GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Material
Language
  • 1
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 109, No. 31 ( 2012-07-31)
    Abstract: Our finding here that miRNAs act as ligands able to bind and activate a receptor in a hormone-like fashion might have broader implications that could go beyond cancer, e.g., for autoimmune diseases and inflammatory diseases. These studies reveal a mechanism by which cancer cells cross-talk with surrounding immune cells and induce them to release cytokines that increase tumor growth and spread. The present study demonstrates the importance of the tumor microenvironment in cancer growth and dissemination and provides molecular targets for the development of anticancer treatments. We first determined which miRNAs are represented more frequently in the exosomes secreted by different populations of lung cancer cell lines, using NanoString technology and subsequent validation by quantitative real-time PCR. We found that miR-21, -29a, and -16 were among the most represented in cancer cell-derived exosomes. Subsequently, we used immunoprecipitation techniques to show that these three miRNAs are able to reach and bind to TLR8 in the endosomes of a cell. Using in situ hybridization with the Nuance system, we were able to demonstrate in primary human lung cancer tumors that cancer cells are the main producers of miR-29a secreted in the exosomes and that miR-29a colocalizes with macrophages, mainly at the interface of tumor and normal tissue. Next, we asked whether the binding of miRNAs to TLRs resulted in activation of the TLR-mediated signaling pathway. We observed that although miR-21 and -29a are able to activate a TLR8-mediated response (activation of the NF-κB pathway and secretion of TNF-α and IL-6), miR-16 does not. We performed an initial analysis on whether this difference might be explained by the sequence of the mature miRNAs by performing mutagenesis of specific nucleotides in the mature sequence of the miRNAs of interest. Although we showed that some nucleotides are critical for the activation of TLRs, further studies are warranted to clarify this aspect. We also showed that the binding of miR-21 and -29a to TLR7 and TLR8 induces activation of immune cells (indicated by activation of CD69) and that this effect is not mediated by binding to human TLR7. Moreover, we showed in an in vivo mouse model of lung cancer [mice injected with Lewis Lung Carcinoma (LLC) cells] that, by binding to TLR7 , cancer cell-secreted miR-21 and -29a in exosomes metastasize preferentially to the lung and reduce overall survival, compared with TLR knockout mice in which this mechanism is impaired. We also found that drugs affecting exosome secretion by cancer cells significantly reduce the metastatic potential of LLC cells, and this effect can be rescued by injecting tumor-bearing mice with exosomes secreted by LLC cells. MiRNAs are expressed aberrantly in most types of cancer ( 1 , 2 ). They are secreted from cells within exosomes and are transferred from cell to cell; miRNAs regulate gene expression in the recipient cells by binding to their target messenger RNAs ( 3 ). MiRNAs also can interact with proteins ( 4 ). For example, members of the TLR family (murine TLR7 and human TLR8) bind viral single-stranded RNA sequences on immune cells, leading to cytokine production ( 5 ). Circulating miRNAs could represent ligands of TLR7 and TLR8 that are released by tumor cells and are involved in intercellular communication in the tumor microenvironment. MicroRNAs (miRNAs) are small noncoding RNAs possessing gene-regulatory functions and are secreted by cancer cells by means of microvesicles called “exosomes.” Here we show that miRNAs contained in cancer cell-secreted exosomes can be transferred to surrounding immune cells and can bind to Toll-like receptors ( TLR7 in mice and TLR8 in human). By binding to TLRs, miRNAs induce cytokine secretion by the immune cells, leading to a prometastatic inflammatory response ( Fig. P1 ). Here, we report that miRNAs can act as hormones by binding to receptors.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2012
