360 BIO, the GSK3 beta blocker, is a potent inhibitor of cell proliferation and inducer of cell death of cervical carcinoma and rhabdomyosarcoma tumor cells

2010 ◽  
Vol 8 (7) ◽  
pp. 114
Author(s):  
M. Majka ◽  
W. Bobela ◽  
K. Miekus
2020 ◽  
Vol 13 (7) ◽  
pp. 156 ◽  
Author(s):  
Angel Escamilla-Ramírez ◽  
Rosa A. Castillo-Rodríguez ◽  
Sergio Zavala-Vega ◽  
Dolores Jimenez-Farfan ◽  
Isabel Anaya-Rubio ◽  
...  

Glioma is the most frequent and aggressive type of brain neoplasm, being anaplastic astrocytoma (AA) and glioblastoma multiforme (GBM), its most malignant forms. The survival rate in patients with these neoplasms is 15 months after diagnosis, despite a diversity of treatments, including surgery, radiation, chemotherapy, and immunotherapy. The resistance of GBM to various therapies is due to a highly mutated genome; these genetic changes induce a de-regulation of several signaling pathways and result in higher cell proliferation rates, angiogenesis, invasion, and a marked resistance to apoptosis; this latter trait is a hallmark of highly invasive tumor cells, such as glioma cells. Due to a defective apoptosis in gliomas, induced autophagic death can be an alternative to remove tumor cells. Paradoxically, however, autophagy in cancer can promote either a cell death or survival. Modulating the autophagic pathway as a death mechanism for cancer cells has prompted the use of both inhibitors and autophagy inducers. The autophagic process, either as a cancer suppressing or inducing mechanism in high-grade gliomas is discussed in this review, along with therapeutic approaches to inhibit or induce autophagy in pre-clinical and clinical studies, aiming to increase the efficiency of conventional treatments to remove glioma neoplastic cells.


PeerJ ◽  
2017 ◽  
Vol 5 ◽  
pp. e4033 ◽  
Author(s):  
Tove Kirkegaard ◽  
Mikail Gögenur ◽  
Ismail Gögenur

Background The perioperative period is important for patient outcome. Colorectal cancer surgery can lead to metastatic disease due to release of disseminated tumor cells and the induction of surgical stress response. To explore the overall effects on surgically-induced changes in serum composition, in vitro model systems are useful. Methods A systematic search in PubMed and EMBASE was performed to identify studies describing in vitro models used to investigate cancer cell growth/proliferation, cell migration, cell invasion and cell death of serum taken pre- and postoperatively from patients undergoing colorectal tumor resection. Results Two authors (MG and TK) independently reviewed 984 studies and identified five studies, which fulfilled the inclusion criteria. Disagreements were solved by discussion. All studies investigated cell proliferation and cell invasion, whereas three studies investigated cell migration, and only one study investigated cell death/apoptosis. One study investigated postoperative peritoneal infection due to anastomotic leak, one study investigated mode of anesthesia (general anesthesia with volatile or intravenous anesthetics), and one study investigated preoperative intervention with granulocyte macrophage colony stimulating factor (GMCSF). In all studies an increased proliferation, cell migration and invasion was demonstrated after surgery. Anesthetics with propofol and intervention with GMCSF significantly reduced postoperative cell proliferation, whereas peritoneal infection enhanced the invasive capability of tumor cells. Conclusion This study suggests that in vitro cell models are useful and reliable tools to explore the effect of surgery on colorectal cancer cell proliferation and metastatic ability. The models should therefore be considered as additional tests to investigate the effects of perioperative interventions.


1993 ◽  
Vol 39 (11) ◽  
pp. 2375-2385 ◽  
Author(s):  
R Schmandt ◽  
G B Mills

Abstract Many of the genes encoding growth factors, growth factor receptors, enzymes, and other effector molecules that regulate normal cell growth are designated protooncogenes. Oncogenes, those genes associated with cellular transformation, differ from their protooncogenic progenitors by being mutated, overexpressed, or expressed at inappropriate times or locations in the cell. One of the activities of growth factors is to prime cells to undergo programmed cell death, which is characterized by a series of morphologic changes called apoptosis. In normal cells, specific mediators must be activated or suppressed to bypass programmed cell death. In tumor cells, either the pathways leading to apoptosis are not functional or the mediators that normally "rescue" cells from this fate are overexpressed or constitutively activated. In addition to the biochemical pathways that drive cell division, there are others that limit cell proliferation; these, designated tumor suppressors, anti-oncogenes, or recessive oncogenes, must be inactivated in normal cells to allow passage through the cell cycle and cell proliferation. In contrast to oncogenes, which are overexpressed or activated in tumors, tumor-suppressor genes are frequently inactivated in tumor cells, either by mutation or deletion. Thus, in normal cells a series of checks and balances must be overcome to allow initiation and continuation of cell division. In tumors, these processes are aberrant, resulting in increased rates of cell division, increases in the proportion of cells in the cell cycle, or increased survival of activated cells. Therefore, tumor cells frequently accumulate genomic alterations, which may result in the activation of a particular array of oncogenes, the inactivation of specific tumor-suppressor genes, and the bypassing of programmed cell death. Trials of antitumor agents that act by exploiting the overexpression of oncogenes in tumors and of the biochemical pathways by which they mediate cell proliferation are currently underway.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3844-3844
Author(s):  
Haiming Chen ◽  
Rui Hao ◽  
Eric Sanchez ◽  
Jing Shen ◽  
Mingjie Li ◽  
...  

