scholarly journals Exogenous hydrogen sulfide exerts proliferation, anti-apoptosis, migration effects and accelerates cell cycle progression in multiple myeloma cells via activating the Akt pathway

2016 ◽  
Vol 36 (4) ◽  
pp. 1909-1916 ◽  
Author(s):  
Dong Zheng ◽  
Ziang Chen ◽  
Jingfu Chen ◽  
Xiaomin Zhuang ◽  
Jianqiang Feng ◽  
...  
FEBS Letters ◽  
2012 ◽  
Vol 586 (24) ◽  
pp. 4311-4318 ◽  
Author(s):  
Anamika Dhyani ◽  
Adriana S.S. Duarte ◽  
João A. Machado-Neto ◽  
Patricia Favaro ◽  
Manoela Marques Ortega ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1694-1694
Author(s):  
Carolina Elosua ◽  
Purificacion Catalina ◽  
Brian A Walker ◽  
Nicholas J Dickens ◽  
Athanasia Avramidou ◽  
...  

Abstract Multiple Myeloma (MM) is a malignancy depicted by clonal expansion of plasma cells in the bone marrow. There are two broad genetic subtypes of multiple myeloma as defined as hyperdiploid multiple myeloma (H-MM), characterized by trisomies of chromosomes 3, 5, 7, 9, 11, 15, 19, and 21, and nonhyperdiploid multiple myeloma (NH-MM) associated with primary translocations involving the immunoglobulin heavy chain (IgH). These two subtypes of multiple myeloma have two different molecular pathogenesis given that characteristic changes of each have been already observed. In order to contribute to the understanding of this malignancy and to unveil the different molecular pathogenesis, our interest is focused on Human Multiple Myeloma Cell lines (HMCLs), as a model, and a broad but specific group of enzymatic proteins: the Kinases. Kinase hyperactivity or lack of it often results in disregulation of cellular pathways involved in proliferation and survival. In our study, we describe the patterns of genetic lesions and molecular pathogenesis of 11 HMCLs with Single Nucleotide Polymorphism (SNP)-based mapping arrays from Affymetrix Human Mapping 500K array set. This technique allows the examination and identification of copy number changes, bi-allelic deletions and the identification of loss of heterozygosity (LOH) due to loss and uniparental disomy, as well as gene localization and identification. The 11 HMCLs utilized are characterized for their structural alterations and not by hyperdiploidy. In addition, so as to fulfill the selection criteria, a minimum of 3 cell lines must present the alterations cited below. The most frequently identified alterations were located as follows: Previously described gains were observed in 1q, 7q, 8, 11q, 18, 19, and 20q; but also found at 4q. The bi-allelic deletions were ascertained on 3p. Similarly, we identified the regions of hemizygotic deletions on 1, 2q, 6q, 8q, 9p, 11q, 12, 13q, 14q, 17p, and 20p. In addition, described regions of homozygotic deletions were detected on 1p, 6q, 8p, 13q, 16q, and 22q, and furthermore located on 2q, 3, 4q, 9, 10q, 12p, and 20p. Finally, the uniparental disomies (UPDs) obtained were traced on 1q, 4q, 8q, 10q, and 22q. These identified alterations are affecting a series of enzymatic genes belonging to targeted pathways. Within the chromosomes 1, 10, 11, 14, and 16 we have localized kinases that are part of the PI3K/AKT pathway, which affect to a number of intracellular and extracellular myeloma growth cytokines. In the chromosomes 1, 6, 12, and 19 we identified a series of Cyclin-Dependent Kinases that are critical regulators of cell cycle progression and RNA transcription, since they regulate and control the cyclins, cell cycle regulatory proteins, which can provoke dysregulation and abnormally accelerated cell cycle progression. And finally on chromosomes 1, 2, 14, 21, and 22 we observed certain Aurora and related kinases, as another family of the cell cycle regulators and often aberrantly activated in human tumor cells, they facilitate transit from G2 through cytokinesis. These mutated kinases may be potential targets for therapeutics. Our data demonstrates the genomic complexity of multiple myeloma enhancing our understanding of the molecular pathogenesis of the disease and the importance of the HMCLs as a model.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5160-5160
Author(s):  
Marc J Braunstein ◽  
Craig M Scott ◽  
Shannon Behrman ◽  
Peter Walter ◽  
Peter Wipf ◽  
...  

