Regulation of the Cyclin D1 Proto-Oncogene by Cell Cycle Regulatory MicroRNAs.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1801-1801
Author(s):  
Anagha Borwankar ◽  
Alessandro Pastore ◽  
Aniruddha Deshpande ◽  
Yvonne Zimmermann ◽  
Christian Buske ◽  
...  

Abstract Mutations, activation or overexpression of cyclin D1 are common features of several human cancers including mantle cell lymphoma (MCL) which bears the characteristic t(11;14) translocation juxtaposing the cyclin D1 gene downstream of the immunoglobulin heavy chain enhancer. The loss of the 3’UTR of this gene has been reported in a majority of MCL patients as well as in cell lines. In order to assess the impact of the 3’UTR on cyclin D1 expression levels, we used YFP tagged cyclin D1 reporter plasmids to quantify cyclin D1 expression in cell lines with different mutations of the 3’UTR. Interestingly, protein expression was significantly higher upon deletion of the cyclin D1 3’UTR compared to the full-length cyclin D1 gene as assessed by flow cytometry (2.1 fold; n=3, P < 0.05). Applying a more sensitive dual-luciferase reporter assay where a constitutively expressed luciferase gene was fused to the cyclin D1 3’UTR, the normalized firefly luciferase activity was reduced significantly to 23% as compared to luciferase only (the empty vactor). We then introduced 3’UTR mutations observed in MCL patients (insertion of adenosine between nucleotides 2308 and 2309 and a deletion of the tri-nucleotide sequence TCA from 2309–2311 of the full length cyclin D1-YFP reporter cDNA), which resulted in a significant increase of cyclin D1 expression (1.3 fold both in Ins308 and Δ309-311) compared to full length cyclin D1, (P< 0.05) showing that these mutations contribute to cyclinD1 overexpression in these patients. Subsequently, the 3’UTR was scanned for elements potentially regulating cyclin D1 expression, and putative microRNA binding sites were identified using the TargetScan and PicTar microRNA target prediction software. The most interesting candidate microRNAs include the miR-15/16 family and the miR-17–92 cluster, both of which have been shown to be involved in lymphoid malignancies and regulate cell cycle progression. In order to confirm whether the cyclin D1 3’UTR is a direct target of these microRNAs, we cloned the cyclin D1 3’UTR target region containing putative miR-15/16 or miR-17/20a binding sites and transfected these reporter constructs into HeLa cells. Upon introduction of oligonucleotide mimics of the miR15/16 microRNAs or a plasmid expressing microRNAs of the miR-17 cluster, the normalized luciferase activity of the respective luciferase reporters was reduced significantly to 41% (miR-15), 33% (miR-16) and 79% (miR-17/20a), respectively. Moreover, introduction of mutations in the seed sequences of the putative microRNA recognition sites rendered these constructs insensitive to inhibition by these microRNAs, confirming the specificity of the microRNA::target interaction. These data confirm that the binding of these microRNAs play an important role in the repression of cyclin D1 mediated by the 3’UTR and mutation or deletion result in cyclin D1 overexpression in MCL as well as other human tumors.

2014 ◽  
Vol 2014 ◽  
pp. 1-9 ◽  
Author(s):  
Guan Sun ◽  
Lei Shi ◽  
Shushan Yan ◽  
Zhengqiang Wan ◽  
Nan Jiang ◽  
...  

Aim. To investigate the role and mechanism of miR-15b in the proliferation and apoptosis of glioma.Methods. The miR-15b mimics were transfected into human glioma cells to upregulate the miR-15b expression. Cyclin D1 was determined by both western blotting analysis and luciferase reporter assay. Methylthiazol tetrazolium (MTT) and flow cytometry were employed to detect the cell proliferation, cell cycle, and apoptosis.Results. Overexpression of miR-15b inhibits proliferation by arrested cell cycle progression and induces apoptosis, possibly by directly targeting Cyclin D1. Both luciferase assay and bioinformatics search revealed a putative target site of miR-15b binding to the 3′-UTR of Cyclin D1. Moreover, expression of miR-15b in glioma tissues was found to be inversely correlated with Cyclin D1 expression. Enforced Cyclin D1 could abrogate the miR-15b-mediated cell cycle arrest and apoptosis.Conclusions. Our findings identified that miR-15b may function as a glioma suppressor by targeting the Cyclin D1, which may provide a novel therapeutic strategy for treatment of glioma.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5287-5287
Author(s):  
Robert W Chen ◽  
Myo Htut ◽  
Britta Hoehn ◽  
Eamon Berge ◽  
William Robinson ◽  
...  

