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
  • American Society of Hematology  (11)
Material
Publisher
  • American Society of Hematology  (11)
Language
Years
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 2937-2937
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 2937-2937
    Abstract: Telomeresplay a vital role in DNA repair activities and protecting chromosomes from degradation[1]. Telomeres are shortened during each cell division because of the end replication problem. After several cell division, telomeres are shortened to a critical length ( 〈 3 kb), and eventually lead to overall genomic instability and triggering the DNA damage response[2-3], which is related to the cancerization of numerous cancers. The commonly used method to estimate telomere length isterminal restriction fragment (TRF) basedon southern blot, which requires thousands of cells and provides only a crude estimate of the average telomere length of all cells analyzed. However, it is believed that the frequency of critically short telomeres, rather than the mean telomere length, is a crucial factor for telomere dysfunction. Therefore, analysis of telomere length at the single-chromosome level is necessary to determine the frequency of critically short telomeres. Here, we describe the development of a high-throughput method for telomere length analysis at the single-chromosome level by using a laboratory-built high-sensitivity flow cytometer (HSFCM)[4] combined with targeted fluorescent peptide nucleic acid (PNA) probes. The unambiguous detection of the telomere signalsfrom a single chromosome was achieved via HSFCM analysis. The fluorescence intensity of single chromosome was converted to the probe number by a calibration curve, and was further transformed to the base pair number of the telomere. Five representative cell lines were analyzed to compare their telomere length and the ratio of critically short telomeres at the single-chromosome level. The potential of using frequency of short telomere for disease treatment monitoring is examined by analyzing the telomere length in lymphocyte of leukemia patients. The abundance of short telomeres was compared between healthy donors and patients with chronic myeloid leukemia (CML) to see whether it can be used to predict the efficacy of therapeutics. Moreover, the quantity of short telomeres was compared among patients with acute leukemia, patients in different phases of CML and healthy donors to see whether it can be used as a marker for disease progression prediction in CML. Reference [1] Blackburn E. H., Epel E. S., Lin J., Science,2015, 350, 1193-1198. [2]Collado M., Blasco M. A., Serrano M., Cell, 2007, 130, 223-233. [3] Deng Y., Chan S. S., Chang S., Nat. Rev. Cancer, 2008, 8, 450-458. [4] Yang LL, Zhu SB, Hang W, Wu LN, Yan XM, Anal. Chem., 2009, 81, 2555-2563. Figure 1 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: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 3333-3333
    Abstract: Frequently mutated in Acute myeloid leukemia (AML), FLT3 is considered as one of the favorable targets for treatment. The FLT3 internal tandem duplication (ITD) mutation enhances kinase activity and causes hyperactivation of downstream signal transduction. Several small molecule FLT3 inhibitors have developed, but their clinical efficacy is limited due to generation of drug resistance. In this study, we define a new mechanism of drug resistance toward tyrosine kinase inhibitors (TKIs). Initially, we found a rapid decrease in the protein level of tumor suppressor p53 in FLT3-ITD-positive MV4-11 and MOLM13 cells and peripheral blood mononuclear cells (PBMCs) from FLT3-ITD AML patients upon treatment with TKIs including sorafenib, sunitinib and quizartinib. The decrease is not caused by changes in mRNA expression as revealed by qPCR analyses but rather by accelerated protease degradation because the p53 protein was stabilized by proteasome inhibitor MG132. Furthermore, treatment of cells with RG7388, a potent disruptor of p53 and MDM2 interaction, prevented the TKI-induced p53 loss. Since MDM2 is the most important E3 ligase responsible for ubiquitination of p53, the data suggest that TKIs may lead to the degradation of p53 by promoting ubiquitination. Indeed, ubiquitination assays verified that TKIs promoted K48 poly-ubiquitination of p53. Previous studies have demonstrated that activations of FLT3 downstream signaling components such as ERKs and Akt reduce p53 protein stability through ubiquitination by activating MDM2. It is somewhat unexpected that inhibition of FLT3-ITD and its downstream signaling pathways