Specific MiRNA Silencing by Lentivirus Mediated Antagomir Expression in Chronic Myeloid Leukemia (CML) Cells.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1017-1017
Author(s):  
Michaela Scherr ◽  
Letizia Venturini ◽  
Karin Battmer ◽  
Michael Schaller-Schoenitz ◽  
Daniel Schaefer ◽  
...  

Abstract Micro RNAs (miRNA) are small non-coding RNAs that regulate gene expression by specific hybridization to complementary sequences in the 3′ untranslated region of corresponding mRNAs. Concomitant recruitment of specific multi-protein complexes results either in inhibition of mRNA translation or mRNA degradation. miRNAs are processed in a regulated multi-step process from primary transcripts into mature miRNAs by cellular components which are also at least partially involved in the process of RNA interference (RNAi). Aberrant expression of specific miRNAs has recently been described in human lymphoma and leukemia. In particular, BCR-ABL and c-MYC dependent over-expression of the polycistronic and oncogenic miR-17-92 cluster (encoding miR-17, miR-18a, miR-19a, miR-20a, miR-19b, and miR-92) has been described in chronic myeloid leukemia (CML) cell lines, primary CD34+ cells from CML patients (Venturini et al. 2007), and in lung cancer. In BCR-ABL positive K562 cells, miR-17-92 encoded miRNAs repress luciferase activity in miRNA-specific reporter assays. In addition, lentivirus-mediated over-expression of miR-17-92 increases both cell proliferation and sensitivity to imatinib induced cell death. To analyse the function of individual miRNAs of the miR-17-92 polycistron, we generated lentivirus-based strategies to induce stable miRNA-specific loss- and gain-of function phenotypes for miR-18a, miR-19b, and miR-20a, respectively. Over-expression of miRNAs embedded within miR-30-derived sequences from an internal SFFV-LTR promoter allows isolation of K562 cells with increased miRNA expression. In contrast, expression of complementary oligonucleotides (antagomirs) from a H1 promoter located in the lentiviral 3′LTR can induce stable hypomorphic miRNA-phenotypes. In lentivirally transduced K562 cells, individual silencing of miR-18a, miR-19b, and miR-20a by the corresponding antagomirs (ant-miR-18a, ant-miR-19b, ant-miR-20a) specifically relieves miRNA-mediated reporter gene repression. Correspondingly, inhibition of miRNA-function correlates to reduced ‘miRNA’-amplification by miRNA-specific quantitative RT-PCR. Furthermore, protein expression of E2F-1, a known miR-20 target, is enhanced by lentivirally expressed anti-miR-20 in a dose-dependent manner, whereas over-expression of miR-20a reduces E2F-1 levels. Finally, combined over-expression of specific miRNAs and antagomirs reveals specific induction of cell proliferation by miR-18a but strong inhibition by miR-20a in K562 cells, respectively. In contrast, anti-miR-18a, but not anti-miR-19b, anti-miR-20a, or control antagomirs inhibits proliferation of K562 cells. These data demonstrate individual and complementary functions of miR-17-92 encoded miRNAs in CML and identify potential targets for specific therapeutic intervention on the miRNA level.

2018 ◽  
Vol 47 (3) ◽  
pp. 981-993 ◽  
Author(s):  
Yu Chen ◽  
Tongtong Wang ◽  
Jing Du ◽  
Yanchun Li ◽  
Xin Wang ◽  
...  

