scholarly journals RGNNV-induced cell cycle arrest at G1/S phase enhanced viral replication via p53-dependent pathway in GS cells

2018 ◽  
Vol 256 ◽  
pp. 142-152 ◽  
Author(s):  
Weijun Mai ◽  
Hongxiao Liu ◽  
Huiqing Chen ◽  
Yajing Zhou ◽  
Yan Chen
2021 ◽  
Author(s):  
Mingjun Su ◽  
Da Shi ◽  
Xiaoxu Xing ◽  
Shanshan Qi ◽  
Dan Yang ◽  
...  

Subversion of the host cell cycle to facilitate viral replication is a common feature of coronavirus infections. Coronavirus nucleocapsid (N) protein could modulate host cell cycle, but the mechanistic details remain largely unknown. Here, we investigated manipulation of porcine epidemic diarrhea virus (PEDV) N protein on cell cycle and its influence on viral replication. Results indicated that PEDV N-induced Vero E6 cell cycle arrest at S-phase, which promoted viral replication ( P < 0.05). S-phase arrest was dependent on N protein nuclear localization signal S 71 NWHFYYLGTGPHADLRYRT 90 and interaction between N protein and p53. In the nucleus, the binding of N protein to p53 maintained consistently high-level expression of p53, which activated p53-DREAM pathway. The key domain of the N protein interacting with p53 was revealed to be S 171 RGNSQNRGNNQGRGASQNRGGNN 194 (N S171-N194 ), in which G 183 RG 185 are core sites. N S171-N194 and G 183 RG 185 were essential for N-induced S-phase arrest. Moreover, small molecular drugs targeting the N S171-N194 domain of PEDV N protein were screened through molecular docking. Hyperoside could antagonize N protein-induced S-phase arrest by interfering with interaction between N protein and p53 and inhibit viral replication ( P < 0.05). The above experiments were also validated in porcine intestinal cells, and resulting data were in line with that of Vero E6 cells. Therefore, these results revealed that PEDV N protein interacted with p53 to activate p53-DREAM pathway, and subsequently induced S-phase arrest to create a favorable environment for virus replication. These findings provided new insight into the PEDV-host interaction and the design of novel antiviral strategies against PEDV.


2012 ◽  
Vol 33 (12) ◽  
pp. 1500-1505 ◽  
Author(s):  
Yu Sun ◽  
Shusheng Tang ◽  
Xi Jin ◽  
Chaoming Zhang ◽  
Wenxia Zhao ◽  
...  

MedChemComm ◽  
2016 ◽  
Vol 7 (6) ◽  
pp. 1132-1137 ◽  
Author(s):  
Hua-Hong Zou ◽  
Jun-Guang Wei ◽  
Xiao-Huan Qin ◽  
Shun-Gui Mo ◽  
Qi-Pin Qin ◽  
...  

Two metallo-complexes inhibited telomerase by interacting with c-myc G4-DNA and induced cell cycle arrest at the S phase.


1998 ◽  
Vol 241 (2) ◽  
pp. 340-351 ◽  
Author(s):  
Nurit Kleinberger-Doron ◽  
Noa Shelah ◽  
Ricardo Capone ◽  
Aviv Gazit ◽  
Alexander Levitzki

2018 ◽  
Vol 70 (1) ◽  
pp. 6-13 ◽  
Author(s):  
Artur Beberok ◽  
Dorota Wrześniok ◽  
Aldona Minecka ◽  
Jakub Rok ◽  
Marcin Delijewski ◽  
...  

Metallomics ◽  
2014 ◽  
Vol 6 (5) ◽  
pp. 1014 ◽  
Author(s):  
Sabine H. van Rijt ◽  
Isolda Romero-Canelón ◽  
Ying Fu ◽  
Steve D. Shnyder ◽  
Peter J. Sadler

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4290-4290
Author(s):  
Ina Fabian ◽  
Debby Haite ◽  
Avital Levitov ◽  
Drora Halperin ◽  
Itamar Shalit

Abstract We previously reported that the fluoroquinolone moxifloxacin (MXF) inhibits NF-kB, mitogen-activated protein kinase activation and the synthesis of proinflammatory cytokines in activated human monocytic cells (AAC48:1974,2004). Since MXF acts on topoisomerase II (Topo II) in mammalian cells, we investigated its effect in combination with another Topo II inhibitor, VP-16, on cell proliferation (by the MTT method), cell cycle, caspase-3 activity and proinflammatory cytokine release in THP-1 and Jurkat cells. THP-1 cells were incubated for 24 h with 0.5–3 μg/ml VP-16 in the presence or absence of 5–20 μg/ml MXF. VP-16 induced a dose dependent decrease in cell proliferation. An additional 2.5-and 1.6-fold decrease in cell proliferation was observed upon incubation of the cells with 0.5 or 1 μg/ml VP-16 and 20 μg/ml MXF, respectively (up to 69% inhibition). To further elucidate the mechanism of the antiproliferative activity of MXF, its effect on cell cycle progression was investigated. In control cultures 1%, 45%,18% and 36% of cells were in G0, G1, S and G2/M phases at 24 h, respectively. In contrast, in cultures treated with 1 μg/ml VP-16 and VP-16+ 20 μg/ml MXF, the number of cells in G1 decreased to 5.4 and 6.5%, respectively, while the number of cells in S phase increased to 25.5 and 42%, respectively and the number of cells in G2/M cells increased to 60 and 44%, respectively. These data provide evidence for S-G2/M cell cycle arrest induced by VP-16 and that addition of MXF shifted the S-G2/M arrest more towards the S phase. Since the antiproliferative effects of MXF could also be attributed to apoptotic cell death in addition to cell cycle arrest, we investigated the effect of the drugs on apoptosis. Using the fluorogenic assay for caspse-3 activity, we show that incubation of THP-1 cells for 6 h with 1.5 μg/ml VP-16 resulted in 630±120 unit/50μg protein of caspase-3 activity while the combination of 1.5 μg/ml VP-16 and 20 μg/ml MXF enhanced caspase-3 activity up to 1700±340 units/50μg protein (vs.233±107 in control cells), indicating that MXF synergises with VP-16 in activation of caspase-3. In Jurkat cells, the addition of 0.5 or 1 μg/ml VP-16, did not affect cell proliferation while in the presence of 20 μg/ml MXF and 1 μg/ml VP-16 there was a 62% decrease in cell proliferation (p&lt;0.05). Exposure of Jurkat cells to 3 μg/ml VP-16 alone resulted in 504±114 units/50μg protein of caspase-3 activity and the addition of 20μg/ml MXF enhanced caspase-3 activity up to 1676± 259 units/50μg protein (vs 226±113 units/50μg protein in control cells). We further examined pro-inflammatory cytokine secretion upon stimulation of THP-1 cells with VP-16, MXF or their combination. VP-16 alone at 3 μg/ml increased IL-8 and TNF-α secretion from THP-1 cells by 2.5 and 1.8-fold respectively. Addition of MXF (5–20 μg/ml) inhibited the two cytokines secretion by 72–77% and 58–72%, respectively. The above combined data indicate that MXF, at clinically attainable concentrations, demonstrates pronounced synergistic effect with VP-16 as an anti-proliferative agent mainly by enhancing caspase-3 activity and apoptosis. At the same time MXF inhibits the pro-inflammatory effects conferred by VP-16 in the tumor cells studied. The clinical significance of the above anti-proliferative and anti-inflammatory effects of MXF in combination with VP-16 should be further investigated in animal models.


Sign in / Sign up

Export Citation Format

Share Document