Here Is the Sub-Title:Apoptosis on Multiple Myeloma U266 Cell by Phosphorylation c-Jun of Brucine

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5112-5112
Author(s):  
Yanping Ma ◽  
Zhihua Li ◽  
Yihua Wang ◽  
Jing Feng

Abstract Abstract 5112 Objective: To investigate the Mechanism of the apoptotic effect of brucine on human multiple myeloma. Method: U266 cells (5×104) were plated in the presence or absence of the brucine (0, 0.05, 0.1, 0.2, 0.4 mg/ml) in 96 well culture plates for 24–72 h. The anti-proliferative response of brucine was assessed by MTT assay. The analysis of cell cycle of U266 cell with or without brucine was mesured by flow cytometry. The expression change of c-Jun after joining brucine, brucine and JNK specific inhibitor SP600125 was detected using RT-PCR. Results The apoptotic effect of brucine show a dose and time dependent manner. Cell cycle analysis using flow cytometry revealed accumulation of cells at sub-G0/G1 phase. The apoptosis rate separately were (4.137±0.01)%, (10.55±0.03)%, (12.31±0.04)%, (27.67±0.08)%, (29.67±0.09)% (p<0.01). Detecting c-Jun gene expression respectively after joining brucine, brucine and JNK specific inhibitor SP600125 by RT-PCR. The gray scale values were (0.7961±0.007),(0.4683±0.003). Conclusions Within the 0.4mg/ml concentration of brucine can induce apoptosis in U266 cells. Brucine by JNK signaling pathway through phosphorylation of c-Jun induced apoptosis in U266 cells. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5727-5727
Author(s):  
Wenjun Wu ◽  
Cai Wu ◽  
Fuming Zi ◽  
Yi Li ◽  
Li Yang ◽  
...  

Abstract Background : Multiple myeloma (MM) is a B cell malignant hematologic cancer. Despite the introduction of new drugs and improvement of chemotherapy, MM is still an incurable disease. Oxymatrine (OMT), the active ingredients of traditional Chinese herbal medicine sophora, has been reported to have antitumor activity. This study was to estimate the therapeutic efficacy of OMT in MM. Methods: The growth inhibition of myeloma cell lines (RPMI8226, U266, ARP-1) or primary cells by OMT was assessed by MTT assay. Apoptosis of MM cells was examined by annexin V-FITC using flow cytometry analysis. DNA content was analyzed by flow cytometry. RT-PCR and western-blot analysis were used to assess the expression of Bcl-2 family proteins and the IAP family proteins. Western blotting was also used to elucidate the signaling pathway that may mediate OMT-induced apoptosis of MM cells. Results: OMT treatment resulted in cell growth inhibition and apoptosis in primary MM cells and all tested MM cell lines in a dose-dependent manner (P <0.05). To elucidate OMT -induced MM cell apoptosis, MM cell lines were treated with or without OMT for 24h and assessed for caspase activation and signaling pathway by Western blotting. The results showed the cleavage of PARP, caspase-3, and caspase-9, and p-AKT were down-regulated after OMT treatment. The mRNA expression of survivin and HIAP by RT-PCR was down-regulated. OMT treatment at 5mM for 48h resulted in increased G-phase cells and decreased S-phase cells in MM cell lines (P <0.05). Cell cycle repressor P21 protein was up-regulated while CDK4, CDK6 and CyclinD1 expression was down-regulated. Our finding also showed a synergistic anti-MM activity of OMT and dexamethasone or adriamycin at a low does (CI<1). In addition, LC3-II expression was significantly increased both in RPMI8226 and U266 cells after treatment with OMT. However, treatment with different doses of OMT and 5 mM autophagy inhibitor 3-MA, significant increased cell apoptosis (P <0.05). Conclusion: Our findings demonstrate the anti-MM activity of OMT and indicate that OMT alone or together with other MM chemotherapeutics may be a prospective treatment for MM. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4228-4228
Author(s):  
Deepti Soodgupta ◽  
Dipanjan Pan ◽  
Grace Hu ◽  
Angana Senpan ◽  
Xiaoxia Yang ◽  
...  