Abstract Mantle Cell Lymphoma (MCL) represents 5–10% of all non-Hodgkins lymphomas, making it an uncommon but difficult form of lymphoma to treat. It has the worst prognosis among the B cell lymphomas with median survival of three years. The genetic hallmark of MCL is the t(11,14)(q13;32) translocation causing amplification of cyclin D1 (CCND1). It is a well known cell cycle regulator. Multiple reports have shown a truncation in the cyclin D1 mRNA 3′ untranslated region. This truncation increases CCND1 protein expression by not only enhancing the half-life of CCND1 mRNA, but also evades microRNA regulation of mRNA translation. The dramatic overexpression of CCND1 mRNA and protein has been associated to poor clinical outcome in patients. We hypothesize that this truncation leads to a more aggressive phenotype and induces chemoresistance in MCL. We have identified 4 MCL cell lines (Granta-519, JVM-2, Jeko-1, and Z138) with different levels of the truncated CCND1 mRNA. We were able to show that Z138 and Jeko-1 have a much higher ratio of truncated CCND1 mRNA to the full length CCND1 mRNA as compared to Granta-519 and JVM-2. We were also able to show that this truncated mRNA leads to an increase in CCND1 protein expression. By using flow cytometry, we correlated the increase in CCND1 protein expression to faster cell cycle progression. We proposed that cell lines with increased CCND1 expression are phenotypically more aggressive, and would be able to continue cell cycle progression without serum support. We were able to arrest JVM-2 in G1 phase after 48 hours of serum starvation. However, we were not able to arrest cell cycle progression in Jeko-1 even after 96 hours of serum starvation. Western blot analysis shows that CCND1 protein expression is decreased in JVM-2 but remains unchanged in Jeko-1 with serum starvation. The same phenomenon was observed in Granta-519 and Z138. The MCL cell lines (Jeko-1 and Z-138) with more CCND1 protein expression were able to continue cell cycle progression in serum free media. The MCL cell lines (JVM-2 and Granta-519) with less CCND1 protein expression were not able to continue cell cycle progression in serum free media. This shows that CCND1 overexpression is associated with a more aggressive phenotype. We then treated the 4 MCL cell lines with varying concentrations of doxorubicin, a standard anthracycline chemotherapy used in the treatment of MCL patients. We used MTS assay to assess cell proliferation after treatment with doxorubicin. We found the IC 50 (inhibitory concentration 50%) of doxorubicin in these cell lines varied from 6nM to 600nM. The cell lines (Jeko-1 and Z-138) with more CCND1 protein expression have a much higher IC 50 as compared to the cell lines (JVM-2 and Granta-519) with less CCND1 protein expression. This demonstrates that CCND1 overexpression is associated with chemoresistance. We conclude truncation in CCND1 mRNA leads to increased CCND1 protein expression and faster cell cycle progression CCND1 overexpression is associated with an aggressive phenotype CCND1 overexpression is associated with chemoresistance.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 417-417
Author(s):  
Goldi A. Kozloski ◽  
Xiaoyu Jiang ◽  
Karen L. Bunting ◽  
Ari M. Melnick ◽  
Izidore S Lossos

