Sequestration of p27Kip1 protein by cyclin D1 in typical and blastic variants of mantle cell lymphoma (MCL): implications for pathogenesis

Blood ◽  
2003 ◽  
Vol 101 (8) ◽  
pp. 3181-3187 ◽  
Author(s):  
Leticia Quintanilla-Martinez ◽  
Theresa Davies-Hill ◽  
Falko Fend ◽  
Julia Calzada-Wack ◽  
Lynn Sorbara ◽  
...  

Abstract p27 is a cyclin-dependent kinase inhibitor that plays a critical role in regulating G1/S progression, and whose activity is, in part, regulated through interactions with D-type cyclins. Mantle cell lymphoma (MCL) is characterized by the t(11;14) translocation resulting in deregulated cyclin D1. We previously showed that p27 expression in MCL, as assessed by immunohistochemistry (IHC), does not show the usual inverse relationship to proliferate seen in most other lymphomas that do not overexpress cyclin D1. This suggested that the normal expression or control of p27 activity on cell growth might be altered through potential interactions with cyclin D1. Using Western blot and coimmunoprecipitation studies, we assessed the interrelationship between cyclin D1 and p27 in several cyclin D1+ cell lines and primary MCL cases. Similar to our previous results by IHC, typical MCLs showed lower expression of p27 when compared to the more highly proliferative blastic cases or cell lines (mean arbitrary units: 58 versus 236 versus 120). Cyclin D1 was expressed at variable levels in both typical and blastic MCLs. p27 protein could be consistently coimmunoprecipitated with cyclin D1 from both cell lines and cases. Using techniques of exhaustive immunoprecipitation, we could demonstrate that most p27 protein was sequestered into complexes containing cyclin D1. We hypothesize that mantle cell lymphomagenesis results not only from direct consequences of inappropriate cyclin D1 expression, but also from the ability of overexpressed cyclin D1 to buffer physiologic changes in p27 levels, thereby rendering p27 ineffective as an inhibitor of cellular growth.

Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2120 ◽  
Author(s):  
Diana Malarikova ◽  
Adela Berkova ◽  
Ales Obr ◽  
Petra Blahovcova ◽  
Michael Svaton ◽  
...  

Mantle cell lymphoma (MCL) is a subtype of B-cell lymphoma with a large number of recurrent cytogenetic/molecular aberrations. Approximately 5–10% of patients do not respond to frontline immunochemotherapy. Despite many useful prognostic indexes, a reliable marker of chemoresistance is not available. We evaluated the prognostic impact of seven recurrent gene aberrations including tumor suppressor protein P53 (TP53) and cyclin dependent kinase inhibitor 2A (CDKN2A) in the cohort of 126 newly diagnosed consecutive MCL patients with bone marrow involvement ≥5% using fluorescent in-situ hybridization (FISH) and next-generation sequencing (NGS). In contrast to TP53, no pathologic mutations of CDKN2A were detected by NGS. CDKN2A deletions were found exclusively in the context of other gene aberrations suggesting it represents a later event (after translocation t(11;14) and aberrations of TP53, or ataxia telangiectasia mutated (ATM)). Concurrent deletion of CDKN2A and aberration of TP53 (deletion and/or mutation) represented the most significant predictor of short EFS (median 3 months) and OS (median 10 months). Concurrent aberration of TP53 and CDKN2A is a new, simple, and relevant index of chemoresistance in MCL. Patients with concurrent aberration of TP53 and CDKN2A should be offered innovative anti-lymphoma therapy and upfront consolidation with allogeneic stem cell transplantation.


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 24 (18_suppl) ◽  
pp. 7573-7573 ◽  
Author(s):  
V. Y. Yazbeck ◽  
G. V. Georgakis ◽  
Y. Li ◽  
A. Younes

