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  (7)
  • 1
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 712-712
    Abstract: The oncogenic BCR/ABL tyrosine kinase induces constitutive DNA damage in Philadelphia chromosome (Ph1)-positive leukemia cells. We find that BCR/ABL kinase - induced reactive oxygen species (ROS) cause chronic oxidative DNA damage as indicated by an enzymatic assay detecting oxidized bases. These DNA lesions result in DNA double-strand breaks (DSBs) detected by comet assay, immunofluorescent gamma-H2AX nuclear foci and linker-ligation PCR (LL-PCR). Combined analysis of the length of LL-PCR products and the sequences of two reference genes DR-GFP and Na+/K+ ATPase revealed that ROS dependent DSBs occurred in the regions containing multiple, 5–9bp long stretches of G/C, in concordance with the notion that oxidative DNA damage is predominantly detected in G/C-rich sequences. Elevated numbers of DSBs were detected in BCR/ABL cell lines, murine bone marrow cells transformed with BCR/ABL and in CML patient samples, in comparison to normal counterparts. Inhibition of the BCR/ABL kinase by STI571 and diminishion of ROS activity by the ROS scavenger PDTC reduced DSBs formation. Cell cycle analysis revealed that most of these DSBs occur during S and G2/M phases, and are probably associated with the stalled replication forks. Homologous recombination repair (HRR) and non-homologous end-joining (NHEJ) represent two major mechanisms of DSBs repair in S and G2/M cell cycle phase. Using the in vivo recombination assay consisting of the DSB-dependent reconstitution of the green fluorescent protein (GFP) gene we found that HRR is stimulated in BCR/ABL-positive cells. In addition, in vitro assay measuring the activity of NHEJ revealed that this repair process is also activated by the BCR/ABL kinase. RAD51 and Ku70 play a key role in HRR and NHEJ, respectively. The reaction sites of HRR and NHEJ in the nuclei could be visualized by double-immunofluorescence detecting co-localization of gamma-H2AX foci (DSBs sites) with RAD51 (HRR sites) or Ku70 (NHEJ sites). Equal co-localization frequency of gamma-H2AX foci with RAD51 and Ku70 was detected, suggesting that both HRR and NHEJ play an important role in reparation of ROS-dependent DSBs in BCR/ABL-transformed cells. Analysis of the DSBs repair products in the reporter DR-GFP gene in BCR/ABL cells identified ~40% of HRR and ~60% of NHEJ events. Sequencing revealed point-mutations in HRR products and large deletions in NHEJ products in BCR/ABL-positive cells, but not in non-transformed cells. We propose that the following series of events may contribute to genomic instability of Ph1-positive leukemias: BCR/ABL → ROS → oxidative DNA damage → DSBs in proliferating cells → unfaithful HRR and NHEJ repair. Since BCR/ABL share many similarities with other members of the fusion tyrosine kinases (FTKs) family, these events may contribute to genomic instability of hematological malignancies caused by FTKs.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2005
    In:  Blood Vol. 106, No. 11 ( 2005-11-16), p. 1982-1982
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 1982-1982
    Abstract: Clinical observations and experimental findings indicated that BCR/ABL stimulates genomic instability leading to mutations and chromosomal abnormalities. The accumulation of genetic errors is believed to be responsible for the transition from a relatively benign CML chronic phase (CML-CP) to the aggressive blast crisis phase (CML-BC) and the resistance to imatinib mesylate. BCR/ABL- positive leukemia cells accumulate an excess of potentially lethal DNA double-strand breaks (DSBs) caused by reactive oxygen species (ROS) or genotoxic treatment. However, BCR/ABL tyrosine kinase facilitates the repair of DSBs and promotes survival. Therefore, the infidelity of DSBs repair processes may contribute to genomic instability in leukemia cells exposed to elevated numbers of spontaneous and/or induced DSBs. To test this hypothesis DSBs repair efficiency and fidelity was examined and compared in parental and BCR/ABL-transformed cells. Nuclear foci detected by γ-H2AX (the form of H2AX histone that is quickly phosphorylated on Serine 139 by ATM, ATR and/or DNA-PKcs kinases on megabase-length fragments near DSB sites) immunofluorescence served as indicators of DSBs. We found that BCR/ABL-positive leukemia cells acquire more DSBs after γ-irradiation in comparison to normal cells. Homologous recombination (HR) and non-homologous end-joining (NHEJ) represent two major mechanisms of DSBs repair in mammalian cells. HR and NHEJ reaction sites in the nuclei can be visualized by double-immunofluorescence detecting co-localization of γ-H2AX foci with RAD51 or Ku70, respectively. NHEJ and HR appear to work in a time-dependent fashion (NHEJ followed by HR) and be more active in BCR/ABL-transformed cells in comparison to normal counterparts. Time-dependent engagements of NHEJ and HR mechanisms in repair of DSBs after γ-irradiation are accompanied by elevated accumulation of Ku70 and RAD51 proteins in cell lysates obtained from BCR/ABL cells. Specific DSBs repair assays confirmed that BCR/ABL leukemia cells in comparison to normal cells displayed enhanced capability of HR and NHEJ. However, analysis of DSBs repair products revealed that the repair mechanisms were less faithful in former cells generating large deletions and point mutations during NHEJ and HR, respectively. In summary, BCR/ABL leukemia cells display facilitated, but unfaithful HR and NHEJ, which may contribute to accumulation of genetic errors in surviving leukemia cells leading to malignant disease progression.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    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. 108, No. 1 ( 2006-07-01), p. 319-327
