Mutually Exclusive Cyclin-Dependent Kinase 4/Cyclin D1 and Cyclin-Dependent Kinase 6/Cyclin D2 Pairing Inactivates Retinoblastoma Protein and Promotes Cell Cycle Dysregulation in Multiple Myeloma

2005 ◽  
Vol 65 (24) ◽  
pp. 11345-11353 ◽  
Author(s):  
Scott Ely ◽  
Maurizio Di Liberto ◽  
Ruben Niesvizky ◽  
Linda B. Baughn ◽  
Hearn J. Cho ◽  
...  
Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 124-124
Author(s):  
John Quinn ◽  
Janet Glassford ◽  
Laura Percy ◽  
Philippa Munson ◽  
Manuel Rodriguez-Justo ◽  
...  

Abstract Abstract 124 Introduction: Understanding how multiple myeloma (MM) cells proliferate and self-renew is crucial to treating resistant disease and preventing relapse. The TNF family members APRIL (A proliferation-inducing ligand) and BAFF (B-cell activating factor) are secreted in the MM bone marrow microenvironment and share two common receptors, TACI and BCMA. APRIL and BAFF and have been shown to exert a mainly anti-apoptotic effect in human myeloma cell lines (HMCLs). Little is known, however, about the proliferative effect of APRIL and BAFF on primary MM cells. Aims: We aimed to determine the effect of APRIL and BAFF on cell cycle progression in primary MM cells and on the modulation of D-type cyclins and other cell cycle proteins. Patients and Methods: We studied the effects of APRIL and BAFF on purified MM cells from 26 patients by flow-cytometry using surface CD138 / Ki67 / propidium iodide staining. A culture system was optimised in which a mean (±SEM) of 73.3±10.9% (n=23) of primary CD138+ MM cells survived for up to 3 days in vitro. D-type cyclin expression was assessed by immuno-histochemistry (IHC) ± western blotting and correlated with FISH for IgH translocations. Fourteen patients expressed cyclin D1, 4 in association with t(11;14); while 12 patients expressed cyclin D2, 1 with t(4;14) and 4 with t(14;16). TACI and BCMA expression were determined by flow cytometry. Western blotting was used to determine expression and modulation of cell cycle proteins and activation of signalling pathways in vitro. Membrane-bound APRIL was detected by IHC in MM trephine biopsies. Results: In-vitro culture with APRIL for 72 hours increased the overall percentage of CD138+ cells in S+G2/M phases (S/G2M) from a mean (±SEM) of 5.5 ± 1.1% to 8.1 ± 1.5% (p<0.05). However, cyclin D2 expressing CD138+ cells demonstrated a greater response to APRIL than D1 expressing cells. In the D2 group (n=12), APRIL increased the S/G2M fraction from 8.3 ± 2.1% to 13.5 ± 2.2%, (p<0.01), while in the D1 group (n=14), proliferation was unaffected (3.0 ± 0.5%, compared with 3.1 ± 0.7% in control). Similar results were obtained when absolute numbers of CD138+ cells in S/G2M were analysed. Proliferative response to APRIL was more marked in those cases with IgH translocations (S/G2M fraction increased from 6.4±1.6% to 14.8±2.0%, p<0.001). In comparison with APRIL, BAFF had a lesser effect, increasing the S/G2M fraction in the D2 group from 8.3 ± 2.1% in control to 11.7 ± 3.5%, p=0.05). BAFF had no significant effect on proliferation in the D1 group. Results were verified by increased 3H-thymidine and bromodeoxyuridine uptake. APRIL-induced proliferation was maximal after 48-72 hours of incubation, was dose-dependent (maximal effect at 200ng/ml) and completely inhibited by TACI-Fc. In the D2 group, proliferation in response to APRIL was accompanied by increased expression of cyclin D2, cdk4, cdk6 and phospho-pRB by western blotting. In contrast, in the D1 group, neither cyclin D1 nor other cell cycle regulators was affected by APRIL. Proliferative response to APRIL was accompanied by increased expression of phospo-PKB. APRIL-induced proliferation showed a positive correlation with TACI expression (p<0.01), but not with BCMA. In contrast to the cell cycle effect, APRIL had no significant effect on survival of primary CD138+ cells either in D2 or D1 groups (viable cell number was 91±22%, and 129±18% of control respectively). IHC revealed the presence of membrane-bound APRIL on MM cells in BM sections from both D1 and D2 patients. IHC was used to examine expression of cell cycle regulators in vivo. In 5 cases where cyclin D1 expression was associated with proliferation, ie phospho-pRb expression, strong expression of cdk6 was seen, with weaker expression of cdk4. Expression of cyclin D2 was always accompanied by phospho-pRb expression (n=5), with strong expression of both cdk4 and -6 in 3, and just cdk6 in 2. Conclusions: APRIL stimulates the proliferation of cyclin D2 expressing primary MM cells, in particular those associated with known IgH translocations, but has minimal effect on cells expressing cyclin D1. These findings suggest that MM cells from different cyclin D/translocation classes rely on different stimuli for proliferation and cell-cycle re-entry. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 117 (3) ◽  
pp. 890-901 ◽  
Author(s):  
John Quinn ◽  
Janet Glassford ◽  
Laura Percy ◽  
Philippa Munson ◽  
Teresa Marafioti ◽  
...  