7573 Background: Mantle cell lymphoma (MCL) is a distinct type of B-cell lymphoma associated with transient response to conventional chemotherapy, continuous relapses and median survival of only 3–4 years. The mammalian target of rapamycin (mTOR) pathway is activated in many human malignancies where it regulates cyclin D1 translation. In a phase II trial, temsirolimus (CCI-779), an inhibitor of mTOR kinase used as single agent achieved an overall response rate of 38% in relapsed MCL patients. Our goal was to determine the activity and the mechanism of action of CCI-779 in MCL cell lines and to examine whether CCI-779 may synergizes with proteasome inhibitors. Methods: The activity of CCI-779 was determined in 3 mantle cell lymphoma cell lines (Jeko 1, Mino, Sp 53). Cell viability was determined by MTS assay, and autophagy by Acridine orange. Analysis of cell cycle was performed by flow cytometry and apoptosis by Annexin-V binding. Molecular changes were determined by western blot . Results: CCI-779 induced cell growth arrest in all cell lines in a time and dose dependent manner. The antiproliferative activity was due to cell cycle arrest in the G0/G1 phase followed by autophagy. CCI-779 decreased S6 phosphorylation in Jeko 1,Sp 53 indicative of mTOR inhibition. Furthermore, CCI-779 downregulated p21 expression in all three cell lines, without altering p 27 expression. Moreover, CCI-779 decreased the expression of the antiapoptotic protein cFLIP and ERK in both Jeko1 and Sp 53, but had no effect on cyclin D1 expression. The proteasome inhibitor bortezomib was also effective in all MCL cell lines, but failed to demonstrate significant synergy with CCI-779. Conclusions: The antiproliferative activity of CCI-779 in MCL is mediated by p21 downregulation and autophagy, without significant effect on cyclin D1 expression. The lack of synergy between bortezomib and CCI-779 should be confirmed using fresh MCL tumor cells. No significant financial relationships to disclose.


Blood ◽  
1997 ◽  
Vol 90 (8) ◽  
pp. 3154-3159 ◽  
Author(s):  
M. Michaela Ott ◽  
Jirina Bartkova ◽  
Jiri Bartek ◽  
Alexander Dürr ◽  
Lars Fischer ◽  
...  

Abstract The cell cycle regulatory protein cyclin D1 is essential for G1-S phase transition in several epithelial and mesenchymal tissues but is apparently not essential in normal mature B cells. An overexpression of cyclin D1 is induced by the chromosomal translocation t(11; 14)(q13; q32), which characterizes non-Hodgkin's lymphomas (NHLs) of mantle cell type. We studied 26 cases of mantle cell lymphoma (MCL) for the expression of cyclins D1 and D3. A total of 23 lymphomas showed a nuclear staining for cyclin D1, whereas reactive B cells of residual germinal centers were constantly negative. When compared with cyclin D3, an inverse staining pattern emerged. Whereas the B cells of residual germinal centers reacted strongly positive for cyclin D3, there was low or missing expression of cyclin D3 in MCL cells. In other B-cell lymphomas (n = 55), including chronic lymphocytic leukemia, low-grade lymphomas of mucosa-associated lymphatic tissue, follicular lymphomas, and diffuse large B-cell lymphomas, no cyclin D1 expression could be detected and 89% of these cases displayed cyclin D3 positivity. Lymphoma cell lines harboring the t(11; 14) showed cyclin D1 protein but no or very low levels of cyclin D3; three other B-cell lines, a T-cell line, and peripheral blood lymphocytes strongly expressed cyclin D3 and reacted negatively for cyclin D1. We conclude that the chromosomal translocation t(11; 14) leads to an abnormal protein expression of cyclin D1 in the tumor cells of MCL and induces a consecutive downregulation of cyclin D3. In contrast to other B-NHLs, cyclin D1 and D3 expression in MCL is not related to the growth fraction.


Blood ◽  
1997 ◽  
Vol 89 (3) ◽  
pp. 965-974 ◽  
Author(s):  
Kaoru Uchimaru ◽  
Toshiyasu Taniguchi ◽  
Miwa Yoshikawa ◽  
Shigetaka Asano ◽  
Andrew Arnold ◽  
...  

Abstract In mantle cell lymphoma, the t(11; 14)(q13; q32) and its molecular counterpart, bcl-1 rearrangement, are consistent features and lead to cyclin D1 (bcl-1, PRAD1) proto-oncogene overexpression. In order to detect cyclin D1 overexpression, we developed a simple assay involving a reverse transcription followed by competitive polymerase chain reaction (PCR). A single upstream primer was derived from a homologous region between cyclin D1 and the other D-type cyclins, cyclins D2 and D3, while three downstream primers were specific to their respective D-type cyclins. Because the upstream primer was shared in PCR amplification of the three sequences, each PCR product served as a competitor and the quantification of the target was made by comparison of the intensity of the three products. With this assay we analyzed 45 hematopoietic cell lines and 40 clinical specimens. Cyclin D1 was rarely expressed in lymphoid cell lines except in t(11; 14)(q13; q32)-bearing B-cell malignancies and/or mantle cell lymphoma, which expressed cyclin D1 predominantly. In myeloid cell lines, the levels of cyclin D1 expression varied and never exceeded the sum of cyclin D2 and D3 levels. Cyclin D3 was ubiquitously expressed while cyclins D1 and D2 were differentially used. The observations suggest that human cyclin D3 may play a fundamental role in hematopoiesis and that cyclins D1 and D2 may have different lineage- or differentiation-dependent functions. With this assay, small aliquots of clinical specimens such as 100 μL peripheral blood were enough to detect cyclin D1 overexpression without a well-controlled standard. The technique was validated as highly comparable with Northern analysis. This rapid and reliable detection of cyclin D1 overexpression may have practical clinical utility in the analysis and management of B-cell malignancies.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1579-1579
Author(s):  
Weiming Xu ◽  
Christine Kang ◽  
Mercedes Delgado ◽  
Sophie M Perrin-Ninkovic ◽  
Patrick W Papa ◽  
...  