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 123, No. 10 ( 2013-10-1), p. 4144-4157
    Type of Medium: Online Resource
    ISSN: 0021-9738
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2013
    detail.hit.zdb_id: 3067-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Cell, Elsevier BV, Vol. 40, No. 11 ( 2022-11), p. 1442-
    Type of Medium: Online Resource
    ISSN: 1535-6108
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2078448-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4467-4467
    Abstract: Introduction: Multiple myeloma (MM) is the second most prevalent hematologic malignancy. Approximately, 80,000 people have died of the disease in the United States and 25,000 new cases are registered every year. Majority of patients develop resistance to current therapeutic treatments and die within 5-10 years of diagnosis. Thus, need of novel therapeutic intervention is extremely urgent. Although the field of oncolytic virotherapy (OV) based on using viruses with natural or engineered tumor selective replication to intentionally infect and kill tumor cells has been extensively explored for the treatment of solid tumors, only few data are available for the treatment of hematopoietic malignancies. Our laboratory was one of the first to show that OV using Reovirus can be an effective therapeutic strategy for the treatment of MM in vitro and in MM patients. In this work we aim at exploring the possibility of using genetically engineered HSV1 (Herpes Simplex Virus) for the treatment of MM. HSV1 is an enveloped, double stranded DNA virus. Engineered HSV1 (HSVQ) has both copies of viral gene important in viral replication in normal cells viz., ICP 34.5 gene deleted and has one copy of GFP inserted into viral ICP6 gene. Such engineered virus has been used for cancer cell selective killing in preclinical and clinical studies for the treatment of several types of solid tumors including melanoma and glioblastoma multiforme. In this study, we investigated the biological and preclinical impact of HSVQ on MM cell in vitro and in vivo. Method: Recombinant HSVQ was amplified in African green monkey kidney epithelial Vero cells, purified by sucrose density gradient centrifugation and titrated by plaque assay on Vero cells. Several MM cell lines (MM1.S, U266, RPMI8226, L363, NIH-H929) were infected with HSVQ at Multiplicity of Infection (MOI) 0.01 to 5. Fluorescence microscopy and flow cytometry analysis were used to assess MM cell infectivity with the virus. RT-PCR was performed to detect presence of viral genome in MM cell lines. Viral replication assays were also performed. Cell proliferation and apoptotic assays including MTT Assay, Tryphan Blue exclusion test, LIVE/DEAD cell viability staining and Annexin/7-AAD assays were done to determine viability of virus infected MM cells. Western Blot analysis was carried out to determine endoplasmic reticulum (ER) stress response mediated by ERK, Hsp90, Bip/GRP78, Hsp40 and apoptosis in HSVQ treated MM cells. Total bone marrow (BM) cells obtained from MM patients were infected with HSVQ and multi parametric flow analysis was performed to determine infectivity and specific killing of CD138+ MM cells by the virus. To study in vivo anti-tumorigenic properties of HSVQ, 12.5 x106 GFP/Luc + MM1.S or NIH-H929 cells were subcutaneously injected into the right flank of 20 NOD-SCID mice. Two weeks after injection, mice with comparable size tumors were randomly divided (5 animal for each treatment group) and treated twice with 1x107PFU (Plaque Forming Unit) HSVQ for 2 weeks or with saline. Tumor growth was measured to determine anti tumorigenic effect of HSVQ on MM tumors. Results and Conclusion: Fluorescence microscopy and flow cytometry revealed that MM cell lines can be effectively infected with and killed by HSVQ even at MOI as low as 0.1. Under such conditions, Western Blot analysis revealed increased BAX expression, decreased BCL2 expression and