scholarly journals Small interfering RNAs targeting cyclin D1 and cyclin D2 enhance the cytotoxicity of chemotherapeutic agents in mantle cell lymphoma cell lines

2011 ◽  
Vol 52 (11) ◽  
pp. 2148-2154 ◽  
Author(s):  
Katrin Tiemann ◽  
Jessica V. Alluin ◽  
Anja Honegger ◽  
Pritsana Chomchan ◽  
Shikha Gaur ◽  
...  
2002 ◽  
Vol 199 (1) ◽  
pp. 84-89 ◽  
Author(s):  
Raymond Lai ◽  
George Z Rassidakis ◽  
L Jeffrey Medeiros ◽  
Vasiliki Leventaki ◽  
Micheal Keating ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2515-2515 ◽  
Author(s):  
Heather Gilbert ◽  
John Cumming ◽  
Josef T. Prchal

Abstract Abstract 2515 Poster Board II-492 Mantle cell lymphoma is a well defined subtype of B-cell non-Hodgkin lymphoma characterized by a translocation that juxtaposes the BCL1 gene on chromosome 11q13 (which encodes cyclin D1) next to the immunoglobulin heavy chain gene promoter on chromosome 14q32. The result is constitutive overexpression of cyclin D1 (CD1) resulting in deregulation of the cell cycle and activation of cell survival mechanisms. There are no “standard” treatments for MCL. Despite response rates to many chemotherapy regimens of 50% to 70%, the disease typically progresses after treatment, with a median survival time of approximately 3-4 years. Mantle cell lymphoma represents a small portion of malignant lymphomas, but it accounts for a disproportionately large percentage of lymphoma-related mortality. Novel therapeutic approaches are needed. In 2007, Nurtjaha-Tjendraputra described how iron chelation causes post-translational degradation of cyclin D1 via von Hippel Lindau protein-independent ubiquitinization and subsequent proteasomal degradation (1). Nurtjaha-Tjendraputra demonstrated that iron chelation inhibits cell cycle progression and induces apoptosis via proteosomal degradation of cyclin D1 in various cell lines, including breast cancer, renal carcinoma, neuroepithelioma and melanoma. Our preliminary data show similar findings in mantle cell lymphoma. To establish whether iron chelation can selectively inhibit and promote apoptosis in mantle cell derived cell lines, the human MCL cell lines Jeko-1, Mino, Granta and Hb-12; the Diffuse Large B cell lymphoma line SUDHL-6; and the Burkitt's Lymphoma lines BL-41 and DG75 were grown with media only, with two different iron chelators (deferoxamine (DFO) and deferasirox) at various concentrations (10, 20, 40, 100 and 250 μM), and with DMSO as an appropriate vehicle control. Cells were harvested at 24, 48 and 72 hours. For detection of apoptotic cells, cell-surface staining was performed with FITC-labeled anti–Annexin V antibody and PI (BD Pharmingen, San Diego, CA). Cell growth was analyzed using the Promega MTS cytotoxicity assay. CD1 protein levels were assessed using standard Western blot techniques. At 24, 48 and 72 hours of incubation with iron chelators, the mantle cell lymphoma cell lines showed significantly increased rates of apoptosis compared to the non-mantle cell lymphoma cell lines (p<0.0001 for all time points). DFO and deferasirox inhibted cell growth with an IC50 of 18 and 12 μM respectively. All of the mantle cell lines had measurable cyclin D1 levels at baseline. None of the non-mantle cell lines expressed baseline measurable cyclin D1. In the mantle cell lines, cyclin D1 protein levels were no longer apparent on western blot after 24 hours of incubation with chelation. We then added ferrous ammonium sulfate (FAS) to DFO in a 1:1 molarity ratio and to deferasirox in a 2:1 ratio, and then treated the same lymphoma cell lines with the FAS/chelator mixture and with FAS alone for 72 hours. Adding iron to the chelators completely negated all the pro-apoptotic effects that were seen with iron chelation treatment. Treating with FAS alone had no effect on cell growth or apoptosis. Iron chelation therapy with both DFO and deferasirox results in decreased cell growth, increased cellular apoptosis, and decreased cyclin D1 protein levels in vitro in mantle cell lymphoma. The cytotoxic effects are prevented by coincubation with ferrous ammonium citrate, confirming that the effects are due to iron depletion. Proposed future research includes further defining the molecular basis of iron chelation effects; studying these therapies in combination with other cancer treatments both in vitro and in vivo; and studying iron chelation therapy in mantle cell lymphoma patients. 1. Nurtjahja-Tjendraputra, E., D. Fu, et al. (2007). “Iron chelation regulates cyclin D1 expression via the proteasome: a link to iron deficiency-mediated growth suppression.” Blood109(9): 4045–54. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 30 (8) ◽  
pp. 923-934 ◽  
Author(s):  
Jordi Camps ◽  
Itziar Salaverria ◽  
Maria J. Garcia ◽  
Esther Prat ◽  
Sílvia Beà ◽  
...  

2018 ◽  
Vol 13 (2) ◽  
pp. 235-245 ◽  
Author(s):  
Valentina Restelli ◽  
Monica Lupi ◽  
Micaela Vagni ◽  
Rosaria Chilà ◽  
Francesco Bertoni ◽  
...  

Mobile DNA ◽  
2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Witold Tatkiewicz ◽  
James Dickie ◽  
Franchesca Bedford ◽  
Alexander Jones ◽  
Mark Atkin ◽  
...  

