scholarly journals C6-ceramide treatment inhibits the proangiogenic activity of multiple myeloma exosomes via the miR-29b/Akt pathway

2020 ◽  
Author(s):  
Liping Liu ◽  
Qinmao Ye ◽  
Langni Liu ◽  
Ji Chen Bihl ◽  
Yanfang Chen ◽  
...  

Abstract Background: The increased bone marrow angiogenesis is involved in the progression of multiple myeloma (MM) with the underlying mechanism poorly understood. Cancer-released exosomes could play an important role in the pathological angiogenesis through exosomal microRNAs (miRs) delivery. MiR-29b has been reported in regulating the tumor angiogenesis. Methods: In this study, we explored the role of C6-ceramide (C6-cer, a Ceramide pathway activator) in the angiogenic effect of MM exosomes and its potential mechanism. MM cells (OPM2 and RPMI-8226) treated with C6-cer were studied for its effects on the endothelial cell (EC) functions . Results: Our results showed that exosomes released from MM cells treated by C6-cer ( Exo C6-cer ) significantly inhibited the proliferation, migration and tube formation of ECs. For mechanism studies, we found that the level of miR-29b was increased in ECs treated by exo C6-cer , while mRNA and protein expressions of Akt3, PI3K and VEGFA were decreased in ECs, indicating the involvement of Akt pathway. Furthermore, downregulation of miR-29b by inhibitor administration could prevent the exo C6-cer -induced cell proliferation, migration and angiogenesis of ECs, accompanied with the increased expressions of Akt3, PI3K and VEGFA. Conclusions: Collectively, our data suggest that exo C6-cer -mediated miR-29b expression participates in the progression of MM through suppressing the proliferation, migration and angiogenesis of ECs by targeting Akt signal pathway.

2020 ◽  
Author(s):  
Liping Liu ◽  
Qinmao Ye ◽  
Langni Liu ◽  
Ji Chen Bihl ◽  
Yanfang Chen ◽  
...  

Abstract Background: The increased bone marrow angiogenesis is involved in the progression of multiple myeloma (MM) with the underlying mechanism poorly understood. Cancer-released exosomes could play an important role in the pathological angiogenesis through exosomal microRNAs (miRs) delivery. MiR-29b has been reported in regulating the tumor angiogenesis. Methods: In this study, we explored the role of C6-ceramide (C6-cer, a Ceramide pathway activator) in the angiogenic effect of MM exosomes and its potential mechanism. MM cells (OPM2 and RPMI-8226) treated with C6-cer were studied for its effects on the endothelial cell (EC) functions . Results: Our results showed that exosomes released from MM cells treated by C6-cer ( Exo C6-cer ) significantly inhibited the proliferation, migration and tube formation of ECs. For mechanism studies, we found that the level of miR-29b was increased in ECs treated by exo C6-cer , while mRNA and protein expressions of Akt3, PI3K and VEGFA were decreased in ECs, indicating the involvement of Akt pathway. Furthermore, downregulation of miR-29b by inhibitor administration could prevent the exo C6-cer -induced cell proliferation, migration and angiogenesis of ECs, accompanied with the increased expressions of Akt3, PI3K and VEGFA. Conclusions: Collectively, our data suggest that exo C6-cer -mediated miR-29b expression participates in the progression of MM through suppressing the proliferation, migration and angiogenesis of ECs by targeting Akt signal pathway.


2020 ◽  
Vol 18 (1) ◽  
Author(s):  
Liping Liu ◽  
Qinmao Ye ◽  
Langni Liu ◽  
Ji Chen Bihl ◽  
Yanfang Chen ◽  
...  

2020 ◽  
Vol 19 (4) ◽  
pp. 745-749
Author(s):  
Hongqing Zhu ◽  
Yejun Si ◽  
Yun Zhuang ◽  
Meng Li ◽  
Jianmin Ji ◽  
...  