    Abstract: Mutations in the BCR/ABL kinase domain play a major role in resistance to imatinib mesylate (IM). We report here that BCR/ABL kinase stimulates reactive oxygen species (ROS), which causes oxidative DNA damage, resulting in mutations in the kinase domain. The majority of mutations involved A/T→G/C and G/C→A/T transitions, a phenotype detected previously in patients, which encoded clinically relevant amino acid substitutions, causing IM resistance. This effect was reduced in cells expressing BCR/ABL(Y177F) mutant, which does not elevate ROS. Inhibition of ROS in leukemia cells by the antioxidants pyrrolidine dithiocarbamate (PDTC), N-acetylcysteine (NAC), and vitamin E (VE) decreased the mutagenesis rate and frequency of IM resistance. Simultaneous administration of IM and an antioxidant exerted better antimutagenic effect than an antioxidant alone. Therefore, inhibition of ROS should diminish mutagenesis and enhance the effectiveness of IM. (Blood. 2006;108:319-327)
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    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. 104, No. 12 ( 2004-12-01), p. 3746-3753
    Abstract: The oncogenic BCR/ABL tyrosine kinase induces constitutive DNA damage in Philadelphia chromosome (Ph)-positive leukemia cells. We find that BCR/ABL-induced reactive oxygen species (ROSs) cause chronic oxidative DNA damage resulting in double-strand breaks (DSBs) in S and G2/M cell cycle phases. These lesions are repaired by BCR/ABL-stimulated homologous recombination repair (HRR) and nonhomologous end-joining (NHEJ) mechanisms. A high mutation rate is detected in HRR products in BCR/ABL-positive cells, but not in the normal counterparts. In addition, large deletions are found in NHEJ products exclusively in BCR/ABL cells. We propose that the following series of events may contribute to genomic instability of Ph-positive leukemias: BCR/ABL → ROSs → oxidative DNA damage → DSBs in proliferating cells → unfaithful HRR and NHEJ repair.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    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 ; 2007
    In:  Blood Vol. 110, No. 11 ( 2007-11-16), p. 1024-1024
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1024-1024
    Abstract: DNA damage and defects in DNA repair pathways may severely predispose to genomic instability which is one of the major factors associated with the formation and progression of chronic myelogenous leukemia (CML). However, leukemia cells transfected by BCR/ABL seem to be better equipped to survive DNA damage generated by reactive oxygen species (ROS) and external factors through activation of double-strand breaks (DSBs) repair by homologous recombination (HR) and non-homologous end-joining (NHEJ). A genome-wide screen was performed to identify genes regulated by BCR/ABL kinase and involved in DSBs repair. Werner syndrome protein (WRN), which exhibits helicase and exonuclease activity, was upregulated in CML cells. WRN is capable of unwinding various DNA structures associated with progressing replication forks as well as promoting Holliday junctions formed as intermediates in DNA recombination. Moreover, the helicase can directly interact with a variety of proteins involved in DSBs repair including Ku complex (NHEJ), and RAD51 (HR). Lack of WRN protein in Werner syndrome is characterized by accumulation of DSBs, genomic instability and a high incidence of cancer. Here we present evidence that BCR/ABL induced the expression of WRN mRNA and protein by activation of c-MYC transcription and inhibition of caspase-dependent cleavage, respectively. Immunoprecipitation and pull-down studies indicated that WRN is phosphorylated by BCR/ABL, and that BCR/ABL SH2 domain interacts directly with phospho-Y1346 of WRN. Drug sensitivity assays performed after downregulation of WRN expression by shWRN in BCR/ABL-positive cells have demonstrated an increased sensitivity to genotoxic stress induced by cisplatin and oxidative stress caused by H2O2. Experiments using TUR90010 lymphoblast cell line established from a Werner syndrome patient (3724C 〉 T) and transfected with BCR/ABL confirmed that WRN plays an important role in response to DNA damage in CML cells. Further studies revealed that BCR/ABL-positive leukemia cells exert an enhanced WRN-mediated helicase activity. Bone marrow cells derived from transgenic mice expressing the helicase-defective WRN mutant (K577M) and transfected with BCR/ABL display increased sensitivity to cisplatin compared to those obtained from the wild-type littermates. The role of