Abstract Background The cell-surface attachment protein (Env) of the HERV-K(HML-2) lineage of endogenous retroviruses is a potentially attractive tumour-associated antigen for anti-cancer immunotherapy. The human genome contains around 100 integrated copies (called proviruses or loci) of the HERV-K(HML-2) virus and we argue that it is important for therapy development to know which and how many of these contribute to protein expression, and how this varies across tissues. We measured relative provirus expression in HERV-K(HML-2), using enriched RNA-Seq analysis with both short- and long-read sequencing, in three Mantle Cell Lymphoma cell lines (JVM2, Granta519 and REC1). We also confirmed expression of the Env protein in two of our cell lines using Western blotting, and analysed provirus expression data from all other relevant published studies. Results Firstly, in both our and other reanalysed studies, approximately 10% of the transcripts mapping to HERV-K(HML-2) came from Env-encoding proviruses. Secondly, in one cell line the majority of the protein expression appears to come from one provirus (12q14.1). Thirdly, we find a strong tissue-specific pattern of provirus expression. Conclusions A possible dependency of Env expression on a single provirus, combined with the earlier observation that this provirus is not present in all individuals and a general pattern of tissue-specific expression among proviruses, has serious implications for future HERV-K(HML-2)-targeted immunotherapy. Further research into HERV-K(HML-2) as a possible tumour-associated antigen in blood cancers requires a more targeted, proteome-based, screening protocol that will consider these polymorphisms within HERV-K(HML-2). We include a plan (and necessary alignments) for such work.


2012 ◽  
Vol 40 (8) ◽  
pp. 646-656.e2 ◽  
Author(s):  
Ana Lopez-Campistrous ◽  
Xiaohua Song ◽  
Adam J. Schrier ◽  
Paul A. Wender ◽  
Nancy A. Dower ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1816-1816
Author(s):  
Robert W Chen ◽  
Katrin Tiemann ◽  
Jessica Alluin ◽  
Stephen Forman ◽  
John Rossi

Abstract Abstract 1816 Introduction: Mantle cell lymphoma is an aggressive B cell neoplasm with a median survival of 3 years. Cyclin D1 overexpression is the genetic hallmark of MCL and regulates cell cycle progression. However, the significance of cyclin D1 in the pathogenesis and treatment of MCL still remains to be defined. The aim of this study is to determine whether down regulation of cyclin D1 with siRNA will lead to enhanced therapeutic effect of chemotherapy in MCL. We used siRNA technology in three well characterized MCL cell lines, and tested traditional chemotherapy agents (doxorubicin and etoposide) as a model system. Material and Methods: We designed three different siRNA targeting cyclin D1 (si-224, 391, 778), one siRNA against cyclin D2 (si-D2), and a dual targeting siRNA against both cyclin D1 and D2 (si-D1/D2). The siRNAs used were 27 mer asymetric duplexes with a 2nd 3′ overhang. Granta-519 cells were transfected by lipofection (Lipofectamin RNAimax, Invitrogen), Z-138 and Jeko-1 cells were transfected with electroporation (BioRad). Western Blot analysis and real time PCR were performed to examine the down regulatory efficiency of the siRNAs on cyclin D1 mRNA and protein. Chemotherapeutics doxorubicin and etoposide were tested for enhancement of cytotoxicity by siRNA. The effect on cell viability of cyclin D1 reduction in combination with chemotherapeutics was analyzed by MTS assay. Results: We achieved cyclin D1 mRNA and protein down regulation in all 3 MCL cell lines, although the efficiency of knockdown varied among the siRNAs and the cell lines of interests. (Table 1) Si-224 has the best activity in Granta-519 while si-778 has the best activity in Jeko-1. We determined the cytotoxic effect of chemotherapy alone as well as in combination with siRNAs by MTS assays. The combination of chemotherapeutic with our siRNAs decreased the IC50 of both doxorubicine and etoposide. In Granta 519, si-224 decreased the IC 50 of doxorubicin by 32% and etoposide by 28%. In Jeko-1, si-778 decreased the IC 50 of doxorubicin by 49% but no effect on etoposide was seen. The magnitude of cyclin D1 down regulation seems to correlates with the percentages changes in IC 50. Klier et al previously reported that knockdown of cyclin D1 leads to an upregulation of cyclin D2 in MCL. Hence we mixed si-224 as well as si-778 targeting cyclin D1 with a si-D2 against cyclin D2 in combination with doxorubicine and etoposide in Granta-519. We also designed a dual-targeting siRNA against CCND1 and CCND2 (si-D1/D2). Targeting both cyclin D1 and D2 decreased the IC 50 of doxorubicin further than targeting cyclin D1 alone. Si224/D2 decreased the IC 50 of doxorubicin by 57% (si-224 alone 32%) and etoposide by 39% (si-224 alone 28%), and si778/D2 decreased the IC 50 of doxorubicine by 58% (si-778 alone 49%). The dual-targeting siRNA showed a decrease in IC 50 of doxorubicin by 45% and etoposide by 48%. Conclusions: Down regulation of cyclin D1 in MCL with siRNA improves the IC 50 of chemotherapeutic agents. Dual inhibition of both cyclin D1 and D2 further enhances the cytotoxic effect of doxorubicine and etoposide. Besides being a cell cycle regulator, cyclin D1 also seems to regulate chemosensitivity in MCL. Footnotes: This work was supported by grants from the Tower Cancer Research Foundation and Tim Nesvig Lymphoma Research Fund and Fellowship, Think Cure, Keck-foundation, SPORE. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document