Abstract Purpose This study investigated alpha 4 beta 1/ Very Late Antigen-4 (α4β1/ VLA-4)-integrin targeted nanotherapy approach to deliver a new lipase-labile prodrug. Experimental Design A phospholipid-based MYC-MAX inhibitor prodrug (MI1-PD) was synthesized, and its inherent anti-proliferate potency was compared to the lipid-free compound (MI1) using mouse multiple myeloma (MM) cell line (5TGM1). VLA-4-targeted perfluorocarbon (PFC) nanoparticles binding to 5TGM1 cells was measured and compared to biomarker expression assessed with flow cytometry using antibodies. The efficacy of MI1-PD incorporated into non-targeted and VLA-4-targeted PFC NP exposed to 5TGM1 cells was assessed with MTT assays, Annexin V and cell cycle analysis. Results MI1-PD was more potent by several orders of magnitude than its free drug counterpart in culture. Targeted NP binding correlated well with biomarker expression assessment by flow cytometry in 5TGM1 cells. Non-targeted NPs had no appreciable binding to 5TGM1 cells. High anti-MM potency of MI1-PD was noted in VLA-4-targeted NPs compared to the non-targeted NPs demonstrating that the efficacy was dependent on expression of the targeted biomarker to afford particle-to-cell drug delivery. Conclusions These results suggest the feasibility of an improved integrin VLA-4- targeted nanotherapy approach to deliver a lipase- labile prodrug construct, MI1-PD. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4947-4947
Author(s):  
Bao-An Chen ◽  
Xiao-hui Cai ◽  
Jun Wang ◽  
Chong Gao ◽  
Jia-hua Ding ◽  
...  

Abstract Abstract 4947 Objective: The aim of this study was to investigate the expression of survivin and the apoptosis induced by DNR and BrTet in the leukemic cells K562/A02. Methods: In a typical experiment, the K562/AO2 cells were treated with daunorubicin (DNR), 5-bromotetrandrine (BrTet), or DNR and BrTet for 48 hours, and the cells treated without any drugs were used as control group. Cell proliferation was analyzed by MTT assay. Cells apoptosis and the concentration of DNR within the cells were measured by Flow cytometry (FCM). The expressions of mRNA and protein of survivin were determined by semi-quantitative reverse transcription PCR (RT-PCR) and Western blot, respectively. Results: The results of MTT assay indicated that DNR and BrTet were both able to inhibit the proliferation of K562/AO2 cells in dose-dependent manner. The fresh evidence from flow cytometry showed that a higher apoptosis rate could be induced and a higer concentration of DNR could be detected in K562/AO2 cells by DNR and BrTet as compared with those by DNR or BrTet in the same concentrations(P<0.01). RT-PCR revealed that the expression of survivin mRNA, a higer expression in K562/AO2 cells with acquired resistance to adriamycin than that in parent K-562 cells, decreased in the DNR and BrTet group (P<0.05), but there was no obvious change in other groups(P>0.05). Western bolt demonstrated that the expression of survivin protein was much lower in the DNR and BrTet group(P<0.05). Conclusion: BrTet could increase the concentration of DNR and reverse the multidrug resistance(MDR) in the K562/AO2 cells. Survivin may play an important role in apoptosis induced by DNR. Survivin could be a target for the treatment of MDR in haematopoietic malignancies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2984-2984
Author(s):  
Sadeaqua S Scott ◽  
Marc J Braunstein ◽  
Christopher Lange ◽  
Christopher Roman ◽  
Eric LP Smith ◽  
...  