also resulted in decreased p53 stability due to increased ubiquitination. We treated FLT3-ITD-containing cells with specific ERK, AKT and STAT5 inhibitors. Interestingly, while inhibition of ERKs and AKT had no significant effect on the stability of p53, STAT5 inhibition resulted in a reduced level of p53 accompanied by increased K48 poly-ubiquitination. We further analyzed the interaction of p53 with MDM2 in AML cells by using immunoprecipitation. The results showed that the p53-MDM2 interaction was significantly enhanced after treatment with TKIs and STAT5 inhibitors, which was diminished in the presence of RG7388. Subcellular fractionation revealed the presence of p53 and STAT5 in both nucleus and cytoplasm. Treatment of cells with TKIs resulted in a decreased level of p53 and STAT5 in the nucleus, and immunoprecipitation of nuclear proteins with a p53 antibody revealed a reduced association of p53 with STAT5. Taken together, the data suggest that FLT3 inhibitors inhibited nuclear translocation of STAT5 and reduced its interaction of p53 thereby facilitating p53/MDM2 interaction and subsequent ubiquitination and degradation of p53. This study reveals a novel mechanism by which drug resistance to TKIs may occur and further support the use of MDM2/p53 interaction inhibitors in combination with TKIs for treatment of AML. 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: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1820-1820
    Abstract: Abstract 1820 Background and Objective: Although there have been introductions of novel and more potent therapeutic regimens, multiple myeloma (MM) remains an incurable hematological malignant disorder. Current goals in MM treatment are focused on finding new therapies that target the deregulated signaling cascades which promote MM cell survival and proliferation, such as STAT3 and nuclear factor-κB (NF-κB) (Bharti AC et al, Blood 2004). Cardamonin (2′,4′-dihydroxy-6′-methoxychalcone) is isolated from Alpinia katsumadai (Zingiberaceae), a plant widely used in traditional Chinese medicine. Previous reports have demonstrated that cardamonin possesses diverse pharmacologic actions, such as anti-inflammation (Hatziieremia S et al, Br J Pharmacol 2006), anti-melanogenesis (Cho M et al, Biochem Biophys Res Commun 2009) and anti-platelet properties (Jantan I et al, Phytomedicine 2008). However, very little is known about anti-myeloma activity of cardamonin. Thus, in the present report we investigated the therapeutic potential of cardamonin against MM, specifically studying the capacity of cardamonin to inhibit STAT3 and NF-κB pathways. Methods and Results: Cardamonin directly inhibited the growth of MM cell lines in a dose-dependent (from 0 μM to 100 μM) and time-dependent (from 0 h to 72 h) manner by using a cell counting kit-8 assay kit. Growth inhibition of MM cell lines, including RPMI 8226, U266 and ARH-77 cells, was demonstrated with an IC50 of less than 50 μM cardamonin at 24 h. Cardamonin also induced chemosensitivity to vincristine, doxorubicin, and dexamethasone in MM cells. Flow cytometric analysis showed that cardamonin caused accumulation of MM cells in G2 phase (from 14.6% to 81.9% for RPMI 8226, 24 h at 10 μM). A more precise evaluation of cell cycle by Hochest 33342 and 5-ethynyl-2′-deoxyuridine (EdU) fluorescence staining indicated that the cells in S phase were reduced in cardamonin-treated cells. Indeed, cardamonin strongly induced cell apoptosis as shown by annexin V-fluoroisothyocyanate analysis (from 5.3% to 49.4% for RPMI 8226, 48 h at 25 μM), which was also proved by a combination of acridine orange and ethidium bromide staining assay. Furthermore, cardamonin significantly enhanced the apoptotic effects of bortezomib from 23.1% to 74.3% and of thalidomide from 20.1% to 65.8%. Because of the pivotal role of STAT3 and NF-κB in MM cell survival and proliferation, we explored whether the above effects of cardamonin were mediated by interfering with STAT3 and NF-κB pathways by western blot analysis and immunofluorescence. We found that cardamonin blocked IL-6-inducible STAT3 phosphorylation and sequent STAT3 nuclear translocation. The constitutive phosphorylation of STAT3 found in certain cells was also abrogated by treatment with cardamonin in a dose- and time-dependent manner. In addition, we discovered that NF-κB, constitutively active in all human MM cell lines examined, was downregulated by cardamonin through suppression of phosphorylation of NF-κB p65 as evaluated by western blot. This correlated with reduction of the nuclear retention of p65. Moreover, cardamonin suppressed