Background/Aims: Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm. Tyrosine kinase inhibitors (TKIs) are commonly used to treat CML; however, drug resistance of CML cells to TKIs has limited their clinical application. Shikonin, a traditional Chinese herb, has long been used to treat leukemia in China, but the roles and related molecular mechanisms of shikonin treatment in CML remain unclear. Here, we aimed to evaluate the effects of shikonin on the proliferation, apoptosis, and migration of K562 cells, a CML cell line. Methods: Firstly, K562 cell proliferation and apoptosis were tested by CCK8 assay and flow cytometry with Annexin V-FITC/PI staining. Cell migration was measured by Transwell migration assay. In addition, western blot was performed to determine the proteins (PI3K, Bax, Bcl-2, cleaved caspase-3, PTEN, p-AKT, AKT, CXCR4, SDF-1, CD44) involved in the mechanism of action of shikonin. Finally, neutrophils from peripheral blood of CML patients were obtained, and cell proliferation and apoptosis were tested by CCK8 assay and flow cytometry. Results: Shikonin reduced the proliferation of K562 cells in a time- and dose-dependent manner and promoted the apoptosis of K562 cells. Moreover, shikonin increased the PTEN level and inactivated the PI3K/AKT signaling pathway, subsequently upregulating BAX in K562 cells. In addition, shikonin could block K562 cell migration via the CXCR4/SDF-1 axis. Finally, shikonin significantly inhibited the proliferation and promoted the apoptosis of neutrophils from CML patients. Conclusion: These results demonstrated that shikonin inhibits CML proliferation and migration and induces apoptosis by the PTEN/PI3K/AKT pathway, revealing the effects of shikonin therapy on CML.


Antioxidants ◽  
2021 ◽  
Vol 10 (10) ◽  
pp. 1603
Author(s):  
Silvia Trombetti ◽  
Raffaele Sessa ◽  
Rosa Catapano ◽  
Laura Rinaldi ◽  
Alessandra Lo Bianco ◽  
...  

GATA-1 is a key regulator of hematopoiesis. A balanced ratio of its two isoforms, GATA-1FL and GATA-1S, contributes to normal hematopoiesis, whereas aberrant expression of GATA-1S alters the differentiation/proliferation potential of hematopoietic precursors and represents a poor prognostic factor in myeloid leukemia. We previously reported that GATA-1S over-expression correlates with high levels of the succinate dehydrogenase subunit C (SDHC). Alternative splicing variants of the SDHC transcript are over-expressed in several tumors and act as potent dominant negative inhibitors of SDH activity. With this in mind, we investigated the levels of SDHC variants and the oxidative mitochondrial metabolism in myeloid leukemia K562 cells over-expressing GATA-1 isoforms. Over-expression of SDHC variants accompanied by decreased SDH complex II activity and oxidative phosphorylation (OXPHOS) efficiency was found associated only with GATA-1S. Given the tumor suppressor role of SDH and the effects of OXPHOS limitations in leukemogenesis, identification of a link between GATA-1S and impaired complex II activity unveils novel pro-leukemic mechanisms triggered by GATA-1S. Abnormal levels of GATA-1S and SDHC variants were also found in an acute myeloid leukemia patient, thus supporting in vitro results. A better understanding of these mechanisms can contribute to identify novel promising therapeutic targets in myeloid leukemia.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4771-4771
Author(s):  
Guangbiao Zhou ◽  
Zheng Hu ◽  
Dapeng Liu ◽  
Fuqun Wu ◽  
Jiang Zhu ◽  
...  