Abstract Abstract 417 MicroRNAs (miRNAs) exhibit differential expression in cancer and can be used as prognostic biomarkers. MiR-181a expression is reported to be associated with survival and outcome in acute myeloid and chronic lymphocytic leukemia patients. We demonstrated that miR-181a levels are independently associated with improved survival of diffuse large B cell lymphoma (DLBCL) patients treated with R-CHOP (Rituximab, Cyclophosphamide, Adriamycin, Oncovin, Prednisolone). However, the mechanism underlying this observation and the function of miR-181a in DLBCL pathogenesis are unknown. MiR-181a was expressed at higher levels in centroblasts compared to naïve and memory B cells, and at significantly higher levels in GCB-like compared to ABC-like DLBCL cell lines (p=0.017). These observations suggested that miR-181a may differentially target critical signaling pathways in GCB and ABC DLBCL. NF-kB serves a critical role in ABC DLBCL survival. Utilizing 3 miRNA target prediction algorithms, multiple NF-κB signaling pathway transcripts harbored putative miR-181a binding sites. Consequently, we tested the effect of miR-181a on CARD11, IBKα, p105/p50, and C-Rel expression in DLBCL cell lines (HBL1, VAL). Compared with a scrambled miRNA control, miR-181a expression decreased protein and mRNA levels of these targets. To confirm the effect was direct, we fused the 3′-UTR sequences of CARD11, IBKα, p105 and C-Rel, each containing miR-181a putative binding sites, to a luciferase reporter gene. Co-transfecting miR-181a with the corresponding constructs, we demonstrated that all the constructs had significantly repressed luciferase activity compared with a non-targeting control. The effect was specific, since miR-181a did not affect luciferase activity of CARD11, IBKα, p105 and C-Rel reporter constructs with mutated binding sites. Using an NF-κB luciferase reporter assay, we next demonstrated that compared to a scrambled control, miR-181a significantly decreased NF-κB reporter activity in DLBCL cell lines (VAL, SUDHL6, OCILY7, OCILY19, HBL1, RCK8). MiR-181a also decreased NF-κB reporter activity induced by anti-IgM and TNFα stimulation. Concordantly, anti-miR-181a increased endogenous p105/p50 and C-Rel protein levels. Because ubiquitinated-IKKγ drives NF-κB signaling, we tested the effect of miR-181a in TNFα-stimulated 293T cells on ubiquitinated-IKKγ. MiR-181a decreased levels of ubiquitinated-IKKγ, corroborating the observed inhibitory effects on NF-κB signaling. We reasoned that NF-κB signaling repression should coincide with a decrease in endogenous transcription activity from NF-κB promoters. Indeed, miR-181a decreased mRNA expression levels of NF-κB target genes (BCL2, IRF4, IL-6, IKBa, FN1, PIM1, BLR1, CCL3, CFLAR, FCER2, TP53) as measured by qRT-PCR in miR-181a-transfected HBL1 cells. Because miR-181a directly targets p105/p50 and REL proteins, we postulated that this may be one of the main mechanisms of NF-κB signaling repression. Indeed, an electrophoresis mobility shift assay along with super-shifts analyses showed a decrease in the p105/p50 protein in HeLa nuclear extracts. To examine the biological significance of differential miR-181a expression between GCB- and ABC-like DLBCL and elucidate its potential role in DLBCL pathogenesis, we next assessed cell death (Annexin V, 7AAD) and cell proliferation (BrdU, 7AAD) in GCB (SUDHL4, OCILY7, OCILY19, VAL) and ABC (HBL1, OCILY10, RCK8, U2932) DLBCL cell lines transfected with GFP labeled precursor miR-181a. MiR-181a expression significantly increased cell death and apoptosis of ABC versus GCB DLBCL (p=0.006). This was associated with a more pronounced G1 phase growth arrest in the ABC DLBCL cells. Our studies demonstrate that miR-181a is a master regulator of canonical NF-kB signaling by regulating the expression of multiple components of this pathway, an effect that may underlie the distinct prognosis of DLBCL with different miR-181a expression levels. Furthermore, miR-181a down regulation may contribute to the pathogenesis of ABC DLBCL. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Hu Chen ◽  
Lequn Bao ◽  
Jianhua Hu ◽  
Dongde Wu ◽  
Xianli Tong