phosphorylation of IκBα, an inhibitor of NF-κB, and phosphorylation of Akt, which has been shown to phosphorylate p65. Additionally, immunoblotting analysis indicated that the expression of STAT3 and NF-κB-regulated gene products associated with proliferation (cyclin D1, TF and COX2), antiapoptosis (Bcl-2, Bcl-xL, Survivin, XIAP and Bfl-1/A1), invasion (ICAM-1), and angiogenesis (vascular endothelial growth factor) were down-regulated by cardamonin. ELISA assay showed that IL-6 release was also suppressed by cardamonin in certain MM cells. Conclusions: Taken together, our results suggest that cardamonin is a potent in vitro inhibitor of STAT3 and NF-κB pathways, which provides the molecular basis for its anti-myeloma activities, including suppression of proliferation, arrest of cell cycle, and induction of apoptosis. 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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1276-1276
    Abstract: Aberrant transcriptional programs play a critical role in the development of acute myeloid leukemias (AMLs). Although persistent over-expression of MEIS1 and HOXA9 has been shown to be essential for the initiation and maintenance of MLL-associated leukemia, it is still poorly understood what additional transcriptional regulators, independent of the MLL fusion-driven MEIS/HOX pathway, dictate the development of MLL leukemia. Considering that AMLs with MLL translocation are typically associated with the monocytic lineage (FAB M4 and M5), we explored the potential role of the monocytic lineage-specific transcriptional program in MLL leukemia. Using 97 genome-wide expression profiles of human MLL leukemias, we constructed an MLL distinctive transcriptional regulatory network. In addition to well-known transcriptional factors in leukemia development such as MEIS1 and HOXA family genes, we identified a highly active monocyte-specific gene signature that includes transcription factor PU.1. In our effort to determine the functional role of PU.1 in MLL leukemia, we found that lower PU.1 expression significantly delayed the onset of MLL- AF9 induced leukemia in primary bone marrow transplantation assay. MLL leukemia failed to maintain in vivo upon induced deletion of the PU.1 gene. To examine the clinical relevance of the PU.1 in AML patients, we further performed multivariate Cox proportional-hazards regression analysis in four published datasets of patients with AML, for whom gene expression and time-to-event data were available. We found that a PU.1-regulated 40-gene signature showed profound concordance with prognosis in segregating high-risk and low-risk AML patients. When specific subgroups of AMLs were examined, the PU.1 expression signature could predict patient outcome for MLL patients, but not in other major AMLs, such as t(8;21), t(15;17) and inv(16). We further explored the molecular mechanisms underlying the critical role of the PU.1 program in MLL leukemia. Functional annotation of this PU.1 expression signature identified the MEIS/HOX pathway (MEIS1, FLT3, KIT), as well as key genes in the inflammatory response (AIF1, NF-KB1 and CD180). We showed that PU.1 is required to maintain high expression of Meis1 and Pbx3 and also important downstream genes in the MEIS/HOX pathway that includes known MEIS/HOX targets c-Kit and Flt3. Using ChIP-sequencing, we demonstrated that PU.1 interacts with the MEIS/HOX regulatory program through co-binding with MEIS1 at the target genomic regions in a MLL-ENL cell line. In our effort to determine the role of PU.1-controlled inflammatory response genes, we found that the growth inhibition in PU.1 knockdown MLL leukemic cells was partially rescued by addition of the monocytic inflammatory cytokine AIF1. AIF1 provides an anti-apoptotic effect through activation of the NF-ƒÛB pathway and additional known apoptosis regulators. Interestingly, AML patients with higher expression of both AIF1 and MEIS1 had a significantly shorter overall survival time than those with lower expression of both genes. Patients with high expression of either MEIS1 or AIF1 had medium survival possibilities. Notably, the prognostic value of AIF1 and MEIS1 remained in those with monocytic AMLs (P=0.00079), but not in the non- monocytic group of patients (p=0.105). Collectively, these results strongly suggest that the monocyte-specific inflammatory cytokine AIF1 is an MEIS/HOX independent essential regulator in monocytic AMLs such as MLL leukemia. Loss of function PU.1 is leukemogenic in mouse models. Suppression of PU.1 activity is also required for the development of human myelocytic M2/M3 leukemia. Here we reveal a converse role for PU.1 as an essential positive regulator in the development of MLL myeloid leukemia, mostly M4/M5 monocytic AMLs. Our study demonstrats that the monocyte-specific PU.1-driven transcriptional program independently contributes to the development of myeloid MLL leukemia, in parallel with the MLL fusion pathway. PU.1 and downstream macrophage specific inflammatory cytokine AIF1 have important prognostic value and may serve as novel therapeutic targets for MLL leukemias. 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: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2014