Abstract STI571/Gleevec/imatinib, a rationally-designed agent that occupies the ATP-binding site of BCR-ABL and stabilizes the protein in its closed, inactive conformation, has been a remarkable success for the treatment of chronic myeloid leukemia (CML). However, a significant proportion of patients chronically treated with STI571 develop resistance because of the acquisition of mutations in the kinase domain of BCR-ABL. Furthermore, the effects of STI571 on CML patients in accelerated phase or blastic crisis are unsatisfactory since many patients relapse after transient remission. Hence, additional drugs or STI571-based combination regimens are desired to circumvent resistance and to improve response rates. Here we reported that PS-341, a proteasome inhibitor which offers great promise to patients with multiple myeloma (MM), significantly enhanced the antileukemia activity of STI571 in vitro and in vivo. We found a synergy exists between low concentrations of PS-341 (5–10 nM) and STI571 (0.1–0.2 μM) in inhibition of cell growth and induction of apoptosis in K562 cell line and CD34+ leukemic cells isolated from CML patients. In K562 cells, combined use of PS-341 and STI571 accelerated activation of caspase-3, 9, and facilitated cleavage of poly-(ADP-ribose) polymerase (PARP) as compared to those in cells treated with PS-341 or STI571 alone. Moreover, PS-341/STI571 combination resulted in potentiated degradation of BCR-ABL and downregulation of phosphorylated BCR-ABL as compared to those in mono treatment. In nude mice inoculated subcutaneously with K562 cells, treatment with PS-341 (injected intraperitoneally, ip) alone (at doses of 0.05, 0.5, 1 mg/kg/d, twice a week for 4 weeks, respectively) decreased tumor growth in a dose-dependent manner. STI571 (ip) at 10 mg/kg/d also inhibited tumor growth. Intriguingly, combinatory administration of low dose PS-341 (0.05 mg/kg/d, twice a week for 4 weeks) and STI571 (10 mg/kg/d) yielded a much more profound inhibition of tumor growth and even clearance of leukemic cells in mice compared to either monotherapy. Taken together, these results demonstrate synergic effects of PS-341 and STI571, and provide the rationale to evaluate PS-341/STI571 combination in treating CML aiming to further improve clinical outcome of patients.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2787-2787
Author(s):  
Stefan Nagel ◽  
Letizia Venturini ◽  
Corinna Meyer ◽  
Hans G. Drexler ◽  
Roderick A.F. MacLeod ◽  
...  

Abstract Homeobox genes of the NK-like familiy, including TLX1, TLX3 and NKX2-5, are ectopically activated in T-cell acute lymphoblastic leukemia (T-ALL) cells mostly via chromosomal aberrations. The pathologic function of these closely related genes is still unclear. Here we analyzed their effect on the C13ORF25 gene, containing the miR-17-92 cluster. Micro RNAs (miRNAs) are a class of small non-coding RNAs which are part of an evolutionarily highly conserved intracellular mechanism, regulating gene expression by hybridization to complementary sequences usually located in the 3′untranslated region of coding transcripts. The primary transcripts (pri-mRNA) are processed to short mature miRNAs, mediating either inhibition of mRNA translation or mRNA cleavage. Aberrant expression of specific miRNAs is involved in oncogenesis as recently described for several human malignancies. The miR-17-92 polycistron encodes miRNAs which decrease E2F1 protein expression. Transcription of both E2F1 and miR-17-92 is induced by MYC, itself a target of E2F1, generating a highly regulated interactive network. Depending on the cellular context, E2F1 performs conflicting tasks by triggering proliferation or inducing apoptosis. We investigated the expression of the miR-17-92 cluster in T-ALL cell lines. Real-time RT-PCR analysis of both miR-17-92 pri-mRNA and mature miRNAs revealed different expression levels in these cells, suggesting a possible implication of the NK-like homeodomain proteins in the regulation of the miR-17-92 cluster in T-ALL. HELA cells transfected with TLX1 or NKX2-5 expression constructs showed elevated miR-17-92 pri-mRNA expression, demonstrating an activating effect. Lentiviral-mediated overexpression of NKX2-5 in the T-ALL cell line MOLT-4 consistently showed increased miR-17-92 pri-mRNA levels and decreased E2F1 protein amounts. For functional analysis of these downstream targets, another T-ALL cell line (PEER) was lentivirally transduced with expression constructs for either miR-17-92 or E2F1, resulting in reduced or elevated E2F1 protein levels, respectively. Overexpression of miR-17-92 or E2F1 did not significantly influenced the cell proliferation. However, induction of apoptosis by treating these cells with etoposide, an inhibitor of topoisomerase II, indicated that overexpression of miR-17-92 and E2F1 resulted in enhanced and reduced cell viability, respectively, as analyzed by MTT assay. In summary, these data indicate an activatory effect of oncogenic NK-like homeodomain proteins on miR-17-92 expression, reducing E2F1 protein levels and thereby enhancing survival of leukemic T-cells.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4242-4242
Author(s):  
Jishi Wang