cleavage of Caspase 3 and PARP indicating apoptosis of virus infected cells. Interestingly, multi parametric flow analysis revealed that HSVQ specifically infects and kills CD138+ MM plasma cells in a total population of BM cellular fraction isolated from MM patients. Moreover in vivo preclinical data show that HSVQ dramatically reduces tumor volume (p 〈 0.001) in both MM.1S and NIH-H929 xenograft mouse models. Thus, from the preliminary observations, it can be concluded that HSVQ can selectively infect and induce apoptosis in myeloma cells. Mechanisms of HSVQ replication in MM cells and induced MM cell killing are being currently investigated. Here for the first time we are providing clear evidences that HSVQ can infect and specifically kill MM cells supporting the idea of the use of HSV for the treatment of MM. Moreover, since the backbone of HSVQ can be further engineered, it can be used to specifically deliver anti-angiogenic and anti-inflammatory genes to MM cells for the treatment of MM. Disclosures Hofmeister: Arno Therapeutics, Inc.: Research Funding; Celgene: Research Funding; Karyopharm Therapeutics: Research Funding; Incyte, Corp: Membership on an entity's Board of Directors or advisory committees; Janssen: Pharmaceutical Companies of Johnson & Johnson: Research Funding; Signal Genetics, Inc.: Membership on an entity's Board of Directors or advisory committees; Takeda Pharmaceutical Company: Research Funding; Teva: Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1453-1453
    Abstract: AML is a clonal hematopoietic disorder characterized by genetic and epigenetic alterations. The prognosis of AML is poor highlighting the urgent need for novel therapeutic approaches. Targeting aberrant DNA hypermethylation by using hypomethylating drugs such as decitabine has been recently investigated in AML. Our group reported a relatively effective (47% complete response) single agent decitabine treatment schedule for older AML patients. These results suggested the opportunity to capitalize on this relatively effective and non-toxic treatment by incorporating this compound into novel molecularly targeted approaches. Recent data indicate that targeting nuclear exporter proteins is a novel therapeutic strategy to overcome cancer. In particular, CRM1/XPO1 is the only nuclear exporter involved in the active transport of the majority of tumor suppressor proteins (TSP) [e.g. p53 and FOX03A] out of the nucleus resulting in their inactivation. We recently reported the anti-leukemic activity of oral SINE CRM1/XPO1 Antagonists in AML. SINEs displayed potent anti-proliferative properties, induced apoptosis, cell-cycle arrest and myeloid differentiation in AML cell lines and patient blasts. In addition, treatment of leukemic mice with oral SINE significantly prolongs their survival. By crossing the lists of genes known to be regulated by DNA methylation in AML with the ones whose nuclear transport is XPO1 dependent, we identified important TSPs such as FOXO3A and p21. Here we hypothesize that treatment of AML cells with decitabine will increase the transcri ption and expression of a subset of TSPs (including FOX03A and p21) whose nuclear anti-leukemic effects could be enhanced by blocking their XPO1 mediated nuclear export using the clinical stage oral SINE (Selinexor). Thus, we expect that the sequential treatment of decitabine followed by Selinexor will be more effective than each drug alone. To confirm this hypothesis first we treated the AML line OCI-AML3 cells with decitabine (500nM) overnight (ovn) followed by Selinexor (200nM, 2 fold lower than IC50) for an additional 24 hours (hrs) and measured cell proliferation using WST-1 assay. Controls include: 1) DMSO ovn +DMSO 24 hours, 2) Selinexor ovn +selinexor 24 hours (200 nM) and 3) decitabine ovn + decitabine 24 hrs (500nM). AML cells that were treated first with decitabine followed by Selinexor exhibited a higher cytotoxicity (absorbance (Abs) 0.7) than cells treated with either decitabine (Abs 1.36), Selinexor (Abs 1, p=0.006) or DMSO (Abs 