    In:  Blood Vol. 124, No. 21 ( 2014-12-06), p. 5509-5509
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 5509-5509
    Abstract: Object: Chronic myeloid leukemia (CML) is a paradigm for neoplasmas that are defined by a unique genetic aberration, the BCR-ABL1 fusion gene. Microvesicles (MVs) are secretory particles released by various cell types including tumor cells. MVs released by CML cells constitute an important part of the microenvironment of leukemia and can modulate the interaction of tumor cells. MicroRNAs (miRNAs) loaded within MVs may also provide an insight in the roles of miRNAs playing in pathological mechanisms of CML. Methods:Blood samples were gathered from patients diagnosed as CML and normal heathy adults. All patients were admitted to Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology during the period from May to September in 2011. Our study was approved by the ethic committee of Wuhan Union Hospital and all subjects signed informed consent. We determined the miRNA expression profiles of CML-derived MVs using Agilent miRNA microarray analysis. Selected miRNAs obtained by microarray profiling were validated using real-time PCR. The putative target genes were predicted by bioinformatic software (TargetScan、miRanda、 PicTar、 MirTart2、PITA). Results:We identified numerous dysregulated miRNAs in MVs derived from CML patients compared with that from the controls by microarray analysis. Of the miRNAs detected, 226 dysregulated miRNAs were present in CML-MVs. With bioinformatic methods, we observed that there were 919 chromosome open reading frame (Corf) genes which were regulated by 169 aberrant MVs miRNAs from CML. Our results indicated that MVs derived from CML were enriched with different groups of altered miRNAs regulating Corf genes. It was interesting that some Corf genes were targeted by one aberrantly expressed MVs miRNAs and that several dysregulated miRNAs targeted one Corf gene. For example, 388 Corf genes were regulated by miR-513a-3p and 186 altered miRNAs targeted C15orf17. These findings suggested that Corf genes were active and complex in non-solid tumors. It was suggested that some members of the Corf genes were closely associated with cancers. For instance, C1orf43, also known as NICE-3 (a novel member of epidermal differentiation complex gene), had been reported to increase tumor cell proliferation and colony formation. In addition, deletion of C8orf4 was correlated with the risk of hematological neoplasmas. Furthermore, C16orf74, negatively associated with development of malignancies, was targeted by over-expressed miR-1299. Given that miRNAs could inhibit the expression of target genes, antineoplastic functions of C16orf74 might be suppressed. Similarly, C11orf30 that was a key oncogene was targeted by down-expressed miR-93, which facilitated C11orf30 to produce tumor-promoting roles. Interestingly, we observed that several Corf genes acting as oncogenes were regulated by over-expressed miRNAs. For instance, C6orf211 and C19orf10 were positively correlated with tumor progression and both of them were regulated by up-regulated miRNAs including miR-1246 and miR-1305. This indicated that Corf genes in diverse tumor microenvironments were likely to exert different influence on carcinogenesis. Conclusion: Briefly, we demonstrated for the first time that CML-derived MVs were enriched with dysregulated miRNAs targeting Corf genes, indicating that miRNAs regulating Corf genes were active in CML-MVs. Furthermore, Corf genes regulated by distinct sets of altered miRNAs might produce similar or alien effects on tumor progression. 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: 2014
    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. 118, No. 21 ( 2011-11-18), p. 1388-1388