Abstract Objective : HO-1 is a microsomal enzyme catalyzing the first, rate-limiting step in degradation of heme, HO-1 is a inducible isoform of HO, it can be strongly induced in response to cellular stress and diverse oxidative stimuli, including its substrate heme, Many studies have convincingly shown that HO-1 is a cytoprotective and antiapoptotic enzyme. the objective of this study was to investigate the influence on the K562 cell growth and apoptosis after hemin-induced HO-1 expression, and to investigate the influence on K562 cells and imatinib-resistant CML cells after ZnPPIX-induced HO-1 inhibition. Methods: different concentrations hemin (0umol/l A20umol/l 30umol/l)was used respectively to induce HO-1 expression of cultured chronic myeloid leukemia cell line K562, then detected HO-1 mRNA expression under different time by RT-PCR, and MTT was used to detected the viability of K562 cells. In addition, we used STI571(2 μmol/L) deal with the hemin-induced cells, then confirm HO-1 protective effect against STI571 use MTT. Then ZnPPIX was used to inhibition HO-1 expression of K562 and imatinib-resistant cells, similarly, RT-PCR and MTT was used for analyzed. Results: The HO-1 mRNA was not tested when absence of hemin, 8h after treated with hemin of 20 μmol/L, we can test the HO-1 mRNA expression, and at 16h the expression is reach to the peak, 16h after treated hemin under different concentrations (10umol/l, 20umol/l, 30umol/l), we found the expression is in a dose-dependent manner. In the group of 10 umol/l and 20 umol/l, the survival of cells is significantly increased in comparison to the control and also have significantly difference in the two groups(p<0.05), in the group of 20 μmol/L, 16h to 48h after hemin-induced, the survival of cells presents a time-dependent manner. In the group of 10μmol/L and 20 μmol/L, exposure of K562 cells to STI571 resulted in a substantial decrease of cell viability in comparison to the STI571 single treatment group(p<0.05). ZnPPIX-induced HO-1 inhibition leads to induction of apoptosis in K562 cells, having significant difference with the control group(p<0.05). ZnPPIX-induced HO-1 inhibition can suppress the survival of imatinib-resistant cells(p<0.05). Conclusion: our studies have shown that hemin-induced HO-1 gene expression may promote the proliferation of K562 cells, and can against the cell apoptosis. And we found hemin-induced HO-1 gene expression can protect K562 cells against STI571-induced apoptosis, ZnPPIX-induced HO-1 inhibition leads to decreased viability of imatinb-resistant CML cells. these all indicates HO-1 may represent a novel targeting in CML.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4264-4264
Author(s):  
Liu Xiaoli ◽  
Na Xu ◽  
Qingfeng Du ◽  
Zhi Liu ◽  
Rong Li ◽  
...  

