE) contained Rhodiola rosea rhizome free phenolic extract (RFE) and Rhodiola rosea rhizome bound phenolic extract (RBE). Compared with RBE, RFE contained higher phenolic contents and possessed stronger antioxidant activity. High-performance liquid chromatography (HPLC) results demonstrated that the main phenolics of were epigallocatechin (EGC), epigallocatechin gallate (EGCG), gallic acid (GA) and catechin. Gas chromatography–mass spectrometry (GC-MS) analysis found that Rhodiola rosea L. was rich in volatile phytochemicals. In addition, many types of vitamin E and a few kinds of carotenoids were found in Rhodiola rosea. In addition, the main compounds in RFE (GA, EGC, EGCG) and RFE all exhibited excellent antiproliferative activity, indicating the antiproliferative activity of RFE was partly attributed to the synergy effects of the main compounds. Further study confirmed that RFE could block 16.99% of HepG2 cells at S phase and induce 20.32% programmed cell death compared with the control group. Specifically, RFE dose-dependently induced cell apoptosis and cell cycle arrest via modulating the p53 signaling pathway including up-regulation of the expression of p53 and Bax while down-regulation of the Bcl-2, cyclin D1 and CDK4 levels. Therefore, RFE exhibited the potential of being developed as an auxiliary antioxidant and a therapeutic agent for cancer.
... We noted that YOH administration significantly attenuated PDGF-BB-induced CDK2 and PCNA upregulation in the VSMCs. A previous study revealed that CRISPR/Cas9-mediated knockout of CDK2 induced G0/G1 arrest and apoptosis in A375 melanocytes [40], and several other studies have shown a substantial downregulation of CDK2 in G0/G1-arrested VSMCs [39,41,42]. This, thus, explains why the expression of PCNA, a classic biomarker of proliferative cells, was suppressed in the YOH-treated MOVAS-1 cells. ...
Article
Full-text available
Yohimbine (YOH) has antiproliferative effects against breast cancer and pancreatic cancer ; however, its effects on vascular proliferative diseases such as atherosclerosis remain unknown. Accordingly, we investigated the inhibitory mechanisms of YOH in vascular smooth muscle cells (VSMCs) stimulated by platelet-derived growth factor (PDGF)-BB, a major mitogenic factor in vascular diseases. YOH (5-20 μM) suppressed PDGF-BB-stimulated a mouse VSMC line (MOVAS-1 cell) proliferation without inducing cytotoxicity. YOH also exhibited antimigratory effects and downregulated matrix metalloproteinase-2 and-9 expression in PDGF-BB-stimulated MOVAS-1 cells. It also promoted cell cycle arrest in the initial gap/first gap phase by upregulating p27Kip1 and p53 expression and reducing cyclin-dependent kinase 2 and proliferating cell nuclear antigen expression. We noted phospholipase C-γ1 (PLCγ1) but not ERK1/2, AKT, or p38 kinase phosphorylation attenuation in YOH-modulated PDGF-BB-propagated signaling pathways in the MOVAS-1 cells. Furthermore, YOH still inhibited PDGF-BB-induced cell proliferation and PLCγ1 phosphorylation in MOVAS-1 cells with α2B-adrenergic receptor knockdown. YOH (5 and 10 mg/kg) substantially suppressed neointimal hyperplasia in mice subjected to CCA ligation for 21 days. Overall, our results reveal that YOH attenuates PDGF-BB-stimulated VSMC proliferation and migration by downregulating a α2B-adrenergic receptor-independent PLCγ1 pathway and reduces neointimal formation in vivo. Therefore, YOH has potential for repurposing for treating atherosclerosis and other vascular proliferative diseases.