BackgroundIn recent years, microRNA-1-3p (miR-1-3p) has been linked to the progression of multiple cancers, whereas little is known about its role in hepatocellular carcinoma (HCC). Herein, we investigated the function of miR-1-3p in HCC, and its regulatory function on origin recognition complex subunit 6 (ORC6).MethodsQuantitative real-time polymerase chain reaction (qRT-PCR) was performed for detecting the expression levels of miR-1-3p and ORC6 mRNA in HCC samples and cell lines. ORC6 expression at the protein level was quantified by Western blot. After gain-of-function and loss-of-function models were established, cell counting kit-8 (CCK-8) assays, Transwell assays, flow cytometry, and 5-Ethynyl-2′-deoxyuridine (EdU) assay were performed for examining cell proliferation, migration, invasion, cell cycle, and apoptosis. The targeting relationship between miR-1-3p and ORC6 was confirmed with bioinformatic analysis and dual-luciferase reporter assays.ResultsThe expression of miR-1-3p was reduced in HCC samples and cell lines. Overexpression of miR-1-3p suppressed the proliferation, migration, and invasion, and induced cell-cycle arrest and apoptosis of HCC cells, whereas the opposite effects were induced by miR-1-3p inhibition. ORC6 is identified as a novel target of miR-1-3p, the expression of which is negatively correlated with miR-1-3p expression in HCC tissues. ORC6 overexpression facilitated the proliferation, migration, invasion, and cell cycle progression, and reduced apoptosis of HCC cells, whereas the opposite effects were induced by ORC6 knockdown. What is more, ORC6 overexpression counteracted the biological functions of miR-1-3p in HCC cells.ConclusionMiR-1-3p targets ORC6 to suppress the proliferation, migration, invasion, and cell cycle progression, and promote apoptosis of HCC cells.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2549-2549
Author(s):  
Leticia Fröhlich Archangelo ◽  
Fabíola Traina ◽  
Philipp A Greif ◽  
Alexandre Maucuer ◽  
Valérie Manceau ◽  
...  

Abstract Abstract 2549 The CATS protein (also known as FAM64A and RCS1) was first identified as a novel CALM (PICALM) interactor that interacts with and influences the subcellular localization of CALM/AF10, a leukemic fusion protein found in acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL) and in malignant lymphoma. CATS is highly expressed in leukemia, lymphoma and tumor cell lines but not in non-proliferating T-cells or in peripheral blood lymphocytes (PBLs). The protein levels of CATS are cell cycle-dependent, induced by mitogens (e.g. PHA) and correlate with the proliferative state of the cell. Thus, CATS is as a marker for proliferation. Using CATS as a bait in a yeast two-hybrid screen we identified the Kinase Interacting Stathmin (KIS or UHMK1) as a CATS interacting partner. KIS is a serine/threonine kinase that positively regulates cell cycle progression through phosphorylation of p27KIP in leukemia cell lines. The interaction between CATS and KIS was confirmed by GST pull-down, and co-immunopreciptation. KIS interaction region was mapped to CATS N-terminal portion. Searching through the phosphorylation site databases PhosphoSitePlus™ (http://www.phosphosite.org) and Phosida (http://www.phosida.com/) we identified 9 residues within CATS shown to be subject of post-translational modification. Phosphorylation assay with recombinant KIS demonstrated that this kinase efficiently phosphorylated full length CATS and its N-terminal part, but not the C-terminal of the protein. To map the KIS phosphorylation site of CATS, peptides comprising all known phospho-sites of CATS N-terminal (S16, S129, S131, T133 and S135) and mutations of the putative KIS target motif (S129 and S131) were tested for KIS phosphorylation. Thereby, we identified CATS S131 as the unique target site for KIS phosphorylation. Western blot analysis of U2OS cells, which had undergone cell cycle synchronization by a double thymidine block, revealed that KIS fluctuated throughout the cell cycle and counteracted CATS levels. Furthermore, we analyzed KIS protein expression on bone marrow mononuclear cells (MNCs) of MDS and AML patients. We studied 5 healthy donors, 13 MDS patients (7 low-risk [RA/RARS] and 6 high-risk [RAEB/RAEBt] according to FAB classification) and 10 AML patients (7 de novo and 3 secondary). Western blot analysis revealed elevated levels of KIS in MDS and AML compared to the control samples. We used a reporter gene assay in order to determine the influence of KIS on the CATS-mediated transcriptional repression and to elucidate the role of KIS-dependent phosphorylation of CATS at serine 131 in this context. Coexpression of GAL4-DBD-CATS and KIS enhanced the inhibitory function of CATS on transactivation of the GAL4-tk-luciferase reporter. This effect of KIS was observed for both CATS wild type and CATS phospho-defective mutant (CATS S131A) but not when the kinase dead mutant KISK54R was used. Moreover, CATS phosphomimetic clone (CATSS131D) exerted the same transcriptional activity as the CATS wild type. These results demonstrate that KIS enhances the transcriptional repressor activity of CATS, and this effect is independent of CATS phosphorylation at S131 but dependent on the kinase activity of KIS. Finally, we investigated whether CATS would affect the CALM/AF10 function as an aberrant transcription factor. Coexpression of constant amounts of GAL4-DBD-CALM/AF10 and increasing amounts of CATS lead to reduced transactivation capacity of CALM/AF10 in a dose dependent manner. Our results show that CATS not only interacts with but is also a substrate for KIS, suggesting that CATS function might be modulated through phosphorylation events. The identification of the CATS-KIS interaction further supports the hypothesis that CATS plays an important role in the control of cell proliferation. Moreover the elevated levels of KIS in hematological malignances suggest that KIS could regulate CATS activity and/or function in highly proliferating leukemic cells. Thus our results indicate that CATS function might be important to understand the malignant transformation mediated by CALM/AF10. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 ◽  
Author(s):  
Yiwei Liu ◽  
Hairong Chen ◽  
Xiangcheng Li ◽  
Feng Zhang ◽  
Lianbao Kong ◽  
...  