Purpose: To identify the biological function of phosphoserine aminotransferase 1 (PSAT1) in regulating cell proliferation and apoptosis in multiple myeloma (MM).Methods: The mRNA and protein levels of PSAT1 were determined using quantitative real-time polymerase chain reaction (PCR) and western blotting, respectively. Cell proliferation was measured using CCK-8 assay.Results: PSAT1 mRNA and protein expression levels were significantly increased in MM cell lines when compared to control cells. Moreover,  downregulation of PSAT1 inhibited MM cell proliferation and induced cell apoptosis, whereas overexpression of PSAT1 promoted MM cell  proliferation and suppressed cell apoptosis. Further analysis demonstrated that the underlying mechanism was via regulation of PI3K/AKT pathway.Conclusion: The results identified a novel role for PSAT1 in the progression of MM, which may provide a therapeutic and a new anticancer target for the therapy of MM. Keywords: Multiple myeloma, PSAT1, Cell proliferation, PI3K/AKT pathway


2020 ◽  
Vol 18 (3) ◽  
pp. 292-296
Author(s):  
Wu Hong ◽  
Zhu Guihua ◽  
Su Yizhou

Bortezomib is an effective drug for the treatment of multiple myelomas. However, its long-term effectiveness is limited by the development of drug resistance. Oridonin, a natural diterpenoid purified from Rabdosia rubescens, has the potential to diminish resistance to bortezomib. To examine the mechanism underlying the diminution of bortezomib-resistance in multiple myeloma by oridonin, we have created a bortezomib-resistant cell line of multiple myeloma cells (RPMI-8226R). Using MTT assay, apoptosis assays and western blot analysis, we evaluated and compared the effects of oridonin on the cell viability, proliferation, apoptosis, and protein expression levels in bortezomib-resistant (RPMI-8226R) and bortezomib-sensitive (RPMI-8226) myeloma cells. The results show that oridonin sensitized multiple myeloma cells leading to increased apoptosis rate via regulating the PTEN/PI3K/AKT pathway. Furthermore, oridonin activated the expression of PTEN, which is a negative regulator of the PI3K/AKT pathway, while inhibitinged the expression of p-Akt. These results demonstrated that oridonin might be a useful natural compound in bortezomib-resistant multiple myeloma treatment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4039-4039
Author(s):  
Chunyan Sun ◽  
Yu Hu ◽  
Xiaomei She ◽  
Zhangbo Chu ◽  
Jun Fang ◽  
...  