    Abstract: Abstract 1388 Microvesicles(MVs) are small exosomes of endocytic origin released by normal healthy or damaged cell types, including leukemic cells. MVs have been considered as cell dust, however, recent data bring evidences that MVs generated during cell activation or apoptosis can transfer biologic messages between different cell types. MicroRNAs (miRNAs) have been demonstrated to be aberrantly expressed in leukemia and the overall miRNA expression could differentiate normal versus leukemia. The MVs expressing miRNAs were found in the primary tumors. However it is currently unknown whether miRNA content changes in MVs derived from leukemic cells. Here we compared the miRNA expression in leukemia-derived MVs to corresponding leukemia cells and analysed their roles in leukemia. K562 cells were cultured and collected. MVs derived from K562 cells were also isolated. The presence and levels of specific miRNAs from both MVs derived from K562 cells and K562 cells were determined by Agilent miRNA microarray analysis probing for 888 miRNAs. Some selected miRNAs were verified by real time qRT-PCR. Bioinformatic software tools were used to predict the target genes of identified miRNAs and define their function. Our results showed that 77 and 122 miRNAs were only expressed in MVs and K562 cells, respectively. There were significant differences in miRNA expression profiles between MVs and K562 cells. We also found that 112 miRNAs were co-expressed in MVs and K562 cells. This observaton may suggest that compartmentalization of miRNAs from cells into to MVs, for at least some miRNAs, is an active (selective) process. Among those abnormally expressed miRNAs, some have been proposed oncomiRNAs or tumor suppressors. For example, miR-155, has been proposed as oncomiRNA, was abnormally expressed only in MVs in our study, suggesting that oncomiRNA was present in MVs. Further analysis revealed that 39 potential target genes regulated by miR-155. Among them, 4 genes involed in oncogenes and the signal genes. OncomiRNAs such as miR-27a and miR-21 expressed in both MVs and corresponding cells, indicating that MVs bear miRNA characteristic of the cell origin. MVs, released into the leukemia microenvironment, play an important role in leukemia. In contrast to oncomiRNAs, if miRNA is associated with tumor suppressive activity, it is regarded as a tumor suppressor (oncosuppressor). The aberrantly expressed miR-125a-3p, miR-125-5p,miR-27b, which have implicated as tumor suppressors, appear in both cellular and MVs of leukemia in our study. MiR-125a-3p, miR-125-5p and miR-27b regulated 308 potential target genes. To our knowledge, 10 of them are tumor suppression genes. It is possible that these aberrantly expressed tumor suppressor miRNAs decreased or lost their roles of tumor suppression, which led to decrease or loss their roles of regulating their target genes including oncogenes, consequently resulted in leukemia. Since K562 cells presented t(9;22), we further examined the predicted function of the 6 expressed miRNAs located in chrosome 9 (hsa-miR-188-5p,hsa-miR-602)and 22(hsa-let-7b,hsa-miR-1249,hsa-miR-130b,hsa-miR-185), which expressed both in the MVs and K562 cells. Using the TargetScan, we found 442 predicted targets regulated by 6 miRNAs. Those miRNAs may play roles in leukemia via these 422 genes. This study is the first to identify and define miRNA expression between K562 cells presented t(9;22), derived from K562 cells and their corresponding cells. We found significant differences in miRNA expression between MVs and corresponding leukemia. K562 cells released MVs riched in miRNAs including oncomiRNAs or tumor suppressor miRNAs into leukemia microenvironment, which play a role in leukemia via regulating their targer genes including oncogenes, consequently resulted in leukemia. 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: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 781-781
    Abstract: Abstract 781 While serving as an initiating event in the disease etiology, chromosomal translocation alone may not be sufficient to drive a particular phenotype of leukemia. Acute leukemia characterized by rearrangements of the histone methyltransferase gene MLL (mixed lineage leukemia) requires additional mutations to develop the full blown malignancy, yet the molecular basis of cooperating events remains under-studied. We reasoned that monozygotic (MZ) twin pairs discordant for human leukemia are well-matched for both inherited genetic background and tissue-specific events, and thereby somatic mutations that arise in the disease twin may play a more prominent role in leukemogenesis. Using whole genome sequencing in a pair of 3-year old monozygotic twins discordant for MLL leukemia, we identified a MLL-NRIP3 fusion gene and mutations in histone H3 lysine 36 methyltransferase