Abstract Abstract 2984 Background: Multiple myeloma (MM), a neoplasm of committed B-lymphocytes within the bone marrow (BM), is the second most common hematologic malignancy in the U.S. Despite prolonged median survival with anti-myeloma strategies aimed at the tumor and its BM microenvironment, MM remains invariably fatal. Endothelial progenitor cells (EPCs) are CD133+/KDR+ cells that originate in the BM and play a key role in supporting tumor growth and MM progression. Using X-chromosome inactivation and RT-PCR analyses, we previously found EPCs from MM patients to be clonally restricted and to display clonotypic IG heavy-chain gene rearrangements identical to the same patients' tumor cells (Braunstein et al., 2006). Based on the shared genetic identity that we and others have demonstrated between tumor cells and EPCs in MM patients, the present study explored the hypothesis that, similar to hemangioblasts, which are CD133-expressing precursors to adult hematopoietic and endothelial cells, EPCs may be a source of vascular and MM progenitor cells. Since hemangioblasts are known to exist predominately in the quiescent phases of the cell cycle, in this study we also examined the cell cycle status of CD133-expressing BM cells from MM patients in order to gain insight into their hemangioblastic traits. Methods: BM aspirates were acquired from MM patients under IRB approval. EPCs (>98% vWF/CD133/KDR+ and CD38-) from BM aspirates of MM patients were outgrown on laminin-coated flasks as previously described. The spleen colony assay was used to determine the stem cell capacity within BM-derived EPCs by i.v. injection into NOD/SCID mice. The spleens and BM of mice were harvested 2–4 weeks later. Cells were analyzed by immunofluorescence (IF) and flow cytometry. Hoechst 33342 (Hst) and Pyronin Y (PY) were used to measure the cell cycle status of CD133+ cells using FACS analysis. Results: Two to four weeks following i.v. injection of MM EPCs, human cell surface marker expression detected by flow cytometry within mouse BM and spleen cells shifted from CD133+/CD45-lo, a progenitor cell phenotype, to CD133−/CD45-hi, a more differentiated phenotype, suggesting the ability of MM EPCs to differentiate in vivo. Cell cycle analysis of the CD133+ population in BM cells of MM patients showed that these cells were predominantly non-cycling. On average, 60.5% of CD133+ cells were found to be in the G0/G1 phase of the cell cycle, as indicated by low levels of IF staining with Hst and PY. Conclusions: CD133+ cells are strong candidates as precursors to MM tumor and vascular progenitor cells. As quiescent cells are non-dividing, they often escape cytotoxic effects of chemotherapy, resulting in relapse, and therefore, identification of these cells is critical. Ongoing studies include the engraftment of CD133+ cells in the subcutaneous NOD/SCID gamma xenotransplant model and their growth in response to anti-myeloma strategies; results of these studies will be discussed. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5632-5632 ◽  
Author(s):  
Jinsong Hu ◽  
Li Lei ◽  
Yanmeng Wang ◽  
Ke Wang ◽  
Xiaoyan Hu ◽  
...  

Abstract Multiple myeloma (MM) is a B-cell malignancy characterized by the clonal proliferation of plasma cells in the bone marrow (BM). CD147, also known as extracellular matrix metalloproteinase inducer (EMMPRIN), is a type I transmembrane glycoprotein that belongs to the immunoglobulin superfamily. CD147 is expressed in a variety of tissues, and is involved in a number of physiological and pathological processes. With regard to MM, it was recently documented that the aberrantly elevated expression of CD147 has been tightly correlated with MM cell colonization and proliferation. However, it is still unclear what mechanism is involved in the dysregulation of CD147 expression in MM. In this study, we found that CD147 can be up-regulated by interleukin-6 (IL-6), which is derived from either autocrine or paracrine sources and plays an essential role in the malignant progression of MM. When serum-starved MM cell lines RPMI8226, MM1.S and NCIH929 were stimulated with 20ng/mL of IL-6 for 24 hours, we found that the expression of CD147 on MM cell membrane was significantly up-regulated, as determined by flow cytometric analysis. Moreover, when using the IL-6 autocrine MM cell line U266, CD147 level was found to be continuously increased in serum-starved MM cells in a time-dependent manner. Interestingly, the increase of CD147 in U266 cells was found to be abrogated when using neutralizing antibody to block the secreted IL-6. Next, we investigated the molecular mechanisms involved in the IL-6-mediated upregulation of CD147 in MM cells. We confirmed that IL-6 can activate the JAK/STAT3 signaling pathway, inducing the phosphorylation of STAT3 at Tyrosine 705 in RPMI8226 and MM1.S cells. Further studies using ChIP analysis demonstrated that there is a STAT3 binding site at the position −659 to −650 in the promoter of the CD147 gene. In addition, when inhibiting of STAT3 phosphorylation with a specific inhibitor S3I-201, the CD147 expression in MM cells was accordingly decreased. Finally, we found that siRNA mediated knock-down of CD147 in RPMI8226 and MM1.S cells can decrease the proliferation and migration of MM cells induced by IL-6 and Cyclophilin A (CypA). In conclusion, our data provide evidence that IL-6 can up-regulate CD147 expression in a STAT3-dependent manner, and offer a compelling rationale for exploring this axis as a therapeutic target for MM. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4817-4817
Author(s):  
Wenming Wang ◽  
Jing Wang ◽  
Mingyi Chen ◽  
Yaoxian Liang ◽  
Zhengqian Li ◽  
...  