Abstract Abstract 3844 Poster Board III-780 Pterostilbene (3,5-Dimethoxy-4'-hydroxy-trans-stilbene) is a key compound found predominantly in blueberries and grapes. Pterostilbene has been shown to exhibit potential anti-cancer characteristics, anti-hypercholesterolemia and anti-hypertriglyceridemia properties. However, the mechanism of anti-cancer effects has not been elucidated and this compound has not been previously evaluated in multiple myeloma (MM). In this study, we first examined the HDAC binding ability of pterostilbene in the RPMI8226 multiple myeloma (MM) cell line and 293 HEK cells by using a novel HDAC screening assay. Briefly, RPMI8226 cells or 293 HEK cells were lysed with M-PER supplemented with protease and phosphatase inhibitors. The lysates were diluted and incubated with pterostilbene in concentrations ranging from 1 to 200 μM or without drug. Proteolysis was performed using thermolysin and aliquots were removed every 5 minutes and western blot analysis completed using antibodies against different HDACs or GAPDH. The results showed that pterostilbene prevents specifically HDAC1 digestion by thermolysin in both RPMI8226 and 293 cells. Next, we examined H4 acetylation of lysine residues in RPMI8226 cells following treatment with pterostilbene using western blot analysis. Increases in histone acetylation in RPMI8226 cells following exposure to pterostilbene treatment occurred in a concentration-dependent manner. Specifically, 1 to10μM of the pterostilbene markedly induced histone acetylation in RPMI8226 cells within 24 hrs. We also analyzed pterostilbene's effect on MM tumor cell proliferation using the MTS assay and apoptosis with flow cytometric analysis following Annexin V staining, in cells from the RPMI8226 and MM1S MM cell lines and freshly obtained tumor cells from MM patients. Pterostilbene alone showed a 50% growth inhibition (IC50) of cells from the RPMI8226 and MM1S lines as well as fresh bone marrow tumor cells from MM patients treated for 48 hours at approximately the same concentration (5 μM) as showing anti-MM effects in the MTS assay. Notably, the combination of pterostilbene and melphalan or the proteasome inhibitor bortezomib showed markedly increased inhibition of MM cell growth compared to treatment of the cells with the drugs alone. Since this effect may have resulted from decreased cell proliferation due to cell cycle arrest or increased cell death, we further determined apoptosis in cells from RPMI8226, U266, and MM1S cell lines as well as fresh tumor cells from MM patients following treatment with pterostilbene. Our data demonstrated that pterostilbene induced apoptotic cell death in a concentration dependent fashion. We also evaluated the combination of this compound with bortezomib (1-6 nM) and showed marked increases in apoptosis using this combination compared to either drug alone. These studies establish pterostilbene as a novel HDACi with potent anti-MM activity alone and also show its ability to enhance the anti-MM effects of chemotherapeutic agents and bortezomib. Currently, we are evaluating pterostilbene alone and in combination treatments using our SCID-hu murine models of human MM. Disclosures: No relevant conflicts of interest to declare.


Planta Medica ◽  
2012 ◽  
Vol 78 (11) ◽  
Author(s):  
HM Chen ◽  
PH Wang ◽  
SS Chen ◽  
CC Wen ◽  
YH Chen ◽  
...  

Author(s):  
Morganna C. Lima ◽  
Elisa A. N. Azevedo ◽  
Clarice N. L. de Morais ◽  
Larissa I. O. de Sousa ◽  
Bruno M. Carvalho ◽  
...  