1.7). Similar results were observed with the MV4-11 cell line. Next we measured the candidate TSPs (FOX03A and p21) mRNA and protein expression in OCI-AML-3 and MV4-11 cell lines after 24 hrs of decitabine treatment. We found a significant up-regulation of p21 in decitabine versus DMSO treated cells (MV4-11, Fold change (FC) 4.67±1.4; OCI-AML3, FC 3.98±1.19, p 〈 0.05). We also detected a modest up-regulation of FOXO3A in both cell lines treated with decitabine when compared to the DMSO controls (MV4-11, FC 2.56±0.74 and OCI-AML3, FC 1.5±0.23, p 〈 0.05). These results were confirmed also at the protein level by using western blot. Next, we asked whether ectopic up-regulation of p21 (mimicking decitabine effects) in OCI-AML3 cells could re-capitulate the decitabine enhancing antileukemic effects of Selinexor. Overexpression of p21 followed by Selinexor (200nM) for 24 hrs resulted in a larger decrease of cell proliferation (Abs 0.5) with respect to controls (Abs 0.7, p 〈 0.05) using the WST assay. Similar results were observed for the MV4-11 cell line. Finally, we tested the efficacy of the sequential decitabine–Selinexor in vivo using the MV4-11 xenograft model. Treatment began one week after leukemic cell inoculation in 4 different cohorts; 1) Vehicle, 2) decitabine i.p. twice weekly (BIW) (0.4mg/kg); 3) Selinexor BIW (20mg/kg by oral gavage) and 4) decitabine BIW i.p (0.4 mg/kg) followed by Selinexor (10 mg/kg BIW). We found no difference in median survival time (MST) between vehicle and decitabine only treated mice. As expected, Selinexor alone treated mice have significantly improved MST at 36.5 days vs. 28.5 days, vehicle, p= 〈 0.01). Most importantly, the sequential treatment of decitabine followed by Selinexor significantly improved MST compared to Selinexor alone 47 vs 36.5, p=0.008). These pre-clinical results hold great promise for the use of this combination in human clinical trials in AML. Disclosures: Schacham: Karyopharm: Membership on an entity’s Board of Directors or advisory committees. Kauffman:Karyopharm Therapeutics Inc.: Employment, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees, Patents & Royalties. Garzon:Karyopharm: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2901-2901
    Abstract: Abstract 2901 FTY720 is a sphingosine analog proposed by the FDA for treating Multiple Sclerosis patients because of its immunosuppressive activity, which depends on its ability to prevent lymphocyte egress into the peripheral blood. To act as an immunosuppressive drug, FTY720 undergoes sphingosine kinase 2 (SPHK2) phosphorylation and internalization upon interaction with the sphingosine-1-phosphate receptor 1 (S1PR1). FTY720 also acts as a potent activator of protein phosphatase 2A (PP2A), a tumor suppressor found inactivated in chronic and blast crisis CML with wild type or imatinib/dasatinib-resistant BCR-ABL1, Ph+ B-ALL, KitD816V AML, Jak2V617F+ MPDs and other leukemias/lymphomas. FTY720 treatment of cell lines and primary progenitors isolated from bone marrow of patients with these malignancies, markedly suppressed leukemic cell proliferation/survival and induced apoptosis in a PP2A-dependent manner. Notably, long-term treatment with FTY720 of mice carrying these hematopoietic malignancies significantly prolonged survival and restored normal myelopoiesis without exerting any toxic effect in hematopoietic and non-hematopoietic organs. However, in vivo administration of FTY720 strongly, albeit reversibly, decreases the number of circulating B and T lymphocytes. Here we report that a synthetically phosphorylated FTY720 (FTY720-P) is unable to induce neither PP2A activation nor apoptosis of BCR-ABL1-, Jak2V617F-, or KitD816V-expressing myeloid precursors, indicating that FTY720 phosphorylation is dispensable for its anti-leukemic activity. Thus, we functionally characterize two FTY720 derivatives, QC-FTYSM and OSU-2S, which were synthesized as molecules unable to undergo SPHK2 phosphorylation. Treatment (2.5 uM; 24h) of FTY720-sensitive 32D-BCR/ABL cells with QC-FTYSM and OSU2S results