Abstract Multiple myeloma (MM) is a malignant disorder characterized by the proliferation of a single clone of plasma cells derived from B cells. Previous studies have demonstrated that both gene-specific hypermethylation and global hypomethylation characterizes the multiple myeloma epigenome. 5-azacytidine as a DNA methylation inhibitor has therapeutic efficacy in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Nevertheless,the effects of 5-azacytidine on MM remains unclear. We used RT-PCR to detect the expression of PTPL1 and used MS-PCR to determine the methylation status of PTPL1 in MM cell lines and after 5-azacytidine treatment. ELISA-like reaction was used to detect global DNA methylation level. The cytotoxic activity of 5-azacytidine was tested using cell viability and apoptosis assays. Flow cytometry was used to detect cell cycle after 5-azacytidine treatment. Our experiments discovered that the PTPL1 gene was hypermethylated in the U266 and H929 cell lines, and the expression of PTPL1 mRNA could be re-inducible by 5-azacytidine. 5-azacytidine also inhibited the proliferation of multiple myeloma cell lines U266 and H929 in a time- and dose-dependent manner, induced G2/M cell cycle arrest and caspase-dependent apoptosis. But in our study 5-azacytidine increased the methylation level for both cell lines. Our study showed that PTPL1 was epigenetically regulated in MM which can be reversed by 5-azacytidine, and highlights 5-azacytidine is a potential therapeutic candidate for MM, but additional studies are needed to determine the effects of genome-wide methylation changes in MM. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3457-3457 ◽  
Author(s):  
Eric D. Hsi ◽  
Roxanne Steinle ◽  
Balaji Balasa ◽  
Aparna Draksharapu ◽  
Benny Shum ◽  
...  

Abstract Background: To identify genes upregulated in human memory B and plasma cells, naïve B cell cDNA was subtracted from plasma cell and memory B cell cDNA. One gene that was highly expressed in plasma cells encodes CS1 (CD2 subset 1, CRACC, SLAMF7), a cell surface glycoprotein of the CD2 family. CS1 was originally identified as a natural killer (NK) cell marker. Monoclonal antibodies (mAbs) specific for CS1 were used to validate CS1 as a potential target for the treatment of multiple myeloma (MM). Methods: Anti-CS1 mAbs were generated by immunizing mice with a protein comprising of the extracellular domain of CS1. Two clones, MuLuc63 and MuLuc90, were selected to characterize CS1 protein expression in normal and diseased tissues and blood. Fresh frozen tissue analysis was performed by immunohistochemistry (IHC). Blood and bone marrow analysis was performed using flow cytometry with directly conjugated antibodies. HuLuc63, a novel humanized anti-CS1 mAb (derived from MuLuc63) was used for functional characterization in non-isotopic LDH-based antibody-dependent cellular cytotoxicity (ADCC) assays. Results: IHC analysis showed that anti-CS1 staining occurred only on mononuclear cells within tissues. The majority of the mononuclear cells were identified as tissue plasma cells by co-staining with anti-CD138 antibodies. No anti-CS1 staining was detected on the epithelia, smooth muscle cells or vessels of any normal tissues tested. Strong anti-CS1 staining was also observed on myeloma cells in 9 of 9 plasmacytomas tested. Flow cytometry analysis of whole blood from both normal healthy donors and MM patients showed specific anti-CS1 staining in a subset of leukocytes, consisting primarily of CD3−CD(16+56)+ NK cells, CD3+CD(16+56)+ NKT cells, and CD3+CD8+ T cells. Flow cytometry of MM bone marrow showed a similar leukocyte subset staining pattern, except that strong staining was also observed on the majority of CD138+CD45−/dim to + myeloma cells. No anti-CS1 binding was detected to hematopoietic CD34+CD45+ stem cells. To test if antibodies towards CS1 may have anti-tumor cell activity in vitro, ADCC studies using effector cells (peripheral blood mononuclear cells) from 23 MM patients and L363 MM target cells were performed. The results showed that HuLuc63, a humanized form of MuLuc63, induced significant ADCC in a dose dependent manner. Conclusions: Our study identifies CS1 as an antigen that is uniformly expressed on normal and neoplastic plasma cells at high levels. The novel humanized anti-CS1 mAb, HuLuc63, exhibits significant ADCC using MM patient effector cells. These results demonstrate that HuLuc63 could be a potential new treatment for multiple myeloma. HuLuc63 will be entering a phase I clinical study for multiple myeloma.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1674-1674 ◽  
Author(s):  
Nicholas Burwick ◽  
Anne-Sophie Moreau ◽  
Xiaoying Jia ◽  
Xavier Leleu ◽  
Judith Runnels ◽  
...  