Abstract Abstract 4039 MicroRNAs (miRNAs) are non-coding small RNAs that negatively modulate protein expression at a post-transcriptional level and are deeply involved in the pathogenesis of a variety of cancers. Two miRNAs, miR-15a and miR-16, which act as putative tumor suppressor by targeting the oncogene BCL2, have been implicated in cell cycle, apoptosis and proliferation. Here we investigated the possible role of miR-15a/miR-16 in the angiogenesis of multiple myeloma (MM). Using stem-loop quantitative reverse transcription-PCR analysis, we showed that miR-15a/miR-16 are significantly underexpressed in primary MM cells as well as MM cell lines (RPMI 8226, ARH-77, OPM-2, KM3, U266 and NIH929). The aberrant expression of miR-15a/miR-16 were detected especially in advanced stage multiple myeloma. The expression of miR-15a and miR-16-1 were remarkably lower in stage III MM patients (n=23), whereas it were significantly higher in healthy individuals (n=18) and stage 2, II MM patients (n=14) (P<0.01 and P<0.001, respectively). In human MM cell lines and normal plasma cell, expression of miR-15a/miR-16 inversely correlated with the expression of vascular endothelial growth factor (VEGF). Consistently with the proposed role of miRNAs as key regulators of angiogenesis, here we identified VEGF-A as a target for miR-15a and -16. Enforced miR-15a or miR-16 expression reduced VEGF-A protein level and the luciferase reporter assays demonstrated that miR-15a and -16 specifically suppress expression of VEGF-A by directly interacting with its 3′-untranslated region. Moreover, Ectopic overexpression of miR-15a and -16 led to decreased pro-angiogenic activity of MM cells. Conditioned medium of pri-miR-15a- and pri-miR-16- transfected RPMI 8226 cells inhibited human bone marrow microvascular endothelial cell (BMEC-1) proliferation, chemotactic motility and capillary formation in vitro as compared with conditioned medium of scramble probe-transfected RPMI 8226 cells (P<0.05). Finally, miR-15a/miR-16 was shown to greatly inhibit the process of tumor formation in an animal model. Infection of lentivirus-miR-15a or lentivirus-miR-16 can significantly inhibit the xenograft tumor growth in nude mice. The average tumor volume after 4 weeks for GFP-transfected cells was 372.5 mm3. While the average tumor volume for miR-15a-transfected cells was 47 mm3 (P<0.05) and for miR-16-transfected cells was 93.6 mm3 (P<0.05). Of interest, neoangiogenesis analysis by immunohistochemistry staining of anti-CD31 showed that there were significant reductions in microvessel density present in the miR-15a group and miR-16 group as compared to control (P<0.05). Take together, our findings suggested that miR-15a and miR-16 could play a role in the tumorigenesis of MM at least in part by modulation of angiogenesis through targeting VEGF-A. These findings have therapeutic implications and may be exploited for future treatment of MM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4019-4019
Author(s):  
Tommasina Guglielmelli ◽  
Emilia Giugliano ◽  
Vanessa Brunetto ◽  
Sokol Rrodhe ◽  
Giuseppe Saglio

Abstract Abstract 4019 Introduction. mTOR is a serine-threonine protein kinase that plays a central role in regulating critical cellular processes. Consistent with its primary target being the translation machinery, mTOR is predominantly localized in the cytoplasm. However, a nuclear localization of mTOR has been found in rhabdomyosarcomas and HCT8 colon carcinoma cells. Furthermore, mTOR becomes nuclear in HEK293 cells treated with leptomycin B, a specific inhibitor of nuclear export receptor Crm1, suggesting that mTOR may be a cytoplasmic-nuclear shuttling protein. Aims of this study is to evaluate cellular localization of mTOR in multiple myeloma (MM) cell lines and primary MM cells and to evaluate the role of pomalidomide (CC-4047) in regulating the mTOR pathway. Methods. We used OPM-2 and RPMI-8226 MM cell lines and primary MM cells. For primary myeloma specimens, plasmacell population was positively selected by the immunomagnetic method using CD138+ microbeads. Proliferation was evaluated by MTT assay on OPM-2 and RPMI-8226 cells following incubation with pomalidomide at concentrations ranging from 0.01 to 10 μM. Apoptosis was assessed by flow cytometry for the detection of annexin V-positive cells in both MM cell lines and in plasmacells from 3 MM patients. Cellular localization of mTOR protein was also evaluated using a confocal scanning microscopy in RPMI-8226 and OPM-2 cells and in plasmacells from 4 MM patients in basal conditions and after pomalidomide treatment. Immunohistochemistry with antibody against phospho-mTOR was performed on bone marrow sections of 92 MM patients. Furthermore, RPMI-8226 and OPM-2 cells untreated or treated with the drug, were fractionated and both cytoplasmic and nuclear fractions were analysed by Western blotting with specific antibodies for mTOR and pospho-mTOR. Results. MTT assay performed on MM cell lines demonstrated that pomalidomide has a dose-dependent activity. Pomalidomide 1 μM at 48 hours inhibited proliferation of OPM-2 and RPMI-8226 cells with 50% and 40% decrease in cell numbers, respectively. Only minor increase of apoptosis could be detected in RPMI-8226 and OPM-2 cells incubated with pomalidomide at varying concentrations for 24, 48 and 72 hours. Pomalidomide 1 μM was effective in plasmacells from 3 MM patients at 24 hours with 23%, 33% and 26% annexin-V positive cells (versus 11%,18% and 3% of annexin-V positive cells cultured with media alone, respectively). Immunofluorescence assays with mTOR antibody, demonstrated that mTOR protein is distributed throughout the cytoplasm and the nucleus at baseline in both MM cell lines and in plasmacells of 3 out 4 MM patients. A clearly increase of the nuclear mTOR protein was detected after pomalidomide treatment in RPMI-8226 and OPM-2 cells (10 μM at 48 hours) and in plasmacells from 3 MM patients (1 μM at 24 hours) (2 with nuclear mTOR localization at baseline and 1 without it). Immunohistochemistry performed on bone marrow sections evidenced that 41 out 92 MM samples (44.4%) stained positive for cytoplasmic phospho-mTOR. A nuclear phospho-mTOR staining was also demonstrated in 11 cases (12%). All patients but one showed both nuclear and cytoplasmic phospho-mTOR staining. Cytoplasmic and nuclear distribution of mTOR and pospho-mTOR was also evidenced by Western blotting in RPMI-8226 and OPM-2 cells. As expected, the mTOR and phospho-mTOR protein levels were significantly higher in the cytoplasm when compared to the nucleus. Treatment with pomalidomide 10 μM at 48 hours increased the nuclear mTOR and phospho-mTOR expression levels in the nucleus with a concomitant decrease of the cytoplasmic phospho-mTOR protein amount. Conclusions. In RPMI-8226 and OPM-2 cell lines and in a fraction of primary MM cells, mTOR is distributed throughout the cell cytoplasm and in some nucleus. The anti-myeloma activity of pomalidomide may be mediated by the downregulation of the mTOR pathway with a nuclear shuttling of mTOR protein and a reduction of the cytoplasmic phospho-mTOR. Further studies are needed to establish the mechanism of mTOR shuttling, to evaluate the role of pomalidomide in regulating the mTOR pathway and to assess the potential synergism between pomalidomide and mTOR inhibitors. Disclosures: Guglielmelli: Celgene: This study was sponsored by Celgene Other, Honoraria, Research Funding.