in ∼80% and 40%, respectively, more efficient suppression of BCR-ABL1 expression and kinase activity than that observed with FTY720. Moreover, QC-FTYSM, OSU-2S and FTY720 (2.5uM; 0–60h) induce a progressive block of proliferation and marked induction of apoptosis of 32D-BCR/ABL cells. In fact, a 96%, 98%, and 79% decrease in viability is observed after treatment with QC-FTYSM, OSU-2S and FTY720, respectively. Notably, viability of non-transformed myeloid 32Dcl3 cells is not significantly affected by treatment with FTY720 or its derivatives. Consistent with the ability of FTY720 to induce apoptosis through rescue of PP2A activity, phosphatase assays show identical ability of FTY720, QC-FTYSM and OSU-2S to restore PP2A functionality. In fact, comparable and marked decrease in the amount of inactive Y307-phosphorylated PP2Ac was detected in 32D-BCR/ABL cells treated with FTY720 or its derivatives. To formally demonstrate that QC-FTYSM and OSU-2S lack immunosuppressive activity, we first assessed their ability to be internalized upon interaction/association with the S1PR1 receptor. Thus, cells were transduced with a GFP-tagged S1PR1 and treated with either QC-FTYSM, OSU-2S, or, as positive control, FTY720-P. Confocal microscopy revealed that treatment FTY720-P resulted in a strong S1PR1 internalization. Conversely, exposure of the cells to QC-FTYSM and OSU-2S did not alter the S1PR1 membrane localization, indicating that these molecules did not undergo SHPK2 phosphorylation. Further demonstration of the inability of these compounds to act as immunosuppressive molecules was gained upon in vivo administration of a single dose of FTY720, QC-FTYSM or OSU-2C (10 mg/kg) to wild type FVB/N mice. As expected, percentage of B220+/CD19+ circulating B-cells decreased of ∼90% in FTY720-treated animals. Conversely, the percentage of B-cells after exposure to QC-FTYSM and OSU-2S remained unchanged (≤ 1% decrease). Likewise, the number or CD4+ and CD8+ cells also was not affect by treatment with the QC-FTYSM compound. Note that effect of OSU-2S on T-cells and the toxicity profile and anti-leukemic activity of these drugs in healthy animals and mouse models of deadly leukemias (e.g. T315I+ and blast crisis CML and Ph+ ALL) as well as Ph− MPDs are currently being assessed. Altogether our data indicate that QC-FTYSM and OSU-2S represent two potentially powerful and safe drugs which could be introduced in the current therapeutic protocols for different types of hematopoietic and non-hematopoietic malignancies characterized by functional inactivation of the PP2A tumor suppressor. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Hematology ; 2007
    In:  Blood Vol. 110, No. 11 ( 2007-11-16), p. 3548-3548
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 3548-3548
    Abstract: Polycythemia vera (PV) is a myeloproliferative disorder (MPD) caused by a valine to phenylalanine mutation in the inhibitory JH2 “pseudokinase” domain of the signaling protein Janus Kinase 2 (Jak2). While Jak2 has several known regulators (e.g. SOCS1, SOCS3, PP2A, and SHP-1), the role played by these signaling molecules in the development of PV is still largely unclear. One of these regulators, the tumor suppressor phosphatase PP2A, has been found functionally inactivated in different hematologic myeloid and lymphoid malignancies characterized by the expression of constitutively activated oncogenic tyrosine kinases (e.g. BCR/ABL). To investigate the role of PP2A in the pathogenesis of Jak2 V617F+ MPDs and the potential therapeutic relevance of PP2A activating drugs (e.g. FTY720), we determined the effects of wild type and V617F Jak2 expression on PP2A activity and assessed the molecular and biological effects of FTY720 in hematopoietic precursor cell lines and/or primary lineage-negative bone marrow cells engineered to expressed either wild type or V617F Jak2 protein. Herein we report that PP2A activity is significantly reduced by about 82% and 78% (P & lt; 0.01) in mJak2 V617F-transduced growth factor-dependent and erythropoietin