Abstract Background: Multiple myeloma (MM) remains incurable, primarily due to the development of dose-limiting toxicity and/or resistance to previously effective drugs. A rational approach is to develop new strategies that are synergistic with existing agents. Recent research indicates that inhibition of the molecular chaperone heat shock protein (Hsp) 70 may facilitate resistance to inhibitors of cellular protein quality control such as bortezomib in MM. This is indicated by upregulation of Hsp70 expression after treatment of MM with inhibitors of the proteasome or Hsp90. Endothelial progenitor cells (EPCs) are bone marrow (BM)-derived hematopoietic precursor cells that augment tumor neovascularization and govern MM severity, suggesting that EPCs are a potential target for novel antimyeloma strategies. In this study, we examined the antimyeloma effects of MAL3-101, a member of a new class of inhibitors of Hsp 70, on MM cell lines and patient-derived MM cells and EPCs. We also determined the synergy between the antimyeloma effects of MAL3-101 and inhibition of the proteasome and Hsp90. Methods: MM cell lines (NCI-H929, RPMI-8266, and U266), BM-derived MM cells, EPCs from untreated patients, control PBMCs, and BM cells were treated with MAL3-101, the proteasome inhibitor MG-132, the Hsp90 inhibitor 17-AAG, or DMSO, alone and in combination. Cell survival and apoptosis were assessed by the MTS assay and Annexin V-PI staining, respectively. Cell cycle progression and immunoblots were performed by standard methods. Synergistic effects were evaluated by determining the combination index (CI) using CalcuSyn software. Results: H929 cells were most sensitive to MAL3-101, with peak cytotoxicity at 40 h (IC50 8.3 μM). In contrast, MAL3-51, a less potent Hsp70 modulator, was less effective. Furthermore, FACS analysis showed that exposure to 10 μM MAL3-101 caused a time-dependent increase in apoptosis and inhibition of cell cycle progression, indicated by a 3-fold increase in the sub-G0/G1 phase and a 2.5-fold decrease in the G2/M phase populations in H929 cells. Also, immunoblot analysis showed a time-dependent increase in the cleavage of caspase-3 and PARP by MAL3-101 in H929 cells, indicating induction of apoptosis. When H929 cells were exposed to a range of concentrations of MAL3-101 or MG-132 alone and in combination, we found that the IC50 for the agents in combination decreased to 0.008 μM. Notably, when examined alone, each compound was ineffective at the IC50 concentration of the combined compounds. The synergistic cytotoxic actions of MAL3-101 and MG-132 on H929 cells occurred over a 10-fold range of concentrations (0.01–0.1 μM) (CI < 1), and also resulted in synergistic apoptosis. Similarly, we found that the combination of MAL3-101 and 17-AAG also decreased the IC50 for 17-AAG, from 0.4 μM to 0.03 μM. These results are consistent with the prediction that the antimyeloma effects of Hsp90 inhibition, which causes upregulation of Hsp70 gene expression, would be potentiated by simultaneous inhibition of Hsp70. Synergistic antimyeloma effects of MAL3-101 and MG-132 were also observed as a decrease in viability of MM cells by 33% ± 8 (mean ± SD; P=.02) and 44% ± 10 (P=.07), by MAL3-101 and MG-132, respectively, but by 75% ± 4 (P=.001) when the agents were combined. In addition, combined MAL3-101 and MG-132 reduced viability of BM EPCs, by 10% ± 14 (P=.7) and 16% ± 17 (P=.3), respectively, but in combination by 60% ± 7 (P=.001). The specificity of MAL3-101’s effect on MM cells was indicated by a lack of cytotoxicity in control cells. When intracellular and secreted IGs were quantitated, we observed that the relative amount of IG secretion was highest in H929 cells, which also demonstrated the highest sensitivity to MAL3-101-induced cytotoxicity, indicating that the sensitivity of MM cells to Hsp70 inhibition most likely arises from the added cellular stress of producing and secreting monoclonal IGs; however, there was no evidence for an upregulation of an unfolded protein response in these cells by XBP-1 mRNA splicing. Conclusions: These results show for the first time that exposure to the Hsp70 inhibitor MAL3-101, a new Hsp70 modulator, sensitizes MM tumor and EPCs to proteasome and Hsp90 inhibition. These data support a preclinical rationale for inhibition of Hsp70 function, either alone or in combination with other agents, as a novel therapeutic strategy for MM.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2811-2811
Author(s):  
Carolina Terragna ◽  
Sandra Durante ◽  
Daniel Remondini ◽  
Giovanni Martinelli ◽  
Francesca Patriarca ◽  
...  