Author(s):  
Xiaorui Luan ◽  
Shang Li ◽  
Jun Zhao ◽  
Junyu Zhai ◽  
Xiaojing Liu ◽  
...  

Abstract The underlying mechanism of the chemokine-C receptor 7 (CCR7) that leads to aberrant trophoblast migration and invasion in recurrent spontaneous abortion (RSA) remains unknown. CCR7 is considered crucial for migration and invasion and has been associated with the risk of miscarriage. However, the functional role of CCR7 in RSA is not fully understood. Our study found that CCR7 mRNA and protein abundance were significantly decreased in the villous from RSA patients compared with healthy controls. Knockdown of CCR7 caused a significant reduction of migration and invasion in JAR and JEG-3 cells. Meanwhile, CCR7 functioned as a positive upstream factor of the AKT pathway contributing to the expression of GATA2, promoting trophoblast migration, and invasion via MMP2. Notably, a decreased abundance of CCR7 was positively correlated with the phosphorylation of AKT and with an abundance of GATA2 and MMP2 in human villous specimens of RSA compared with the control group. CCL19, a ligand of CCR7, could promote trophoblast migration and invasion by activating the deregulation of the CCR7-mediated pathway in RSA. We are convinced that CCR7 and its downstream factors may be possible mechanisms for the pathogenesis of RSA.