receptor-expressing 32Dcl3 (32D-EPO) and Ba/F3 cells, respectively, compared to MigR1-transduced controls. Furthermore, addition of the PP2A activator FTY720 (2.5μM) for 10 hours restored PP2A activity to 66% and 75% respectively compared to that of MigR1 controls. Mechanistically, we demonstrated that inactivation of PP2A was due to constitutive Jak2 activity. In fact, treatment of V617F Jak2-expressing cells with Jak inhibitor I (1μM; 10 hours) restored PP2A activity to 80% of controls in 32Dcl3 cells, while 600nM was sufficient to restore activity to 108% of controls in Ba/f3 cells. Likewise, transduction of murine lineage-negative bone marrow cells with wild-type Jak2 produced a 62% reduction in PP2A activity (P & lt;0.01), while expression of the Jak2 V617F mutant produced a 97% reduction (P & lt; 0.01). Moreover, as we previously reported for BCR/ABL, pharmacologic restoration of PP2A activity by treatment of Ba/F3 or 32D-EPO cells with the PP2A activator FTY720 (2.5μM for 10 hours) led to reduced expression and dephosphorylation of wild type and V617F Jak2. This, in turn, resulted in a 60% reduction in the colony forming ability of IL-3 cultured cells overexpressing wild type Jak2, while a 94% suppression of colony formation was evident in Jak2 V617-expressing cells maintained in the absence of cytokines. Moreover, addition of IL-3 to the FTY720-containing semisolid medium restored viability to levels similar to those of FTY720-treated wild type Jak2-expressing cells (70% suppression of colony formation upon treatment), indicating that restoration of PP2A activity counteracts cytokine-independent pathways triggered by the V617F Jak2 mutated tyrosine kinase. Altogether, these results suggest that inactivation of PP2A is essential for Jak2 (V617F mutant included) -driven cell/proliferation and survival and that pharmacologic activation of PP2A might represent a potential avenue for treatment of PV and, perhaps, of other MPDs characterized by the expression of a mutated Jak2 kinase.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 855-855
    Abstract: Abstract 855 MicroRNAs (miRs) and heterogeneous ribonucleoproteins (hnRNPs) are post-transcriptional gene regulators that bind mRNA in a sequence-specific manner. We have reported that a) hnRNP-E2 suppresses CEBPA mRNA translation and inhibits myeloid maturation of bone marrow (BM) progenitors from chronic myelogenous leukemia patients in myeloid blast crisis (CML-BCCD34+; Perrotti et al, Nat Genet 2002); and b) miR-328 expression is lost in myeloid CML-BCCD34+ progenitors (n=6) and its restored expression at physiological levels rescues granulocytic differentiation and impairs clonogenic potential of primary BCR/ABL+ blasts (Eiring et al, ASH 2007). Here we show by Northern blot, real-time PCR, and microarray analyses that miR-328 levels increase during granulocytic differentiation of normal human CD34+ and mouse Lin− BM progenitors, but not during differentiation towards erythroid, megakaryocytic or monocytic lineages. BCR/ABL uses the same MAPKERK1/2-hnRNP-E2 signaling pathway to suppress both C/EBPα and miR-328, as pharmacologic or shRNA-mediated inhibition of these molecules restored miR-328 expression in BCR/ABL+ cells. In fact, two functional C/EBPα binding sites are present in the miR-328 promoter region and C/EBPα interacts in vivo with these regulatory elements to enhance miR-328 transcription. Importantly, we also show that restored maturation of BCR/ABL+ blasts requires direct interaction of hnRNP-E2 with the C-rich regions of miR-328. Indeed, RNA-immunoprecipitation (RIP) assays demonstrated that miR-328 directly binds to hnRNP-E2 independent of the RNA-induced silencing complex (RISC). Furthermore, ectopic miR-328, but not miR-181b, resulted in decreased in vivo binding of hnRNP-E2 to the uORF/spacer region of CEBPA mRNA, thereby releasing CEBPA from hnRNP-E2 translation inhibition and rescuing C/EBPa-driven neutrophil maturation (decoy activity). Differentiation of miR-328-expressing CML-BCCD34+ blasts (88.8±2.4% post-mitotic