SETD2 in the leukemia twin. Retrovirus-mediated ectopic expression of MLL-NRIP3 in mouse hematopoietic cells was able to induce the same type of myeloid leukemia as the patient's in a transplant mouse model. A relative delay in disease onset suggested the occurrence of cooperating events in addition to the initial hit of MLL-NRIP3, in the development of induced leukemia. SETD2 mutations were recurrent (5.4%) in 241 acute leukemia patients, particularly in those with MLL-rearranged myeloid leukemia (22.2%). The identified SETD2 mutations are loss-of-function in nature, characterized by biallelic and truncating mutations, and accompanied by a global loss of trimethylation of H3K36 (H3K36me3) in the patient leukemic blasts. These data suggest that SETD2 acts as tumor suppressor gene in leukemia development. Functionally, transfection of SETD2 shRNA in MLL-AF9 knockin bone marrow cells resulted in decreased levels of SETD2 expression and H3K36 trimethylation. Notably, SETD2 knockdown significantly accelerated the development of MLL-AF9 leukemia in the mouse bone marrow transplantation experiment. Moreover, SETD2 knockdown yielded a significantly higher number of total colonies through the second and third rounds of serial replating of colony-forming cell (CFC) assay, suggesting that loss of SETD2 increased the self-renewal and proliferation potential of MLL leukemia-initiating cells. Finally, we showed that SETD2 deficiency was able to activate gene expression in MAPK, Jak-STAT and mTOR signaling, and dysregulates multiple metabolic and DNA repair pathways that are known to directly contribute to leukemogenesis. This comprehensive study provides compelling evidence for SETD2 as a novel tumor suppressor for leukemia, and suggests that the disruption of distinct histone modifying enzymes, MLL and SETD2, synergistically promotes the development of human leukemia. In addition, our study illustrates that whole-genome sequencing of phenotypically discordant monozygotic twins provides an effective approach, in combination with mutational analysis in patients and functional assays using experimental models, to uncover disease-causal genes. 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: 2012
    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. 126, No. 23 ( 2015-12-03), p. 4897-4897
    Abstract: Xiaomei Chen and Jianyu Weng contributed equally to this study. The outcomes ofrelapsed or refractory acute myeloid leukemia (RR-AML) are poor and effective salvage regimens are urgently needed. We present a study of 14 patients with RR-AML (median age 42years, range 18-65years; male n=12, female n= 2) treated with CLAT regimen, which consisted of cladribine 5mg/m² per day i.v. 2-3 hour on days 1-5, cytarabine 1.0g/m² per day i.v. 4 hours after cladribine on days 1-5, topotecan 1.25mg/m² per day i.v. 4 hours after cytarabine on days1-5 and G-CSF 300ug per day subcutaneous injection on days until neutrophile granulocyte recovery. Total of fourteen patients were included into the study from June 2013 to June 2015, Two (14.3%) patients were relapsed and twelve (85.7%) patients were refractory, 4 of 14 patients were relapsed or refractory after allogeneic-HSCT. Two patients died of invasive fungal infection before the assessment. Seven patients (58.3%) achieved complete remission (CR), and one patient (8.3%) achieved partial remission (PR), the rest patients (33.3%) did not respond (NR). The overall response rate was 66.7%. Following CLAT treatment, four patients with CR underwent allogeneic hematopoietic stem cell transplantation (HSCT) or microtransplantation. The median relapse-free survival (RFS) for RR-AML patients receiving CLAT regimen was 8.6 (range 2-16) months. Thirteen patients developed grade 4 granulocytopenia and thrombocytopenia, the median duration was 13(range 2 to 21) days and12 (range 2 to 21) days, respectively. The most common non-hematological side effects included nausea, vomiting, diarrhoea, and were grade 1/2. The CLAT regimen seems promising for the treatment of patients, and it was well tolerated. This regimen offers an alternative treatment for those patients with RR-AML who have received severe intensive treatment, especially with anthracycline-containing chemotherapy. The project was sponsored by grants from National Natural Science Foundation of China (No. 30972790; No.81270648; No.81370665; No.81300446) Provincial Natural Science Foundation of Guangdong (No. S2012010009560) Provincial Science and Technology Planning Project of Guangdong (No.2013B021800186; No.2013B021800201), and Science and Technology Planning Project of Guangzhou (No. 201400000003-4, 201400000003-1). 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: 2015