Abstract A proliferation-inducing ligand (APRIL) promotes survival and drug resistance in multiple myeloma (MM) cell lines. We studied the effect of APRIL on cell-cycle behavior in primary MM cells and correlated our findings with D-type cyclin expression by immunohistochemistry and/or Western blotting. In MM cases, expressing cyclin D2 APRIL significantly increased the percentage of CD138+ cells in S + G2/M phase (from 8.4% ± 1.9% to 14.3% ± 2.6%, n = 15, P < .01), whereas a lesser effect was seen in cases expressing cyclin D1 (n = 18). Cell-cycle response to APRIL was most marked for cyclin D2-expressing cases with IgH translocations (P < .01) and was accompanied by increased expression of cyclin D2, CDK4, CDK6, and phospho-retinoblastoma protein. Cell-cycle proteins in cyclin D1+ cells were not modulated by APRIL. Surface expression of B-cell maturation antigen and transmembrane activator and calcium-modulating cyclophilin ligand interactor was not significantly different between cyclin D1+ and D2+ MM cells. We observed activation of nuclear factor-κB and PI3-kinase pathways in response to APRIL in both cyclin D1+ and D2+ MM cells. In conclusion, APRIL stimulates G1/S progression in cyclin D2+ MM cells bearing IgH translocations but has minimal effect on cyclin D1+ cells, suggesting MM cells from different cyclin D/translocation classes rely on different mechanisms for cell-cycle re-entry.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 501-501
Author(s):  
Scott Ely ◽  
Maurizio Di Liberto ◽  
Ruben Niesvizky ◽  
Eunice Hatada ◽  
Hearn Cho ◽  
...  

Abstract Multiple myeloma (MM), the second most common hematopoietic cancer, ultimately becomes refractory to treatment when self-renewing MM cells begin unrestrained proliferation by unknown mechanisms. Cell cycle reentry and progression is modulated by the balance between positive cell cycle regulators [cyclins and cyclin-dependent kinases (Cdk)] on the one hand and Cdk inhibitors (CKI) on the other. Here we show that one, but not more than one, of the three early G1 D cyclins is elevated in each case of MM. However, cyclin D1 or D3 overexpression rarely leads to phosphorylation of the retinoblastoma protein Rb and G1 cell cycle progression, unless cyclin-dependent kinase 4 (Cdk4) is correspondingly elevated in the absence of Cdk6. By contrast, cyclin D2 and Cdk6 are coordinately increased, thereby overriding inhibition by Cdk inhibitors, p18INK4c and p27Kip1, and phosphorylating Rb in conjunction with Cdk4. Thus, Cdk4 pairs exclusively with cyclin D1 whereas Cdk6 pairs exclusively with cyclin D2, although Cdk4 can also pair with cyclin D2 in MM cells. The basis for the exclusive Cdk4-cyclin D1 and Cdk6-cyclin D2 pairing is coordinated transcriptional activation. Cyclin D1 or D3 expression is scattered or uniform among bone marrow MM cells and at a constant frequency in the clinical course. By contrast, phosphorylation of Rb by Cdk6-cyclin D2 occurs in discrete foci of bone marrow MM cells early in the clinical course before cell proliferation, then is heightened with proliferation and disease progression. Mutually exclusive Cdk4-cyclin D1 and Cdk4/6-cyclin D2 pairing, therefore, critically controls cell cycle re-entry and G1 progression, which underlies the expansion of self-renewing MM cells in MM progression.