Abstract Mantle cell lymphoma (MCL) is a distinct sub-type non-Hodgkin lymphoma characterized by overexpression of cyclin D1 (CCND1) in 95% of patients due to the cytogenetic change of chromosome translocation t(11;14) (q13;q32). It remains one of the most challenging lymphomas associated with shorter response duration to conventional chemotherapy as well as continuous relapses and refractory to current drugs. However, dysregulation of cyclin D1 biology alone is insufficient to develop MCL. The emerging data suggest that the mammalian target of rapamycin (mTOR) plays a crucial role in the proper transmission of proliferative and anti-apoptotic signals through the PI3K/AKT pathway that makes it an attractive therapeutic target for hematological malignances including mantle cell lymphoma. As a single agent, rapamycin analogs such as temsirolimus (CCI-779) achieved 38% overall response rate in heavily pretreated MCL and prolonged progression free survival (PFS) in relapsed and refractory mantle cell lymphoma (4.8 months in temsirolimus vs. 1.9 months in investigator’s choice, ASCO 2008). mTOR regulates two distinct complexes TORC1 and TORC2. TORC1 complex is involved in cell cycle regulation by phosphorylating p70S6K and 4E-BP1, two molecules that are important for translational control of cyclin D1 and c-myc as well as ribosomal biogenesis whereas TORC2 complex mainly regulates phospho- AKT serine 473 leading to cell survival and proliferation. mTOR kinase also negatively regulates autophagy, a process of cellular bulk protein degradation by fusion to lysosomes upon the nutrient deprivation. We have developed mTOR kinase selective inhibitors which exhibit distinct biological profile from rapamycin in many cancer cell lines. Here we demonstrate that a selective mTOR kinase inhibitor displays potent anti-proliferative activity in JeKo-1 and Mino cells associated with decreased phosphorylation of S6, p70S6K, AKT S473, 4E-BP1 as well as decreased cyclin D1 levels leading to G1 arrest. The inhibitor also promotes autophagic cell death at 72h and 96h post-treatment. Furthermore a selective mTOR kinase inhibitor but not rapamycin induces a significant apoptosis in JeKo-1 and Mino cells. The observed apoptosis is correlated with caspases mediated PARP cleavage as well as inhibition of anti-apoptotic protein Mcl-1, suggesting TORC2/AKT S473 complex may provide survival signaling for mantle cell lymphoma. A timecourse study demonstrated that JeKo-1 and Mino cells undergo apoptosis at 24h and 48h followed by significant autophagic cell death at 72h and 96h in a dose dependent manner when exposed to our mTOR kinase inhibitor. In conclusion, mTOR kinase inhibitors are able to induce G1 cell cycle arrest, caspase-dependent apoptosis and autophagic cell death that contribute to the anti-tumor activity. Therefore it may provide a powerful alternative targeted therapy for mantle cell lymphoma.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 989-989
Author(s):  
Elliot M. Epner ◽  
Jing Huang ◽  
Hui Liu ◽  
Jin Wang ◽  
Shugiang Liang ◽  
...  

Abstract Cyclin D1 expression in B cells is deregulated by long distance chromosome translocations involving the immunoglobulin heavy chain (IgH) locus in mantle cell lymphoma (MCL). Ge. Using chromatin immunoprecipitation (ChIP) assays, we show that the cyclin D1 (CCND1) loci in MCL cell lines and MCL patient samples are packaged in chromatin containing CCCTC binding factor (CTCF) and nucleophosmin (NPM). Using allelic chromatin immunoprecipitation (ChIP) assays, we demonstrate that both the translocated and nontranslocated cyclin D1 alleles bind both CTCF and NPM, while RNA polymerase II is only bound to the translocated allele. Sequential ChIP assays demonstrated CTCF and NPM binding at the cyclin D1 loci in MCL cells. By immunoFISH, we demonstrate that the translocated and nontranslocated cyclin D1 loci colocalize at the nucleolus. CTCF and NPM are bound at the IgH 3′ regulatory elements only in MCL cell lines. NPM shRNA effects a specific growth arrest in these cells. These data demonstrate a role for CTCF and NPM in long distance cyclin D1 deregulation in mantle cell lymphoma.


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