    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. 126, No. 23 ( 2015-12-03), p. 3319-3319
    Abstract: Introduction: Clinical trial data for MDS pts may not be representative of the general MDS pt population because some pts may be ineligible for clinical trials or may decline to participate. The GE Centricity™ EMR database (GE Healthcare IT, Princeton, NJ, USA), which is anonymized and compliant with the Health Insurance Portability and Accountability Act of 1996, contains clinical practice data from 〉 38 million US pts from 1994 onward (Asche CV, et al. ISRN Cardiol. 2011;2011:924343). Pt diagnoses in the database can be determined using International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) codes; however, cohorts selected this way may include pts without MDS due to coding errors. We accessed the GE EMR database to identify a large, real-world cohort of MDS pts and compared them with MDS pts reported to the well-established, population-based Surveillance, Epidemiology, and End Results (SEER) Program. Methods: The GE EMR database is generally representative of the US population and contains data from single-physician offices to large practices and networks, covering 49 US states. Of 〉 30,000 participating clinicians, two-thirds are primary care physicians; the remainder are specialists. Using this database, we assembled a retrospective cohort of MDS pts with data entered during January 2006-February 2014. MDS pts were initially identified by the presence of ≥ 1 MDS-specific ICD-9-CM diagnosis code (codes: 238.72-238.75). Several different approaches were developed, evaluated, and applied to refine this cohort by excluding pts with a lower likelihood of MDS (Figure). Results: The Initial Cohort comprised 9,645 pts with ≥ 1 MDS-specific ICD-9-CM code; 31% of pts received supportive treatment (hematopoietic growth factors, iron-chelating therapy, or transfusions) and 8% received active treatment (azacitidine, decitabine, or lenalidomide). However, only 9% of pts in this cohort received transfusions; fewer than expected. This may be due to unrecorded transfusions occurring outside the EMR system, a limitation that may apply to other EMR databases. The Initial Cohort included 563 pts with ≥ 2 MDS-specific ICD-9-CM codes (Figure; 1st Modified Cohort); 15% of these pts had received transfusions; considerably higher than in the Initial Cohort. To exclude pts unlikely to have MDS in the Initial Cohort, the inclusion criteria were modified to include only pts with ≥ 1 MDS entry plus an entry for MDS-specific active treatment (N = 1,208) (Figure; 2nd Modified Cohort); supportive treatment was insufficient for inclusion as it is not MDS-specific. As diagnosis of MDS requires hemoglobin (Hb) measurement and bone marrow (BM) aspirate or biopsy prior to diagnosis, inclusion criteria were refined to include only those pts with either ≥ 2 MDS entries or ≥ 1 entry for MDS plus ≥ 1 of the following: prior MDS-specific active treatment; ≥ 2 Hb tests ≤ 1 year prior to code entry; or ≥ 1 BM procedure ≤ 1 year prior to code entry (N = 5,623) (Figure; 3rd Modified Cohort). This cohort was then further refined by excluding pts whose EMR included terms indicative of a non-diagnosis of MDS, such as "rule out," resulting in a Final Cohort of 5,162 pts most likely to have MDS (Figure; Final Cohort) whose baseline characteristics were generally comparable to MDS pts reported to the SEER Program during 2001-2011 (Table). In this Final Cohort, 35% received supportive care alone while 13% received active treatment; unrecorded transfusions may have contributed to underreporting of supportive care. Treatment patterns and the impact of variables such as age, insurance type, geographical location, and comorbidities will be presented. Conclusions: By leveraging the GE Centricity EMR database and evaluating different approaches for the ascertainment of MDS pts, we identified a large, real-world cohort of MDS pts whose baseline characteristics are comparable to MDS pts from the SEER Program. Although characteristics between the modified cohorts were similar, the Final Cohort was selected based on clinical and diagnostic features associated with the diagnosis of MDS in real-world clinical practice. Our methodology can inform other investigators interested in utilizing EMR databases for cancer outcomes research, and the cohort we identified will be useful in the further characterization of treatment patterns and outcomes of MDS pts who are more representative than participants of clinical trials. Disclosures Ma: Incyte Corporation: Consultancy; Celgene Corporation: Consultancy. Swern:Celgene Corporation: Employment, Equity Ownership. Kiselev:Celgene Corporation: Consultancy. Fliss:Celgene Corporation: Employment, Equity Ownership. Lu:Celgene Corporation: Employment. Scott:Celgene Corporation: Consultancy, Speakers Bureau. Steensma:Incyte: Consultancy; Amgen: Consultancy; Celgene: Consultancy; Onconova: Consultancy. Sugrue:Celgene Corporation: Employment, Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2011