2000 ◽  
Vol 23 (1) ◽  
pp. 73-83 ◽  
Author(s):  
Francois Roudier ◽  
Elena Fedorova ◽  
Janos Gyorgyey ◽  
Attila Feher ◽  
Spencer Brown ◽  
...  

Endocrinology ◽  
2004 ◽  
Vol 145 (12) ◽  
pp. 5439-5447 ◽  
Author(s):  
Maofu Fu ◽  
Chenguang Wang ◽  
Zhiping Li ◽  
Toshiyuki Sakamaki ◽  
Richard G. Pestell

Abstract Cyclin D1 encodes the regulatory subunit of a holoenzyme that phosphorylates and inactivates the retinoblastoma protein and promotes progression through the G1-S phase of the cell cycle. Amplification or overexpression of cyclin D1 plays pivotal roles in the development of a subset of human cancers including parathyroid adenoma, breast cancer, colon cancer, lymphoma, melanoma, and prostate cancer. Of the three D-type cyclins, each of which binds cyclin-dependent kinase (CDK), it is cyclin D1 overexpression that is predominantly associated with human tumorigenesis and cellular metastases. In recent years accumulating evidence suggests that in addition to its original description as a CDK-dependent regulator of the cell cycle, cyclin D1 also conveys cell cycle or CDK-independent functions. Cyclin D1 associates with, and regulates activity of, transcription factors, coactivators and corepressors that govern histone acetylation and chromatin remodeling proteins. The recent findings that cyclin D1 regulates cellular metabolism, fat cell differentiation and cellular migration have refocused attention on novel functions of cyclin D1 and their possible role in tumorigenesis. In this review, both the classic and novel functions of cyclin D1 are discussed with emphasis on the CDK-independent functions of cyclin D1.


2003 ◽  
Vol 178 (2) ◽  
pp. 319-329 ◽  
Author(s):  
K Takahashi ◽  
M Ohmichi ◽  
M Yoshida ◽  
K Hisamoto ◽  
S Mabuchi ◽  
...  

The proliferation of vascular smooth muscle cells (VSMC) is a crucial pathophysiological process in the development of atherosclerosis. Although estrogen is known to inhibit the proliferation of VSMC, the mechanism responsible for this effect remains to be elucidated. In addition, the effect of raloxifene on VSMC remains unknown. We have shown here that 17beta-estradiol (E(2)) and raloxifene significantly inhibited the platelet-derived growth factor (PDGF)-stimulated proliferation of cultured human VSMC. Flow cytometry demonstrated that PDGF-stimulated S-phase progression of the cell cycle in VSMC was also suppressed by E(2) or raloxifene. We found that PDGF-induced phosphorylation of retinoblastoma protein (pRb), whose hyperphosphorylation is a hallmark of the G1-S transition in the cell cycle, was significantly inhibited by E(2) and raloxifene. These effects were associated with a decrease in cyclin D1 expression, without a change in cyclin-dependent kinase 4 or cyclin-dependent kinase inhibitor, p27(kip1) expression. ICI 182,780 abolished the inhibitory effects of E(2) and raloxifene on PDGF-induced pRb phosphorylation. Next, we examined which estrogen receptor (ER) is necessary for these effects of E(2) and raloxifene. Since VSMC express both ERalpha and ERbeta, A10, a rat aortic smooth muscle cell line that expresses ERbeta but not ERalpha, was used. The dose-dependent stimulation of A10 cell proliferation by PDGF was not inhibited by E(2) or raloxifene in contrast to the results obtained in VSMC. Moreover, E(2) and raloxifene significantly inhibited the PDGF-induced cyclin D1 promoter activity in A10 cells transfected with cDNA for ERalpha but not in the parental cells. These results suggested that E(2) and raloxifene exert an antiproliferative effect in VSMC treated with PDGF, at least in part through inhibition of pRb phosphorylation, and that the inhibitory effects of E(2) and raloxifene may be mainly mediated by ERalpha.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 953-953 ◽  
Author(s):  
Linda Resar ◽  
Joelle Hillion ◽  
Katrina Alino ◽  
Michelle Rudek ◽  
Judith Karp