Abstract BACKGROUND: Multiple myeloma (MM) is a plasma cell malignancy that depends on interactions with the bone marrow (BM) microenvironment for growth and survival. In turn, adhesion of MM cells to the BM stroma provides a mechanism of resistance from standard chemotherapeutic agents. Recently, our lab has shown that by disrupting this adhesion using a selective CXCR4 inhibitor named AMD3100, MM cells are more sensitive to the proteasome inhibitor Bortezomib (Ghobrial lab, unpublished data). CXCR4 has been a particularly attractive target because its ligand SDF-1 is known to induce p42/44 MAPK, AKT, and the down-stream anti-apoptotic protein bad in MM cells, leading to increased MM growth and survival. Until recently, CXCR4 was thought to be a canonical receptor for the SDF-1 ligand. However, a second chemokine receptor for SDF-1 was subsequently discovered and named CXCR7. CXCR7 is a novel chemokine receptor that is important in cell adhesion, growth and survival in several tumor types. However, the role of CXCR7 in multiple myeloma (MM) has yet to be explored. Furthermore, the ability of SDF-1 ligand to regulate MM function via CXCR7 has not been studied. METHODS: The MM cell lines (U266, MM1.S, RPMI, OPM2, OPM1) were used. After informed consent was obtained, primary bone marrow samples from MM patients were collected. CD138 positive mononuclear cells were isolated by microbead selection. The expression of CXCR7 on MM cell lines and patient samples was confirmed using flow cytometry and RT-PCR analysis. For functional in vitro and ex-vivo assays, the CXCR7 selective antagonist 733 was used (ChemoCentryx Inc., Mountain View, CA). RESULTS: Here we show that CXCR7 was expressed on all tested MM cell lines and primary patient samples as demonstrated by flow cytometry and RT-PCR. Furthermore, CXCR7 was found to regulate SDF-1 induced MM cell adhesion, as demonstrated by in vitro assays using a small molecule compound specific for CXCR7 (733). The CXCR7 antagonist showed significant inhibition of adhesion of MM cell lines and patient samples to fibronectin, endothelial cells and stromal cells, with 50% reduction of adhesion at 5nM of the CXCR7 inhibitor, and with similar activity compared to 20uM of AMD3100 (CXCR4 inhibitor). However, unlike CXCR4, CXCR7 did not effect trans-well migration to SDF-1 chemokine. Interestingly, both receptors were found to be important for trans-endothelial migration of MM cells. Moreover, pre-treatment with 733 reduced homing of MM cells to the BM niche in vivo. Previous studies have failed to show signaling in response to CXCR7 in many tumor types. Here, we demonstrate that treatment with 733 inhibited SDF-1 induced pERK and pAKT, ribosomal pS6Kinase, pGSK3, pSTAT3, pFAK and pPAK signaling pathways, confirming a role for CXCR7 in facilitating SDF-1 signaling. This effect was further confirmed using immunofluorescence. To investigate whether CXCR7 and CXCR4 interact directly, we examined the effect of 733 and AMD3100 on CXCR4 expression and found that AMD3100 significantly inhibited CXCR4 expression, while 733 had no effect on CXCR4 expression, even in the presence of SDF-1. The CXCR7 inhibitor had no effect on the survival of MM cells using MTT and flow cytometry analysis, while high doses of 733 (1uM) had modest inhibition of proliferation. Interestingly, 733 prevented the growth advantage induced by 30nM SDF-1 at 24 hrs. CONCLUSION: Together, these results demonstrate the importance of CXCR7 in regulating MM adhesion and homing, and highlight the differential effects of CXCR4 and CXCR7 in regulating SDF-1 signaling in MM, thus providing a rationale for targeting the SDF-1/CXCR7 axis in MM.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4158-4158
Author(s):  
Jing Liu ◽  
Ji-liang Hu ◽  
Yang He ◽  
Bi-Wei Shi ◽  
Wei-Xin Hu