    In:  Blood Vol. 118, No. 21 ( 2011-11-18), p. 4411-4411
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 4411-4411
    Abstract: Abstract 4411 MicroRNAs (miRNAs) are small non-coding RNA sequences of about 22nt and play an important role in disease progression including carcinogenesis. Recent evidences reveal that genetic exchange of miRNAs between cells can be accomplished through microvesicles(MVs). MVs are small exosomes of endocytic origin released not only by activated platelets but also by hematologic malignancies such as leukemia. Sheded from the plasma membrane MVs move into the extracellular environment to facilitate communication between cells. MVs containing miRNAs would enable intercellular cross-talk in vivo. This prompted us to investigate specific variations of miRNA expression patterns in MVs derived from leukemia. We examined the miRNA expression profile of MVs both from chronic myeloid leukemia cell line K562 and normal human volunteers’ peripheral blood. Agilent miRNA microarray was employed for detection and then real-time PCR for verification. Bioinformatic software tools were used to predict the target genes of identified microRNAs and define their function. Our study figures out miRNAs of MVs from leukemia and normal cells and characterizes specific miRNAs expression. We found that MVs from K562 cells express 348 miRNAs of 888 miRNAs. While 77 miRNAs displayed down regulation, 134 were upregulated. Interestingly, most of the miRNAs dysregulated in MVs display up regulated expression, suggesting their prevalent roles as tumor promotors. Among the aberrantly expressed miRNAs, miR-1290 was identified whose expression levels was more than 900 times than that of normal cell derived MVs. While the expression of miR-125a-3p was up-regulated by more than 300 times. And miR-654-5p, miR-654-5p, miR-1268 and miR-1246 were up-regulated more than 200 times. Five of the disregulated miRNAs (miR-1290, miR-125a-3p, let-7a, let-7f, miR-26a) were further assayed and validated by Q-RT-PCR results which correlated well with the microarray data. Of note, upexpression of miR-663, miR-1237, miR-149, miR-634, miR-1181, miR-92b, miR-130b as well as downregulation of let-7a, let-7f, miR-26a, miR-26a, miR-26b, miR-266, miR-126, miR-93, miR-451, miR-103, miR-107, miR-27a were similar to what was previously reported about leukemia, thus supporting the general roles of these miRNAs as tumor suppressors or oncomiRNAs in leukemia. Meanwhile we noticed a reduced expression of miR-1237, miR-365, miR-223b, miR-27b, miR-151-5p, miR-23a, miR-21, miR-30e, miR-361-5p, miR-484, miR-185, miR-374a, miR-197 in our study, as recently stated in solid tumor, thus suggesting that significantly lower abundance of these miRNAs is shared in leukemia. In addition to identify the already known leukemia-associated miRNAs, we had checked out dozens of novel miRNAs without any articles published until now, namely miR-502-3p, miR-718, miR-877, miR-1470, miR-720, miR-1267, miR-127, miR-767-3p, miR-1974-v14.0, miR-361-5p, miR-374b and so on. Using bioinformatic tools (TargetScan), we predicted potential targets for those miRNAs that exhibited altered expression in MVs from leukemia cells. Of particular interest, we found that hsa-miR-125a-3p which was refered above may regulate 34 potential genes of which five are located around the chromosome open reading frame. We hypothesised that miR-125a-3p may participate in the modulation of leukemia through these genes by affecting chromosome. Taken together, our study identifies miRNAs of MVs from leukemia and normal cells and characterizes specific miRNAs expression. These findings highlight a number of miRNAs from leukemia-derived MVs that may contribute to the development of hematopoietic malignancies. Further investigation will reveal the function of these differentially expressed miRNAs and may provide potential targets for novel therapeutic strategies. 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: 2011
    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...