Abstract Abstract 2811 Poster Board II-787 Introduction The recurrent translocation t(4;14)(p16;q32) occurs in less than 20% of patients with newly diagnosed Multiple Myeloma (MM) and is associated with a poor clinical outcome following either conventional or high-dose chemotherapy. Recently, it has been reported that patients carrying t(4;14) are prognostically heterogeneous and that the novel agents bortezomib and lenalidomide may overcome the poor prognosis related to this cytogenetic abnormality. In the present study, we analyzed the gene expression profile of patients who carried or not t(4;14) and were primarily treated with a bortezomib-based regimen. Patients and methods Two hundred thirty six patients with MM who received a combination of bortezomib-thalidomide-dexamethasone (VTD) as first-line therapy were evaluated for the presence at diagnosis of t(4;14). Of these, 41 patients (17.3%) were t(4;14) positive. On an intention-to-treat basis, the rate of CR and near CR (nCR) to VTD induction therapy among patients carrying t(4;14) was 41%, a value higher than the 29% observed among t(4;14) negative patients. In 218 patients for whom data on t(4;14), del(13q) and del(17p) were available, the differential gene expression of CD138+ enriched plasma cells was evaluated by means of expression microarray using the Affymetrix platform. The analysis was performed in t(4;14) negative patients and patients carrying t(4;14), either alone or combined with other abnormalities; t(4;14) negative patients included those with del(13q) alone and with any of these abnormalities. Results In 27 patients, t(4;14) was associated with either del(13q) (24 patients) or del(17p) (3 patients); the remaining 14 patients carried t(4;14) alone. The expression profiles of patients carrying either t(4;14) alone or t(4;14) combined with del(13q) significantly clustered apart when compared with those of cytogenetic negative patients. Similarly, the expression profiles of patients with del(13) alone clustered with those of cytogenetic negative patients. De-regulated expression of similar molecular pathways was demonstrated in patients carrying t(4;14) alone or combined with del(13q). Thus, the analysis of gene expression profiles according to response or no response to VTD was performed in two subgroups of patients, including those carrying t(4;14) alone or combined with del(13q) and those carrying either del(13q) alone or without cytogenetic abnormalities. By comparing the lists of genes differentially expressed (P '0.05) in patients who responded (e.g. those who achieved CR+nCR) and failed to respond (NR) to VTD according to the presence or absence of t(4;14), we found that the differential expression of 3719 genes characterized CR+nCR vs NR patients in the t(4;14) positive subgroup. At the opposite, the differential expression of 3182 genes characterized CR+nCR vs NR patients in the t(4;14) negative subgroup. 271 genes which were common to the two groups of genes were excluded from the list of genes found to be differentially expressed in t(4;14) positive patients who responded to VTD. Among these patients, we observed the de-regulated expression of genes involved in cell cycle progression (e.g. MDM2, CDK6 and SMAD2), Wnt signalling pathway (e.g. FZD7, WNT10A, MMP7,WNT2B, WNT6, WNT9A and DAAM2), and Hedgehog signalling pathway (GAS1, STK36 and GLI1). Overall, genes involved in cell cycle progression resulted over-expressed, thus suggesting a more aggressive phenotype of t(4;14) positive plasma cells of responder patients; nevertheless, the overall down-regulation of genes involved in Wnt and Hedgehog signalling pathways (known to be involved in the maintenance of a putative tumoral stem cell compartment) might mitigate this phenotype and predispose t(4;14) positive plasma cells to more favourably respond to VTD induction therapy. Supported by: BolognAIL, Fondazione Carisbo, Progetto di Ricerca Finalizzata (M.C). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1808-1808
Author(s):  
Nesrine Lajmi ◽  
Julia Templin ◽  
Sara Yousef ◽  
Tim Luetkens ◽  
Stefanie Spock ◽  
...  