cells) correlated with induction of C/EBPa protein expression, whereas CEBPA mRNA and hnRNP E2 protein levels remained unchanged. The existence of a direct miR-328/hnRNP-E2/CEBPA interplay was formally demonstrated in vitro using RRL-directed translation assays and in vivo using the 6.15 clone of 32D-BCR/ABL cells that do not express endogenous CEBPA mRNA and require ectopic C/EBPα (wt-uORF-CEBPA) for differentiation. Addition of miR-328, but not miR-330, to hnRNP-E2-containing RRL reactions increased newly synthesized 35S-C/EBPa levels by 〉 100%. Likewise, forced miR-328 expression in vivo resulted in decreased hnRNP-E2 binding to CEBPA mRNA, induction of C/EBPa protein but not mRNA and rescued granulocytic differentiation of 6.15-wt-uORF-CEBPA but not vector-transduced 6.15 cells. While hnRNP-E2 was not found in complex with basic RISC components (Dicer, TRBP2 and Ago2), RIP assays detected miR-328 associated to Dicer and Ago2 in miR-328-expressing cells, suggesting that it also acts through canonical RISC-dependent base-pairing with mRNA targets. Indeed, we identified the BCR/ABL-regulated PIM1 serine-threonine kinase as a bona fide miR-328 target in BCR/ABL+ cells. Ectopic miR-328 suppressed PIM1 protein but not mRNA levels, and this effect required integrity of the miR-328 binding site present in the PIM1 3'UTR. Forced expression of a wild-type but not kinase-deficient PIM1 lacking the 3'UTR into miR-328-expressing cells fully rescued BCR/ABL clonogenicity, suggesting that miR-328-induced PIM1 suppression accounts for reduced survival of miR-328-infected BCR/ABL+ blasts. To show that miR-328 acts on PIM1 in a RISC-dependent manner, we mutated the miR-328 seed sequence (miR-328-Mut) while retaining its C-rich character. Similar to wild-type miR-328, miR-328-Mut efficiently interacted with hnRNP-E2, restored C/EBPa protein expression and rescued granulocytic differentiation, but was unable to silence PIM1 in 32D-BCR/ABL cells, indicating that the C-rich character of miR-328 is essential for its decoy activity, while its seed sequence integrity is necessary for RISC-dependent pairing to mRNA targets. Thus, the discovery of dual activities for miR-328 not only adds a new layer of complexity to the mechanisms regulating CML disease progression, but also highlights the ability of miRNAs to alter mRNA metabolism by acting as molecular decoys for RNA-binding proteins. Disclosures: Cortes: Novartis: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 938-938
    Abstract: Abstract 938 KIT is a receptor tyrosine kinase (RTK) and its aberrant activities resulting from protein overexpression and/or activating mutations are associated with a number of malignancies including core binding factor (CBF) AML [e.g., patients with t(8;21) or inv(16) or molecular equivalent RUNX1/RUNX1T1 or CBFB/MYH11, respectively]. RTK inhibitors (e.g. PKC412) have been shown to suppress aberrant KIT activity and delay tumor growth, but they are active only on distinct types of KIT mutations (KITmut). Furthermore, resistance to these inhibitors, as a result of secondary mutations or KIT overexpression, is emerging. Thus, we hypothesize that direct inhibition of KIT gene transcription may be a valuable therapeutic approach to override aberrant KIT expression and activity. Here, we described the regulatory and functional role of Sp1/NFkB-miR29b feedback loop in KIT-driven leukemia that can be targeted pharmacologically. Applying chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assays (EMSA) to RUNX1/RUNX1T1-positive Kasumi-1 cells, we demonstrated that while, the Sp1/NFkB complex was enriched on KIT promoter and acted as gene transactivator thereby leading to KIT overexpression, Sp1/NFkB recruited HDAC1 and HDAC3 to miR29b regulatory region thereby epigenetically repressing miR29b. This microRNA, when expressed, targeted Sp1 and eventually decreased Sp1/NFkB-mediated gene transactivation, including that of KIT. In agreement with these, we showed that when Sp1, NFkB and HDAC1 were transiently overexpressed in Kasumi-1 