2020 ◽  
Vol 7 (5) ◽  
pp. 192152
Author(s):  
Yao Zhang ◽  
Changqing Zhen ◽  
Qing Yang ◽  
Bing Ji

Multiple myeloma (MM) is an incurable disease with relatively high morbidity and mortality rates. Great efforts were made to develop nuclear factor-kappa B (NF-κB)-targeted therapies against MM disease. However, these treatments influence MM cells as well as normal cells, inevitably causing serious side effects. Further research showed that NF-κB signalling promotes the survival of MM cells by interacting with JNK signalling through growth arrest and DNA damage-inducible beta (GADD45β), the downstream module of NF-κB signalling. The GADD45β-targeted intervention was suggested to be an effective and MM cell-specific treatment. However, the underlying mechanism through which GADD45β promotes the survival of MM cells is usually ignored in the previous models. A mathematical model of MM is built in this paper to investigate how NF-κB signalling acts along with JNK signalling through GADD45β and MKK7 to promote the survival of MM cells. The model cannot only mimic the variations in bone cells, the bone volume and MM cells with time, but it can also examine how the NF-κB pathway acts with the JNK pathway to promote the development of MM cells. In addition, the model also investigates the efficacies of GADD45β - and NF-κB-targeted treatments, suggesting that GADD45β-targeted therapy is more effective but has no apparent side effects. The simulation results match the experimental observations. It is anticipated that this model could be employed as a useful tool to initially investigate and even explore potential therapies involving the NF-κB and JNK pathways in the future.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 24-24
Author(s):  
Shirong Li ◽  
Jing Fu ◽  
Jun Yang ◽  
Markus Y Mapara ◽  
Christophe Marcireau ◽  
...  

Introduction: RAS oncogenes are the most frequently mutated gene family in human cancers. 50% of newly diagnosed multiple myeloma patients carry a RAS/MAPK pathway mutation, with a rising percentage in the relapsed situation1. Thus, targeting RAS mutations in multiple myeloma will increase therapeutic efficiency and potentially overcome drug-resistance. Unfortunately, RAS mutations have been considered "undruggable" due to a lack of traditional small molecule binding pockets on the proteins. Therefore, key component in the RAS/MAPK pathway may represent an alternative therapeutic target for MM. Germinal center kinase (GCK), also named mitogen-activated protein kinase kinase kinase kinase 2 (MAP4K2), is an upstream activator in the MAPK pathway. Indeed, we recently discovered the critical role of GCK in RAS mutated (RASmut) MM cell survival and growth. GCK knockdown in RASmut MM cells induced MM cell growth inhibition both in vitro and in vivo. However, the detailed mechanism is yet to be defined. Methods and Results: Our previous data showed that GCK knockdown induces MM cell growth inhibition, associated with the blockage of MKK4/7-JNK phosphorylation and the downregulation of critical transcriptional factors (TFs) including IKZF1/3, BCL-6, and c-MYC proteins. To confirm that GCK knockdown downregulates IKZF1/3 etc at protein level but not mRNA level, we conducted real-time PCR on GCK knockdown MM cells and compared the expression of GCK and TFs to the empty vector (EV) infected MM cells. Results showed that shRNA induced GCK silencing only led to the significantly decreased GCK mRNA, however, did not affect IKZF1 and c-MYC expressions at mRNA level. Consistent with the effects of GCK knockdown, the GCK inhibitor TL4-12 dose-dependently downregulated IKZF1 and BCL-6 proteins, inhibited MM cell proliferation and induced cell apoptosis. IKZF1/3 are the key targets of the immunomodulatory drugs (IMiDs), which are the backbone of MM therapy. IMiDs bind to cereblon (CRBN) and induce IKZF1/3 protein degradation, which subsequently lead to MM cell growth inhibition. Importantly, our data showed that IMiDs-resistant RPMI-8226 MM cells have high expression of GCK. GCK knockdown and inhibition induced IKZF1 downregulation, triggered growth inhibition and cell apoptosis in RPMI-8226 cells, suggesting that GCK regulates IKZF1 degradation via a CRBN-independent mechanism. To confirm this hypothesis, we silenced CRBN in N-Rasmut H929 MM cells by shRNA lentiviral infection and examined the response to IMiDs and GCK inhibitor. CRBN knockdown was confirmed by western blotting. CRBN silencing in H929 cells resulted in lenalidomide (LEN) resistance, evidenced by the WTS proliferation assay. In contrast, CRBN silencing failed to rescue N-Rasmut H929 MM cells from TL4-12 induced proliferation inhibition and IKZF1 downregulation, confirming that GCK regulated IKZF1 and cell growth is independent of CRBN. Conclusion: Taken together, our data demonstrated that GCK inhibition induces cell growth inhibition and triggers apoptosis especially in RASmut MM cells. Importantly, GCK inhibitor downregulates IKZF1 via a CRBN-independent mechanism. Our findings thus provide a rationale for the clinical evaluation of targeting GCK in RASmut MM patients and further mechanistic insight into the role of GCK in MM tumorigenesis as well as drug resistance. GCK inhibitors may represent a novel therapy for the treatment of RASmut MM patients, especially those who are resistant to IMiDs as well as with refractory or relapsed MM. References Walker, B.A., et al. Mutational Spectrum, Copy Number Changes, and Outcome: Results of a Sequencing Study of Patients With Newly Diagnosed Myeloma. J Clin Oncol33, 3911-3920 (2015). Disclosures Marcireau: Sanofi: Current Employment. Lentzsch:Karyopharm: Research Funding; Mesoblast: Divested equity in a private or publicly-traded company in the past 24 months; Janssen: Consultancy; Sorrento: Consultancy; Caelum Biosciences: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Celularity: Consultancy; Magenta: Current equity holder in private company; Sanofi: Research Funding.