Proteasome 26S subunit ATPase 2 (PSMC2) plays a pathogenic role in various cancers. However, its function and molecular mechanism in hepatocellular carcinoma (HCC) remain unknown. In this study, tissue microarray (TMA) analysis showed that PSMC2 is highly expressed in HCC tumors and correlates with poor overall and disease-free survival in HCC patients. Multivariate Cox regression analysis revealed that PSMC2 is an independent prognostic factor for HCC patients. Furthermore, our results showed that PSMC2 knockdown inhibited cell proliferation and suppressed tumorigenesis in vivo. Knockdown of PSMC2 increased the expression of p21 and therefore decreased the expression of cyclin D1. Dual-luciferase reporter assays indicated that depletion of PSMC2 significantly enhanced the promoter activity of p21. Importantly, PSMC2 knockdown-induced phenotypes were also rescued by downregulation of P21. Taken together, our data suggest that PSMC2 promotes HCC cell proliferation and cell cycle progression through the p21/cyclin D1 signaling pathway and could be a promising diagnostic and therapeutic target for HCC patients.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2001-2001 ◽  
Author(s):  
Irena Misiewicz-Krzeminska ◽  
Maria Eugenia Sarasquete ◽  
Carolina Vicente-Dueñas ◽  
Patryk Krzeminski ◽  
Katarzyna Wiktorska ◽  
...  

