• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • No language data
  • Tagged with
  • 3
  • 3
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

SRC INHIBITORS POTENTIATE UCN-01-INDUCED APOPTOSIS IN HUMAN MULTIPLE MYELOMA CELLS THROUGH A RAS/RAF/MEK/ERK-DEPENDENT MECHANISM

Shah, Rena Ashwin 01 January 2007 (has links)
The goal of this study is to determine whether SKI-606, a Src/abl inhibitor, potentiates chk1 inhibitor UCN-01 to induce apoptosis in multiple myeloma cells, and what mechanism maybe involved. We found that the co-exposure of human myeloma cells (e.g., U266, RPMI8226, MM.1S and its dexamethasone-resistant counterparts MM.1R) to minimally toxic concentration of SKI-606 (e.g., 1-2 ƒÝM) and UCN-01 (e.g., 100-150 nM) resulted in dramatic increase in mitochondrial damage and apoptosis. In our previous reports, it has been well demonstrated that activation of Ras/Raf/MEK/ERK pathway represents a critical cytoprotective response in cells exposed to UCN-01. Moreover, Src is required to activate this pathway by growth factors and cytokines. To this end, we examined whether the Src inhibitor enhances UCN-01 lethality through interruption of Ras/Raf/MEK/ERK signaling cascade. Firstly, it was found that co-adminstration of SKI-606 markedly diminished ERK phosphorylation/activation induced by UCN-01, accompanied by an increase in cdc2 activation. Furthermore, myeloma cells with ecpotic expression of either active mutant Ras (Q61L) or constitutive active MEK1 were significantly resistant to combined treatment with SKI-606 and UCN-01, indicating Src inhibition acts upstream of Ras/Raf/MEK to potentiate UCN-01 lethality. Conversely, stable expression of dominant-negative mutant Ras (S17N) markedly sensitized myeloma cells to this combination regimen. Lastly, ectopic expression of kinase inactive (K297R) or dominant-negative (K296R/Y528F) mutant Src blocked ERK activation by UCN-01 and thereby sensitized myeloma cells to UCN-01. Together, these findings indicate that Src inhibitors act through a Ras/Raf/MEK-dependent mechanism to prevent ERK activation in UCN-01-treated cells, resulting in the synergistic induction of cell death.
2

HMG-CoA Reductase Inhibitors Act Synergistically with UCN-01 Through RAS Inhibition

Khanna, Payal 01 January 2006 (has links)
The primary objective of this study is to elucidate the mechanism by which the reagent UCN-01 induces apoptosis when administered to leukemia cells along with HmG-CoA reductase inhibitors, mainly the statins. In this study, we demonstrated that exposure of leukemia cell lines to lovastatin (20 uM, 18 hours) and UCN-01 (100 nM, 18 hours) resulted in mitochondria dysfunction, procaspase 3 and 9 cleavage, and PAW degradation along with marked cytochrome C release and apoptosis. Although similar molecular mechanisms have not yet been confirmed in other cancers, our hypothesis holds that enhanced apoptotic effects of UCN-01 are due in part to lovastatin's ability to block formation of geranylgeranylpyrophosphate and farnesylpyrophosphate by interfering with the rate-limiting step of the mevalonate pathway. Geranylgeranylpyrophosphate and farnesylpyrophosphate induce post-translational modifications in RAS that anchor the protein to the cell membrane so that it acts as a signal transducer to the nucleus, promoting cell proliferation.
3

Combinatorial Modulation of Multiple Signaling Pathways to Gain Therapeutic Response in Breast and Prostate Cell Carcinomas

Hawkins, William Tressel, II 01 January 2006 (has links)
Our laboratory is primarily interested in novel pharmacological intervention of cell proliferation and survival pathways expressed in various types of cancer. These cyto-protective pathways can be activated in response to growth factor stimulation, toxic insult and radiation. In our studies, we utilized novel drug combinations with and without radiation to enhance breast & prostate tumor cell death both in vitro and in vivo. Previous studies from our group have shown that UCN-01 and MEK1/2 inhibitors interact to cause tumor cell death in transformed cell lines in vitro. We extended this observation to an in vivo animal model system using the estrogen dependent breast cell carcinoma line MCF-7 and the estrogen independent breast cell carcinoma line MDA-MB-231. This drug combination was shown to profoundly reduce tumor cell proliferation in vivo and also exhibited the ability to significantly reduce ex-vivo tumor cell colony formation 30 days after cessation of the combination drug treatment. In addition, tumor cell death coincided with decreased ERK112 phosphorylation, reduced immunoreactivity of Ki67 and CD31. Overall, these studies demonstrate that UCN-01 and MEK112 inhibitors have the potential to suppress mammary tumor growth in vivo which is independent of p53 status, estrogen dependency, caspase-3 levels or oncogenic K-RAS expression. In our LnCap prostate carcinoma cell studies we demonstrated the impact of hCG and lovastatin in combination with ionizing radiation to radiosensitize and enhance tumor cell lethality. This enhancement was attributed to the hCG-induced activation of ERBB1 via a GPCR, MEK112 and metalloprotease dependent paracrine mechanism which was further enhanced by radiation. This enhanced cell killing effect was shown to involve prolonged activation of PARP1 which could be suppressed by inhibition of ERBB1, MEKl , PI3 kinase or PARP1. Therefore, the combination of hCG, lovastatin and radiation may represent a novel approach to kill prostate cancer cells and potential new therapy.

Page generated in 0.0272 seconds