Abstract Abstract 4264 The bone marrow microenvironment supports growth, differentiation and proliferation of normal hematopoietic cells and can also contribute to malignant growth. Recently,it is considered that except for the point mutant of BCR-ABL kinase contribute to imatinib-resistant therapy for patients with chronic myeloid leukemia(CML), environment-mediated drug resistance (EM-DR) is a potential factor in imatinib resistance. Our previous studies found that Integrin, focal adhesion kinase(FAK), RhoA(a small GTPase) are important adhesion molecules,and related to imatinib resistance. But how and what they crosstalk with each other is still open to debate. In order to simulated bone marrow microenvironment, we used the major components of bone marrow microenvironment- Fibronectin (Fn) co-cultured with human leukemia K562 cells.and then K562 cells were inoculated with Fn, collagen-coated plate(Co) and suspended cultures as control(mask) group,and then treated with 0.4μM,0.8μM,1.6μM,3.2μM,6.4μM imatinib for 24h,48h and 72h, detected cell apoptosis and proliferation by MTT and AnnexinV-PI assay, examined p-FAK and Rho-GTP by Wersten Blotting and Pull down-Wersten Blotting. The data showed that compared to the Co and mask groups, the cells growth inhibition and apoptosis in Fn co-culture group was significantly reduced. The protein expression of p-FAK and Rho-GTP was higher in the Fn group,and in time-dependent manner. When K562 cells in Fn group were transfected with 150nM siRNA-RhoA for 48h, there was no significant difference compared with the Co and mask groups. Furthermore, the above groups treated with anti-integrin monoclonal antibody (anti-CD29 mAb), we found that p-FAK was significant lower compared with without anti-CD29 mAb in the Fn group; but there was no significant difference of Rho-GTP compard with without anti-CD29 mAb in Fn group. These results indicate that Fn adhesion co-culture could reduce imatinib-induced cell growth inhibition and apoptosis, and this mechanism may be correlated to Rho-GTP activity,and anti-integrin monoclonal antibody could not completely block the integrin binding to Fn on K562 cells, or there was other pathway activated RhoA. The mechanism of EM-DR is complex,and which is well worth us to speculate and study. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 6 (1) ◽  
Author(s):  
Dan Wang ◽  
Huan Yang ◽  
Yun Zhang ◽  
Rong Hu ◽  
Dongjie Hu ◽  
...  

AbstractIncreased endogenous hydrogen sulfide (H2S) level by cystathionine β-synthase (CBS) has been shown to closely relate tumorigenesis. H2S promotes angiogenesis, stimulates bioenergy metabolism and inhibits selective phosphatases. However, the role of CBS and H2S in chronic myeloid leukemia (CML) remains elusive. In this study, we found that CBS and H2S levels were increased in the bone marrow mononuclear cells of pediatric CML patients, as well as in the CML-derived K562 cells and CBS expression levels were correlated with different disease phases. Inhibition of CBS reduced the proliferation of the CML primary bone marrow mononuclear cells and induced growth inhibition, apoptosis, cell cycle arrest, and migration suppression in K562 cells and tumor xenografts. The knockdown of CBS expression by shRNA and inhibiting CBS activity by AOAA decreased the endogenous H2S levels, promoted mitochondrial-related apoptosis and inhibited the NF-κB-mediated gene expression. Our study suggests that inhibition of CBS induces cell apoptosis, as well as limits cell proliferation and migration, a potential target for the treatment of chronic myeloid leukemia.


Blood ◽  
2007 ◽  
Vol 109 (10) ◽  
pp. 4399-4405 ◽  
Author(s):  
Letizia Venturini ◽  
Karin Battmer ◽  
Mirco Castoldi ◽  
Beate Schultheis ◽  
Andreas Hochhaus ◽  
...  

Abstract Aberrant micro RNA (miRNA) expression has been described in human malignancies including B-cell lymphomas. We here report BCR-ABL– and c-MYC–dependent regulation of miRNA expression in chronic myeloid leukemia (CML) using microarray analysis (miCHIP) and miRNA-specific quantitative real-time reverse transcriptase–polymerase chain reaction (miR-qRT-PCR). In 3 bcr-abl–positive cell lines, expression of miRNAs encoded within the polycistronic miR-17-92 cluster is specifically down-regulated (2- to 5-fold) by both imatinib treatment and anti–BCR-ABL RNA interference (RNAi). In addition, anti–c-MYC RNAi reduces miR-17-92 expression in K562 cells in which miRNAs can specifically repress reporter gene expression, as demonstrated by specific miRNA inhibition with antagomirs. Furthermore, lentivirus-mediated overexpression of polycistronic miRNAs in K562 cells confers increased proliferation, partial resistance against anti–c-MYC RNAi, and enhanced sensitivity to imatinib-induced cell death. Finally, we determined miR-17-92 expression in purified normal (n = 4), early chronic-phase (CP) (n = 24), and blast-crisis (BC) (n = 7) CML CD34+ cells and found up-regulation of polycistronic pri-miRNA transcripts in CML and mature miRNAs in CP but not in BC CML. These data are in accordance with a BCR-ABL–c-MYC–miR-17-92 pathway that mediates enhanced miRNA expression in CP but not BC CML CD34+ cells. Altered miRNA expression may contribute to the pathophysiology of the disease and may provide potential targets for therapeutic intervention.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1208-1208
Author(s):  
Naoto Takahashi ◽  
Wei-Feng Dong ◽  
Matthew Bainbridge ◽  
Andrea Hull ◽  
Elodie Pastural ◽  
...  

