scholarly journals Overexpression of dominant negative PARP interferes with tumor formation of HeLa cells in nude mice: Evidence for increased tumor cell apoptosis in vivo

Oncogene ◽  
1999 ◽  
Vol 18 (50) ◽  
pp. 7010-7015 ◽  
Author(s):  
Margrit-Airin Hans ◽  
Marcus Müller ◽  
Mirella Meyer-Ficca ◽  
Alexander Bürkle ◽  
Jan-Heiner Küpper
MedChemComm ◽  
2018 ◽  
Vol 9 (10) ◽  
pp. 1663-1672 ◽  
Author(s):  
Kun Hu ◽  
Chensi Liu ◽  
Jingui Li ◽  
Fupei Liang

Three Cu(ii) complexes (C1–C3) were designed and synthesized.C3, in particular, having a ligand derived from benzocaine, exhibited greater selectivity toward HeLa cells, arrested cell cycles, and promoted tumor cell apoptosis.


Science ◽  
1991 ◽  
Vol 254 (5029) ◽  
pp. 293-295
Author(s):  
SF Dowdy ◽  
CL Fasching ◽  
D Araujo ◽  
KM Lai ◽  
E Livanos ◽  
...  

Wilms tumor has been associated with genomic alterations at both the 11p13 and 11p15 regions. To differentiate between the involvement of these two loci, a chromosome 11 was constructed that had one or the other region deleted, and this chromosome was introduced into the tumorigenic Wilms tumor cell line G401. When assayed for tumor-forming activity in nude mice, the 11p13-deleted, but not the 11p15.5-p14.1-deleted chromosome, retained its ability to suppress tumor formation. These results provide in vivo functional evidence for the existence of a second genetic locus (WT2) involved in suppressing the tumorigenic phenotype of Wilms tumor.


2021 ◽  
Author(s):  
Guanyou Lin ◽  
Qingxin Mu ◽  
Richard Revia ◽  
Zachary Stephen ◽  
Mike Jeon ◽  
...  

PSA was conjugated onto IONP by first covalently linking Neu onto a 10 nm IONP core and then attaching PSA to neu via biotin–avidin interaction. When applied in vivo, IONP-Neu-PSA-Cy5.5 produced persistent near-IR signal specifically for monitoring tumor apoptosis.


2013 ◽  
Vol 34 (5) ◽  
pp. 1105-1114 ◽  
Author(s):  
Xiaolin Hu ◽  
Mary A. Zimmerman ◽  
Kankana Bardhan ◽  
Dafeng Yang ◽  
Jennifer L. Waller ◽  
...  

2016 ◽  
Vol 52 (40) ◽  
pp. 6693-6696 ◽  
Author(s):  
Xiaojun Liu ◽  
Bo Hu ◽  
Ranran Cheng ◽  
Fanpeng Kong ◽  
Xiaohong Pan ◽  
...  

Simultaneous fluorescence imaging of selenol and hydrogen peroxide with 5-FAM-peptide-AuNPs and QCy7-H2O2 was realized during tumor cell apoptosis induced by selenite.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2241-2241
Author(s):  
Shiva Keshava ◽  
L. Vijaya Mohan Rao ◽  
Usha R. Pendurthi