Abstract Abstract 4158 Lycorine displays various biological functions including remarkable anti-tumor effect. We previously reported that lycorine induced anti-leukemia effect via arresting cell cycle and inducing apoptosis on human acute promyelocytic leukemia (APL) cell line HL-60. To explore the molecular mechanism how lycorine induced HL-60 cell apoptosis, cDNA microarray was used to investigate the expression profile of 494 apoptosis-associated genes. Real-time RT-PCR, Western blotting and immunocytochemistry were used to analyze the expression of related genes, as well as the modification and distribution of related proteins in the presence of lycorine. The results showed that 89 differential genes were expressed significantly (Cy3:Cy5 > 2 or < 0.5) among all the 494 apoptosis-related genes. 78 genes were up-regulated and 11 genes were down-regulated. We are particularly interested in the expression increase of p21 (9.271 folds) and TNF-α (8.242 folds). Furthermore, we found that lycorine could down-regulate p21-related gene expression including Cdc2, Cyclin B, Cdk2 and Cyclin E, promote the truncation of Bid protein and the release of cytochrome c from mitochondria, decrease the phosphorylation of IκB, block the nuclear import of NF-κB and down-regulate expression of survivin. This study revealed that lycorine decreased HL-60 cells survival through p21-mediated cell cycle arrest and stimulation of TNF-α signaling pathway which induced apoptosis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4896-4896
Author(s):  
Qingxian Bai ◽  
Qifa Liu

Abstract Abstract 4896 BackgroundF Multiple myeloma(MM) is a malignant plasma disease, which is characterized as high relapse rate and high resistance to chemotherapy. Curcumin is a polyphenol derived from the rhizome of Curcuma spp. It possesses diverse pharmacologic actions, such as antitumor, anti-inflammatory,anti- oxidation properties .Curcumin has the property of inhibit multiple tumor cell lines, in which included multiple myeloma cell. The real mechanism is not completely clear yet. We explored the mechanisms of curcumin on human multiple myeloma cell lines (RPMI8226 and H929), and investigated whether the combination of curcumin and adriamycin(Adr) has a synergistic effect. MethodsF The effect of curcumin on proliferation of RPMI8226 and H929 was observed with MTT assay. The synergetic effect of curcumin and Adr was analyzed by median-effect principle. Cell cycle distribution and apoptosis were studied with flow cytometry. Expression of surviving, bcl-2, bax mRNA was detected by RT-PCR. ResultsF Curcumin could inhibit the proliferation of RPMI8226 and H929 cells in a time- and dose-dependent manner. The IC50 values for RPMI8226 and H929 cell line were 12.15 μmol/L,17.24μmol/L respectively. The combination of curcumin and Adr showed synergistic effect even at low concentration of Adr. Apoptotic ratio of treated cells was significantly higher than untreated controls (36.9% vs 10.6%, p<0.05). Cells treated with curcumin showed cell cycle arrest at G2/M phase. Curcumin upregulated expression of survivin, bcl-2, while bax mRNA was significantly downregulated. ConclusionF Curcumin could suppress the proliferation of multiple myeloma cells and induce apoptosis. Adr combining with curcumin can show synergistic effect at low concentration of Adr. The mechanism of curcumin's antitumous effect might be related to down-regulation of surviving, bcl-2 mRNA and up-regulation of bax mRNA. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document