Abstract RIZ1 (PRDM2) is a tumor suppressor gene whose expression and activity are reduced by genetic and epigenetic aberrations in many cancers. In chronic myeloid leukemia (CML), blastic transformation is associated with loss of heterozygosity at 1p36, the region where RIZ1 is located, suggesting that RIZ1 has an essential role in CML pathogenesis. In CML patients and in the CML blast crisis cell line K562, we observed aberrant RIZ1 promoter methylation. To further characterize RIZ1 tumor suppressor properties that are related to CML, we analyzed RIZ1-induced changes in proliferation, apoptosis, and differentiation in CML myeloid blast crisis (CML-BC) cell lines (K562, ERY-1, YN-1, and JURL-MK1) that express low levels of endogenous RIZ1. Forced RIZ1 expression in CML-BC cell lines substantially decreased proliferation, increased apoptosis, and increased the population of cells in G2/M phase of the cell cycle. RIZ1 expression also promoted differentiation as assessed by benzidine staining in CML-BC cell lines expressing immature erythroid cell features (K562, ERY-1, YN-1) and by CD117 and CD33 expression in JURL-MK1, a CML-BC cell line expressing megakaryoblastic features. To identify genes and pathways influenced by RIZ1 expression, we used 42k cDNA microarrays to globally monitor how RIZ1 expression changes the gene expression profile of K562 cells. We discovered 25 RIZ1-regulated genes that are involved in a variety of biological processes with a subgroup of genes involved in IGF-1 signaling. The most strongly RIZ1 down-regulated genes (IGF1) and up-regulated genes (SPARC, IGFBP2) are involved in IGF-1 signaling. Using chromatin immunoprecipitation (ChIP), we determined that RIZ1 associates with IGF-1 and SPARC promoters. RIZ1 contains a PR domain that has Histone H3 lysine 9 methylation activity, which is implicated in gene repression. Using ChIP assays, we found that RIZ1 expression in K562 cells increased Histone H3 lysine 9 methylation of the IGF-1 promoter. The increased Histone H3 lysine 9 methylation is associated with decreased IGF-1 expression as monitored by RT-PCR and Western analysis. We observed autocrine production of IGF-1 in K562 cells, by culturing cells in serum free media and monitoring IGF-1 production and signaling. We detected receptor bound IGF-1 by flow cytometry, and compared the growth properties of sorted IGF-1 positive and negative cells. IGF-1 positive cells have increased number of cells in G2/M and S phase, a reduced number of cells in G1 and higher numbers of mitoses and Ki-67 positive nuclei compared with IGF-1 negative cells. RIZ1 expression in K562 decreased the amount of receptor bound IGF-1, reduced IGF-1 receptor activation, and reduced the activity of downstream IGF-1 signaling pathways. RIZ1 expression in K562 substantially reduced AKT1 and ERK1/2 phosphorylation. Our study demonstrates that RIZ1 reduces cell proliferation, increases apoptosis, and enhances differentiation of CML blast crisis cell lines. RIZ1 also controls autocrine production of IGF-1 and blocks the activity of IGF-1 signaling pathways. These activities may in part be responsible for RIZ1 tumor suppressor activity and point to the therapeutic potential of IGF-1 pathway inhibition in the acute phase of CML.


Sign in / Sign up

Export Citation Format

Share Document