Abstract Our recent studies showed that endothelial cell protein C receptor (EPCR) functions as a crucial negative regulator of cancer progression in malignant pleural mesothelioma (MPM) (Keshava et al., Cancer Research 73: 3963-73, 2013). In these studies, the introduction of EPCR expression to aggressive MPM cells lacking EPCR completely attenuated their tumorigenicity whereas the knock-down of EPCR expression in non-aggressive MPM cells that constitutively express EPCR increased their tumorigenicity. This study also revealed that EPCR in MPM cells promotes tumor cell apoptosis in vivo. These data are quite intriguing as EPCR-mediated cell signaling typically activates cell survival and anti-apoptotic pathways and is shown to promote tumor growth in breast and lung cancers. The aim of the present study is to understand the mechanism(s) by which EPCR promotes tumor cell apoptosis in MPM. Evaluation of thoracic fluids obtained from nude mice implanted with MPM cells orthotopically showed a significant increase in the levels of mTNFα and mIFNγ in mice implanted with REN (+EPCR) cells compared to mice implanted with parental REN MPM or REN cells transfected with the vector control [REN(z)]. Analysis of tumor cell apoptosis of cultured cells by TUNEL assay showed that REN (+EPCR) cells were highly susceptible to TNFα+IFNγ-induced cell death compared to parental REN or REN(z) cells (80% vs. 20% cell death at 72 h). In agreement with the concept that EPCR in MPM cells promotes the apoptosis, M9K MPM cells that constitutively express EPCR (non-aggressive MPM cell line) were highly susceptible to TNFα+IFNγ-induced cell death; the knock-down of EPCR in these cells rendered them resistant to cytokine-induced cell death. EPCR-induced tumor cell apoptosis in MPM cells was not dependent on activated protein C (APC). Deletion of the cytoplasmic tail of EPCR failed to diminish its cytotoxic effect. Analysis of REN and REN(+EPCR) tumor cell extracts using PathScan intracellular signaling antibody array kit (Cell Signaling Technology, MA) and immunoblot analysis with specific antibodies against proteins involved in apoptotic pathways revealed that EPCR expression in MPM cells, in comparison to MPM cells lacking EPCR, resulted in a sustained cleavage of poly ADP ribose polymerase (PARP) and an enhanced activation of caspase 3 in response to TNFα+ IFNγ treatment. In addition, REN(+EPCR) cells exhibited an increased activation of caspase 8 and a decreased phosphorylation of anti-apoptotic protein BAD. Further signaling studies showed a delayed activation of NF-kB, suppression of activation of STAT3 and AKT in REN(+EPCR) cells compared to REN cells in response to TNFα+ IFNγ treatment. In addition, EPCR expression suppressed the constitutive activation of the proline-rich AKT substrate, 40 kDa (PRAS40). Overall, our studies indicate that EPCR in MPM cells controls the progression of MPM by exerting its effect at multiple levels. First, EPCR in MPM cells elicits elaboration of cytokines TNFα and IFNγ in vivo and then renders the MPM cells susceptible to TNFα+ IFNγ-induced cell death. The EPCR-mediated cytotoxic effect in MPM cells is independent of APC and the cytoplasmic tail and involves the suppression of STAT3 and AKT activation. At present, it is unclear exactly how EPCR initiates the activation of the apoptotic pathway in MPM cells and whether it involves the interaction of EPCR with the endogenous ligand(s) specific to MPM cells. Studies are underway to identify EPCR interacting proteins in MPM cells. Disclosures No relevant conflicts of interest to declare.


2009 ◽  
Vol 29 (8) ◽  
pp. 2243-2253 ◽  
Author(s):  
Jun Yang ◽  
Afshan Ahmed ◽  
Evon Poon ◽  
Nina Perusinghe ◽  
Alexis de Haven Brandon ◽  
...  

ABSTRACT The p53 tumor suppressor protein negatively regulates hypoxia-inducible factor 1α (HIF-1α). Here, we show that induction of p53 by the small-molecule RITA (reactivation of p53 and induction of tumor cell apoptosis) [2,5-bis(5-hydroxymethyl-2-thienyl) furan] (NSC-652287) inhibits HIF-1α and vascular endothelial growth factor expression in vivo and induces significant tumor cell apoptosis in normoxia and hypoxia in p53-positive cells. RITA has been proposed to stabilize p53 by inhibiting the p53-HDM2 interaction. However, induction of p53 alone was insufficient to block HIF-1α induced in hypoxia and has previously been shown to require additional stimuli, such as DNA damage. Here, we identify a new mechanism of action for RITA: RITA activates a DNA damage response, resulting in phosphorylation of p53 and γH2AX in vivo. Unlike other DNA damage response-inducing agents, RITA treatment of cells induced a p53-dependent increase in phosphorylation of the α subunit of eukaryotic initiation factor 2, requiring PKR-like endoplasmic reticulum kinase activity, and led to the subsequent downregulation of HIF-1α and p53 target proteins, including HDM2 and p21. Through the identification of a new mechanism of action for RITA, our study uncovers a novel link between the DNA damage response-p53 pathway and the protein translational machinery.


Sign in / Sign up

Export Citation Format

Share Document