Abstract CCND2 is highly expressed in most of multiple myeloma (MM) samples without CCND1 or CCND3 overexpression. D-type cyclins are highly homologous proteins and there is a growing body of evidence that the functions of the D cyclins are mostly exchangeable. During mouse development, the three D-type cyclins are expressed following an often mutually exclusive pattern and their function may be tissue-specific. Likewise in MM CCND1 or CCND2 expression is commonly mutually exclusive. The mechanisms by which CCND2 is upregulated in a set of MMs are not completely deciphered. In this study, we investigated the role of post-transcriptional regulation through the interaction between miRNAs and their binding sites at3'UTR in CCND2 overexpression in MM. First, we observed that ectopic transfection of MM cell lines with several miRNAs, directly targeting CCND2 3'UTR according to luciferase reporter assay, decreased the level of cyclin D2, although not in all the cell lines. This fact suggested the possible disruption of miRNA target sites. Indeed, we detected the presence of short CCND2 mRNA, both in MM cell lines and primary cells, using four different methodological approaches: qRT-PCR, Northern blot, mRNA FISH and 3’RACE PCR with product sequencing. The short CCND2 isoform was observed by qRT-PCR in the majority of patients and in all MM cell lines expressing CCND2. This finding was confirmed by Northern blot results. The abundance of each CCND2 mRNA isoform was also assessed by two-color mRNA FISH designed to discriminate the two mRNA different in length. This approach also enabled us to notice that no subpopulation of cells distinguishable by the load of one isoform with respect to another was present. The results obtained by RACE experiments in MM cell lines support the idea that changes in CCND2 3’UTR length are explained by alternative polyadenylation (APA). The functional consequences of 3’UTR shortening is the mRNA stabilization due to the loss of miRNA sites and regulatory elements located in the 3’UTR. Accordingly, the luciferase assays using plasmids harboring the truncated CCND2 mRNA strongly confirmed the loss of miRNA sites in the shorter CCND2 mRNA isoform. The short 3’UTRs lacking miRNA-binding sites have been associated with increased expression of different genes at both the mRNA and the protein level. Here, we observed significant higher level of overall CCND2 mRNA expression in those MMs with greater abundance of the shorter 3'UTR isoform. The previous intriguing observation showing that the level of cyclin D2 increased in U266 when cyclin D1 was silenced, was confirmed by our experiments. Moreover, functional analysis showed significant mRNA shortening after CCND1 silencing, suggesting that cyclin D1 could downregulate CCND2 level by modification of polyadenylation/clavage reaction. Since CCND2 expression is undetectable in myeloma cells with t(11;14), we extended our investigation to explore if DNA methylation might play a role in abolishing CCND2 expression. We observed that the CpG island more proximal to CCND2 TSS was highly methylated in MM cell lines with t(11;14). In summary, our results reveal that CCND2 expression in MM is mainly regulated by post-transcriptional mechanisms. Downregulation of specific miRNAs directly targeting CCND2 contributes to overexpression of CCND2 in a set of MM. Moreover, the shortening of CCND2 3'UTR by alternative polyadenylation with the consequent loss of miRNA binding sites is also participating in CCND2 upregulation. In fact, this mechanism seems to play a decisive role in the regulatory network between CCND1 and CCND2 in MM. Disclosures No relevant conflicts of interest to declare.


2015 ◽  
Vol 112 (38) ◽  
pp. E5261-E5270 ◽  
Author(s):  
Gero Doose ◽  
Andrea Haake ◽  
Stephan H. Bernhart ◽  
Cristina López ◽  
Sujitha Duggimpudi ◽  
...  

Despite the established role of the transcription factor MYC in cancer, little is known about the impact of a new class of transcriptional regulators, the long noncoding RNAs (lncRNAs), on MYC ability to influence the cellular transcriptome. Here, we have intersected RNA-sequencing data from two MYC-inducible cell lines and a cohort of 91 B-cell lymphomas with or without genetic variants resulting in MYC overexpression. We identified 13 lncRNAs differentially expressed in IG-MYC-positive Burkitt lymphoma and regulated in the same direction by MYC in the model cell lines. Among them, we focused on a lncRNA that we named MYC-induced long noncoding RNA (MINCR), showing a strong correlation with MYC expression in MYC-positive lymphomas. To understand its cellular role, we performed RNAi and found that MINCR knockdown is associated with an impairment in cell cycle progression. Differential gene expression analysis after RNAi showed a significant enrichment of cell cycle genes among the genes down-regulated after MINCR knockdown. Interestingly, these genes are enriched in MYC binding sites in their promoters, suggesting that MINCR acts as a modulator of the MYC transcriptional program. Accordingly, MINCR knockdown was associated with a reduction in MYC binding to the promoters of selected cell cycle genes. Finally, we show that down-regulation of Aurora kinases A and B and chromatin licensing and DNA replication factor 1 may explain the reduction in cellular proliferation observed on MINCR knockdown. We, therefore, suggest that MINCR is a newly identified player in the MYC transcriptional network able to control the expression of cell cycle genes.