Background: Zika virus is an emerging arbovirus of global importance. ZIKV infection is associated with a range of neurological complications such as the Congenital Zika Syndrome and Guillain Barré Syndrome. Despite the magnitude of recent outbreaks, there is no specific therapy to prevent or to alleviate disease pathology. Objective: To investigate the role of P-MAPA immunomodulator in Zika-infected THP-1 cells. Methods: THP-1 cells were subjected at Zika virus infection (Multiplicity of Infection = 0.5) followed by treatment with P-MAPA for until 96 hours post-infection. After that, the cell death was analyzed by annexin+/ PI+ and caspase 3/ 7+ staining by flow cytometry. In addition, the virus replication and cell proliferation were accessed by RT-qPCR and Ki67 staining, respectively. Results: We demonstrate that P-MAPA in vitro treatment significantly reduces Zika virus-induced cell death and caspase-3/7 activation on THP-1 infected cells, albeit it has no role in virus replication and cell proliferation. Conclusions: Our study reveals that P-MAPA seems to be a satisfactory alternative to inhibits the effects of Zika virus infection in mammalian cells.


Author(s):  
Nivedita Bhardwaj ◽  
Nancy Tripathi ◽  
Bharat Goel ◽  
Shreyans K. Jain

: During cancer progression, the unrestricted proliferation of cells is supported by the impaired cell death response provoked by certain oncogenes. Both autophagy and apoptosis are the signaling pathways of cell death, which are targeted for cancer treatment. Defects in apoptosis result in reduced cell death and ultimately tumor progression. The tumor cells lacking apoptosis phenomena are killed by ROS- mediated autophagy. The autophagic programmed cell death requires apoptosis protein for inhibiting tumor growth; thus, the interconnection between these two pathways determines the fate of a cell. The cross-regulation of autophagy and apoptosis is an important aspect to modulate autophagy, apoptosis and to sensibilise apoptosis-resistant tumor cells under metabolic stress and might be a rational approach for drug designing strategy for the treatment of cancer. Numerous proteins involved in autophagy have been investigated as the druggable target for anticancer therapy. Several compounds of natural origin have been reported, to control autophagy activity through the PI3K/Akt/mTOR key pathway. Diosgenin, a steroidal sapogenin has emerged as a potential candidate for cancer treatment. It induces ROS-mediated autophagy, inhibits PI3K/Akt/mTOR pathway, and produces cytotoxicity selectively in cancer cells. This review aims to focus on optimal strategies using diosgenin to induce apoptosis by modulating the pathways involved in autophagy regulation and its potential implication in the treatment of various cancer. The discussion has been extended to the medicinal chemistry of semi-synthetic derivatives of diosgenin exhibiting anticancer activity.


2019 ◽  
Vol 19 (5) ◽  
pp. 610-619 ◽  
Author(s):  
Xue-Qing Zhang ◽  
Lu-Ting Yu ◽  
Pei Du ◽  
Tian-Qi Yin ◽  
Zhi-Yuan Zhang ◽  
...  

Background:Regenerating islet-derived gene family member 4 (Reg4), a well-investigated growth factor in the regenerative pancreas, has recently been reported to be highly associated with a majority of gastrointestinal cancers. Pathological hyper-expression or artificial over-expression of Reg4 causes acceleration of tumor growth, migration, and resistance to chemotherapeutic 5-Fluorouracil (5-FU). Until now, no method has been successfully established for eliminating the effects of Reg4 protein.Methods:This study reports the production of an engineered immunoglobin, a single-chain variable fragment (scFv-Reg4), to specifically bind Reg4 and block the bioactivity. The complementary-determining regions (CDRs) against Reg4 were assigned using MOE and ZDOCK servers. The binding affinity (KD) was determined by bio-layer interferometry (BLI). MKN45 and AGS cell proliferation was determined by Thiazolyl blue tetrazolium bromide (MTT) method and the cell apoptosis was detected by flow cytometry assay.Results:The KD of scFv-Reg4 to Reg4 was determined to be 1.91×10-8. In MKN45 and AGS cell lines, scFv- Reg4 depressed Reg4-stimulated cell proliferation and the inhibitory rates were 27.7±1.5% and 17.3±2.6%, respectively. Furthermore, scFv significantly enhanced 5-FU-induced cell death, from 23.0±1.0% to 28.4±1.2% in MKN45 and 28.2±0.7% to 36.6±0.6% in AGS cells. Treatment with scFv alone could lyse cancer cells to a certain extent, but no significance has been observed.Conclusion:The single-chain antibody (scFv-Reg4) significantly inhibited gastric cancer cell proliferation and synergistically enhanced the lethal effect of 5-FU. Thus, traditional chemo-/radio- therapeutics supplemented with scFv-Reg4 may provide advances in the strategy for gastrointestinal cancer treatment.


Sign in / Sign up

Export Citation Format

Share Document