Abstract Acute leukemia in adults continues to be a formidable clinical challenge that demands further investigation to identify more rational therapies. To optimize anti-leukemia therapy, we are investigating the prototypical cyclin dependent kinase (cdk) inhibitor, flavopiridol, in refractory or poor-risk disease. Flavopiridol is a cytotoxic molecule that is thought to induce cell cycle arrest by blocking cyclin-dependent kinase (cdk) function, thereby interfering with RNA Polymerase II activity and globally down-regulating gene expression. In the setting of pan-cdk inhibition, E2F1 is released and appears to drive apoptosis in transformed cells. Consistent with these proposed mechanisms of action, a previous study from our group showed that flavopiridol induces apoptosis in vitro in leukemic blasts from patients with refractory leukemia. Administration of flavopiridol was associated with a decrease in one or more of the following proteins in the leukemic blasts: RNA Polymerase II, STAT3, cyclin D1, Bcl-2, and Mcl-1. Serum VEGF levels also decreased in most patients. We are now investigating mRNA levels of the genes encoding these proteins by quantitative, RT-PCR in leukemic blasts from adult patients with refractory or poor-risk leukemia before and after flavopiridol therapy. We have treated 26 patients with flavopiridol at an escalating, hybrid dose followed by ara-c and mitxantrone. Adequate RNA from leukemic blasts before and after flavopiridol administration was available from 8 of 11 patients studied thus far. All cases (8/8) exhibit a marked decrease in mRNA for VEGF following flavopiridol. mRNA levels for other putative flavopiridol target genes is also decreased in a subset of leukemic blast samples after therapy, as follows: E2F1 (6/8), STAT3 (6/8), Mcl-1 (6/8), RNA Polymerase subunit 2a (3/3), and cyclin D1 (2/3). In contrast, bcl-2 mRNA levels increased after flavopiridol in most cases (7/8), which could represent a compensatory mechanism of leukemic blasts to avoid apoptotic cell death. Our preliminary studies indicate that flavopiridol is cytotoxic in poor-risk and refractory acute leukemia. Studies are underway to determine if down-regulation of any putative target genes correlates with pharmacologic data or clinical responses.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 20039-20039
Author(s):  
M. Bhutani ◽  
A. K. Pathak ◽  
G. Sethi ◽  
B. B. Aggarwal

20039 Background: Agents that can block activated STAT3, a central player for proliferation, cell survival and chemoresistance, have a potential as therapeutic agents for the treatment of Multiple Myeloma (MM). Capsaicin, a spicy component of hot pepper, is a homovanillic acid derivative that preferentially induces certain cancer cells to undergo apoptosis. We have previously shown that Capsaicin blocked activation of NF-kappa B in human myeloid ML-1a cells. In this study we evaluated the effect of Capsaicin on STAT3 in MM cells. Methods: We used U266, a well-characterized MM cell line, which constitutively expresses activated STAT3. MM cells treated with Capsaicin were subjected to western blot analysis with specific antibodies to STAT3, tyrosyl phosphorylated STAT3 and STAT5. The effect of Capsaicin on nuclear-cytoplasmic compartment of STAT3 was studied by immunocytochemistry. The antiproliferative effect of Capsaicin was determined by the MTT assay and the effect on the cell cycle was determined by flowcytometry. Apoptosis of cells was measured using the Live and Dead assay. To determine the downstream targets like antiapoptotic proteins (Bcl-xL, Bcl-2, and Survivin), and cell cycle-regulators (cyclin D1) immunoblot analysis of Capsaicin treated cells was performed. Results: Capsaicin suppressed the constitutive activation of STAT3 in human MM cells in a dose- and time-dependent manner, prior to cell death. Capsaicin’s effect on STAT3 was specific as STAT5 was unaffected. Capsaicin depleted nuclear pool of STAT3 in U266 cells. Abrogation of constitutive STAT3 phosphorylation in MM cells induced G1 cell cycle arrest. The antiapoptotic proteins BCl-xl, suvivin, cyclin D1, and Bcl-2, which are encoded in target genes of STAT3, were down regulated by Capsaicin, followed by induction of apoptosis through activation of caspase-3. We further demonstrated that low dose combined Capsaicin and thalidomide/ bortezomib treatment triggered synergistic cytotoxicity. Conclusions: These findings suggest that the antitumor activity of Capsaicin is at least partially due to inhibition of STAT3 pathway and provide a basis for potential application of Capsaicin for treatment of relapsed and refractory MM. No significant financial relationships to disclose.


Sign in / Sign up

Export Citation Format

Share Document