cells, increased KIT expression and decreased miR29b transcription occurred. In contrast, siRNA knockdown of Sp1, NFkB and HDAC1 augmented miR29b level and decreased KIT transcription. Moreover, ectopic miR29b expression impaired Sp1/NFkB repressor complex on the promoter of endogenous miR29b, thereby resulting in re-expression of the endogenous microRNA and further inhibition of Sp1/NFkB-dependent KIT transcription. Importantly, the activity of Sp1/NFkB/HDACs complex can also be pharmacologically modulated leading to restored miR29b transcription and abrogated KIT expression. We showed that pharmacologic interference with Sp1/NFkB/HDACs using their respective inhibitors, such as bortezomib (0, 6, 20, 60 and 100nM for Sp1 and NFkB), mithramycin A (150 and 300ng/ml for Sp1), bay 11-7082 (3μM for NFkB) and OSU-HDAC42 (1μM for HDAC), upregulated miR29b at early time point (6 hours) and decreased Sp1 and in turn KIT expression in KIT overexpression cell lines (e.g., Kasumi-1) and AML patient blasts. EMSA and ChIP assay demonstrated that bortezomib or HDAC42-mediated KIT repression and miR29b upregulation occurred through the dissociation of Sp1/NFkB complex from the corresponding promoter. To further investigate the therapeutic potential of targeting KIT over-expression in leukemia, we stably expressed KIT wild type (KITwt) or KITmut (D816V) in FDC-P1 cell line (murine non-tumorigenic cells derived from myeloid precursors), and we evidenced that both KITwt and KITmut promoted cell proliferation that was overcome by bortezomib in clonogenic assay. In in vivo study, when NOD/SCID mice were engrafted with FDC-P1/KITmut cells (5×106/mouse), they developed significant splenomegaly and marrow blast infiltration through KIT overexpression. When leukemia-carrying mice were treated with bortezomib (1mg/kg) for 48 hours, we observed an obvious increase of endogenous miR29b expression and a significant reduction of KIT expression. Leukemic mice that received 1mg/kg of bortezomib twice/week for 3 weeks starting on day 21 after engraftment (n=5 mice/group) showed no evidence of splenomegaly and had a significantly longer median survival [59 days (twice/week) vs 28 days (vehicle-treated), p=0.0036] , compared to vehicle-treated mice that instead showed massive splenomegaly. Cytospin of marrow and histopathology of spleen and liver showed that vehicle-treated mice displayed extensive blast infiltration that was instead absent in bortezomib-treated mice. Altogether, our study revealed a previously unrecognized protein-microRNA regulatory network whose imbalance contributes to KIT-driven leukemia. As the aberrant activity of this network is pharmacologically targetable, this discovery may be quickly translated into the clinic as a novel therapeutic approach for KIT-driven AML and other malignancies. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 119, No. 20 ( 2012-05-17), p. 4786-4797
    Abstract: Acute graft-versus-host disease (aGVHD) remains a major complication of allogeneic hematopoietic stem cell transplant (alloHSCT), underscoring the need to further elucidate its mechanisms and develop novel treatments. Based on recent observations that microRNA-155 (miR-155) is up-regulated during T-cell activation, we hypothesized that miR-155 is involved in the modulation of aGVHD. Here we show that miR-155 expression was up-regulated in T cells from mice developing aGVHD after alloHSCT. Mice receiving miR-155–deficient donor lymphocytes had markedly reduced lethal aGVHD, whereas lethal aGVHD developed rapidly in mice recipients of miR-155 overexpressing T cells. Blocking miR-155 expression using a synthetic anti–miR-155 after alloHSCT decreased aGVHD severity and prolonged survival in mice. Finally, miR-155 up-regulation was shown in specimens from patients with pathologic evidence of intestinal aGVHD. Altogether, our data indicate a role for miR-155 in the regulation of GVHD and point to miR-155 as a novel target for therapeutic intervention in this disease.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...