2012 ◽  
Vol 111 (suppl_1) ◽  
Author(s):  
Aili Guan ◽  
Hui Gong ◽  
Yong Ye ◽  
Jianguo Jia ◽  
Guoping Zhang ◽  
...  

It is well established that angiotension II (Ang II) is an important regulator in vascular homeostasis. Under certain conditions, Ang II could exert anti-angiogenic effects in cardiovascular system. However, the potential mechanism is unclear. P53 has been reported to suppress angiogenesis by promoting hypoxia-inducible factor-1 (Hif-1α) degradation. This study was conducted to determine the contribution of P53 and the underlying mechanism to the anti-angiogenic effect of Ang II. Angiogenesis was determined by tube formation from the cardiac microvascular endothelial cells (ECs). Microvessel density and cardiac function were analyzed in mice subjected to Ang II infusion (200 ng/kg/min ) or vehicle for 2 weeks. Ang II (1μM) greatly inhibited tube formation and stimulated phosphorylation and upregulation of P53 in cultured cardiac ECs. P53 inhibitor, pifithrin-α (PFT-α,3.0mg/kg), significantly reversed the inhibitory effect of Ang II on tube formation. Vascular endothelial growth factor (VEGF ) and Hif-1α has been reported as important pro-angiogenetic factors. The present study indicated that Ang II decreased VEGF concentration in cultured medium and downregulated Hif-1α expression in cultured ECs. Interestingly, Ang II also stimulated the upregulation of Jagged 1, a ligand of Notch, but it didn't affect the Delta-like 4 (Dll 4) , another ligand of Notch, expression in cardiac ECs. However, PFT-α partly abolished these effects of Ang II. These results were consistent with the study in vivo. Further research revealed that siRNA-Jagged 1 transfection in cultured ECs dramatically abolished the phosphorylation of P53 and the downregulation of Hif-1α induced by Ang II. Additionally, Ang II- induced inhibitory effect on capillary formation was blocked by siRNA-Jagged 1 transfection in cultured cardiac ECs. In conclusion, Ang II promoted the phosphorylation and upregulation of P53, and increased Jagged 1 expression, the upregulation of Jagged 1 in turn stimulated the phosphorylation of P53, which resulted in the downregulation of Hif-1α and VEGF, then induced the inhibitory effects of Ang II on capillary formation. The present data suggest that Ang II exerts anti-angiogenesis via the cooperation of P53 and Jagged 1 in vitro and in vivo.


Sign in / Sign up

Export Citation Format

Share Document