Abstract Abstract 1808 Background: Cancer-testis antigens belonging to the MAGE class I family of genes are commonly expressed in Multiple Myeloma (MM). Expression of MAGE class I genes is associated with an aggressive clinical course of MM and resistance to chemotherapy, suggesting that MAGE genes may confer a survival advantage on myeloma cells. MAGE-C2/CT10 is member of the MAGE class I family of genes thought to be a good candidate for cancer immunotherapy given its very frequent expression in primary myeloma. In normal cells, MAGE-C2/CT10 seems to suppress p53 expression by promoting its polyubiquitination and degradation. However, the function of MAGE-C2/CT10 in malignancies is completely unknown. We, therefore, investigated for the first time the role of MAGE-C2/CT10 in tumor cells derived from patients with MM. Material and Methods: MAGE-C2/CT10 expression was analysed by real-time PCR and western blot in myeloma cell lines (N=8) and in PBMC from healthy donors (N=8). For the functional evaluation of MAGE-C2/CT10 we decided to use myeloma cell line U-266 which constitutively expresses MAGE-C2/CT10 and a missense mutant p53 (A161T) that has partially lost its transcriptional activity. The biological role of MAGE-C2/CT10 was investigated by stably silencing its expression using lentiviral short hairpin RNA (shRNA). The effects of silencing MAGE-C2/CT10 expression on myeloma cell biology were examined by determining the number of viable or apoptotic cells using a colorimetric MTT assay and annexin-V/7AAD staining followed by flow cytometry, In addition, we measured myeloma cell proliferation and the anchorage-independent growth using a BrdU incorporation assay and a colony formation assay, respectively. Finally, we investigated cell cycle phase distribution by flow cytometry and we analyzed the expression of key molecules involved in cell cycle progression and apoptosis using a real-time PCR array as well as western blot. Results: We found MAGE-C2/CT10 to be constitutively expressed in all myeloma cells lines but not in PBMC from healthy donors. Lentivirus-mediated silencing of MAGE-C2/CT10 inhibited significantly the proliferation and the anchorage-independent growth of myeloma cells. Cell cycle analysis demonstrated that the anti-proliferative effect of MAGE-C2/CT10 silencing in U-266 was due to a 70% decrease of cells in the S phase, a cell cycle arrest at both G0/G1 and G2/M transitions and an increase in the subG0/G1 population due to an activation of apoptotic cell death. The serine-threonine checkpoint effector kinase 2 (CHK2) and its substrate, the tumor suppressor protein p53, are essential for cell cycle control, DNA repair and apoptosis. We found that the loss of MAGE-C2/CT10 expression was associated with the activation of CHK2 through phosphorylation at Thr68 as well as the activation of p53 by phosposphorylation at Ser20. Furthermore, we observed a three-fold increase in the endogenous level of p53 protein which correlated with an up-regulation of two transcriptional targets of p53, the cyclin-dependant kinase inhibitor p21WAF1 and the growth arrest and DNA-damage-inducible alpha protein (GADD45A), known to be essential for p53-induced G1 and G2 arrest, respectively. Finally, using the Human Apoptosis Profiler PCR array that contains a number of p53 target genes, we found that apoptosis induced by MAGE-C2/CT10 knockdown was due to a more than two-fold increase in the transcription of pro-apoptotic genes like BAX (Bcl2-associated × protein), BAD (BCL2-associated agonist of cell death), Cytochrome c, APAF1 (Apoptosis activating factors) as well as several caspases, which are the down-stream mediators of p53-dependant apoptosis in response to DNA damage. Conclusions: Our collected findings support an anti-apoptotic function of MAGE-C2/CT10 in MM, likely through the regulation of key molecules involved in cell cycle progression and p53-mediated apoptosis. The central role of MAGE-C2/CT10 in the biology of myeloma strongly suggest that this cancer-testis antigen represents a promising target for myeloma-specific immunotherapies or other targeted modes of therapy for MM. Disclosures: Kröger: Fresenius Biotech: Honoraria, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document