2002 ◽  
Vol 22 (12) ◽  
pp. 4309-4318 ◽  
Author(s):  
Latha Shivakumar ◽  
John Minna ◽  
Toshiyuki Sakamaki ◽  
Richard Pestell ◽  
Michael A. White

ABSTRACT The RASSF1A locus at 3p21.3 is epigenetically inactivated at high frequency in a variety of solid tumors. Expression of RASSF1A is sufficient to revert the tumorigenicity of human cancer cell lines. We show here that RASSF1A can induce cell cycle arrest by engaging the Rb family cell cycle checkpoint. RASSF1A inhibits accumulation of native cyclin D1, and the RASSF1A-induced cell cycle arrest can be relieved by ectopic expression of cyclin D1 or of other downstream activators of the G1/S-phase transition (cyclin A and E7). Regulation of cyclin D1 is responsive to native RASSF1A activity, because RNA interference-mediated downregulation of endogenous RASSF1A expression in human epithelial cells results in abnormal accumulation of cyclin D1 protein. Inhibition of cyclin D1 by RASSF1A occurs posttranscriptionally and is likely at the level of translational control. Rare alleles of RASSF1A, isolated from tumor cell lines, encode proteins that fail to block cyclin D1 accumulation and cell cycle progression. These results strongly suggest that RASSF1A is an important human tumor suppressor protein acting at the level of G1/S-phase cell cycle progression.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4236-4236
Author(s):  
Brigitte Sola ◽  
Juliette Gauduchon ◽  
Sophie Krieger ◽  
Mikel Roussel ◽  
Denis Lesage ◽  
...  

Abstract Cyclin D1 is an essential regulator of cell cycle progression. Over-expression of cyclin D1 is associated with tumorigenesis both in solid cancers and in hematological malignancies such as mantle cell lymphoma (MCL), hairy cell leukemia and multiple myeloma (MM). The CCND1 gene coding for cyclin D1 exhibit an A/G polymorphism at the nucleotide 870 which is responsible for the generation of an alternative splice variant called form b. The cyclin D1b is identical to the typical form a, except in the C-terminal part. The cyclin D1b has lost the PEST domain responsible for protein degradation and the threonine 286, the site of phosphorylation by the glycogen synthase kinase-3 essential for the nuclear export. Cyclin D1b has been recently described as a nuclear oncogene (Lu et al., Cancer Res63, 7056, 2003; Solomon et al., J Biol Chem278, 30339, 2003). Using real time quantitative RT-PCR, we have detected cyclin D1b mRNA in all cyclin D1a-expressing MM and MCL cell lines and in MCL patients. The cyclin D1b protein, detected with an antibody specific of the form b (a generous gift of Dr Alan Diehl) is present in a limited number of samples (1/6 MM and 1/1 MCL cell lines) and localized both in the nucleus and in the cytoplasm. We have generated B-cells conditionally (ecdysone system) expressing cyclin D1a or cyclin D1b proteins. B-cells were stimulated with ponasterone A to induce cyclin D1 synthesis and then with cycloheximide. The two alternative proteins forms displayed similar stability: calculated half-lives are 35 min and 50 min for a and b respectively. By co-immunoprecipitation and western blots experiments, we determined that, although cyclin D1b was able to bind efficiently CDK4 (to the same extent than cyclin D1a), cyclin D1b/CDK4 complexes weakly catalyzed the phosphorylation of pRB. We further showed by cytofluorometric analysis of propidium iodide-stained cells, that cyclin D1b did not allow the entry of quiescent cells within the cell cycle and did not allow the progression through the G1 phase. This implies that cyclin D1b plays a minor role in cell cycle regulation. However, cyclin D1b, such as cyclin D1a (Duquesne et al., Cell Death Different8, 51, 2001) potentiated the apoptosis induced by growth factor deprivation. It us tempting to speculate that the contribution of cyclin D1b for B-cell hemopathies does not occur through deregulation of the pRb pathway but more likely through deregulation of apoptotic pathways. We will study these aspects in the future.


Sign in / Sign up

Export Citation Format

Share Document