WRN in BCR/ABL-induced DSBs repair pathways, HR and NHEJ, was examined. NHEJ activity was measured in nuclear cell lysates of BCR/ABL-positive leukemia cells using linearized double-stranded plasmid as a substrate. Removal of WRN by immunoprecipitation did not affect the efficacy of NHEJ reaction. HR was assessed using cells containing one copy of the modified gene for GFP containing a unique I-SceI restriction site with two stop codons as a recombination reporter and a truncated fragment of the GFP gene as a template for homologous repair. A HR event restores functional GFP expression. Downregulation of WRN protein by shRNA abrogated HR activity induced by BCR/ABL. Therefore BCR/ABL-dependent overexpression of WRN helicase seemed to be important for HR, but not NHEJ. Finally, an enhanced interaction between WRN and RAD51 upon DNA damage in BCR/ABL-positive cells supported that conclusion. In summary, BCR/ABL-mediated overexpression and enhanced activation of WRN helicase played an essential role in response of CML cells to elevated numbers of DBSs induced by oxidative and genotoxic stress.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 714-714
    Abstract: Imatinib mesylate (IM) [Gleevec, STI571], a selective inhibitor of ABL kinase activity, revolutionized the treatment of BCR/ABL-positive leukemias. Unfortunately, resistance to IM has become a rising clinical problem. Although the resistance could be achieved by various mechanisms, mutations in the BCR/ABL kinase domain seem to play a major role. Identification of the mechanisms causing these mutations may have a significant impact on the effectiveness of IM therapy. Mutations usually result from enhanced DNA damage and/or deregulated mechanisms responsible for DNA repair. Much endogenous DNA damage arises from intermediates of oxygen reduction generating potentially damaging molecules called reactive oxygen species (ROS). We report here that BCR/ABL-mediated transformation is associated with the kinase-dependent stimulation of ROS production and elevation of DNA oxidative damage. Immunofluorescence visualized more 8-oxoG nuclear lesions and enzymatic assays detected more oxidized bases in DNA in BCR/ABL-transformed cells (CML patient cells and leukemic cell lines) in comparison to normal cells. To examine the potential effect of ROS on mutations in BCR/ABL kinase freshly transformed 32D-BCR/ABL cells (no mutations in BCR/ABL) were cultured in the absence (control) or presence of IM (to inhibit BCR/ABL kinase and ROS production), PDTC (ROS scavenger) or IM+BSO (to inhibit BCR/ABL kinase and elevate ROS because BSO prevents its decay) for 8 weeks in medium supplemented with IL-3; then IM-resistance was examined. About 0.1% of control cells displayed IM-resistance, which was dramatically reduced in the presence of IM or PDTC, implicating BCR/ABL kinase-dependent ROS-mediated mechanism. IM+BSO treatment resulted in partial inhibition of IM-resistance, suggesting that another BCR/ABL-induced mechanism may collaborate with ROS in mutagenesis. IM-resistance was associated with mutations in the sequence encoding bcr/abl kinase. The majority of detected mutations were GC→AT transitions causing amino acid substitutions; most of them are already proven to be clinically relevant. Similar conditions were applied to study the mutagenesis in Na+K+ ATPase (mutations may cause the resistance to ouabain) in 32D parental and freshly transformed 32D-BCR/ABL cells. BCR/ABL increased the ouabain resistance rate ~12-times, which was associated with mutations in the alpha1 subunit of Na+K+ ATPase, essential for ouabain resistance. Again the majority of mutations were GC→AT transitions. To confirm that similar mechanisms of BCR/ABL-dependent mutagenesis operate in living organisms, IM-resistance was examined in SCID mice injected with freshly transformed 32D-BCR/ABL cells and fed with normal or vitamin E-rich (VE) diet (to decrease ROS) for 8 weeks. Leukemia cells harvested from bone marrow and spleen of the mice fed with VE diet displayed ~2 -fold reduction in the frequency of IM-resistance in comparison to the animals obtaining regular food. Mutations in the BCR/ABL kinase were detected in IM-resistant clones. Altogether, these studies suggest that BCR/ABL kinase facilitates ROS-dependent mutagenesis of in different genes, including bcr/abl gene, and that this process is essential for IM-resistance. Early treatment with IM can prevent mutagenesis, but if mutations occur it will select the outgrowth of mutant IM-resistant cells. In addition, reduction of ROS should greatly diminish the mutagenesis in BCR/ABL kinase associated with IM-resistance.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    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. 106, No. 11 ( 2005-11-16), p. 2873-2873
    Abstract: A genome-wide screen suggested that BCR/ABL kinase might stimulate WRN, a member of the RecQ-like DNA helicases family. The Werner syndrome protein (WRN) exerts DNA helicase and 3′-5′ exonuclease activities. Inactivating mutations in the WRN gene causes Werner syndrome, characterized by premature aging, genomic instability and cancer predisposition. The WRN helicase unwinds unusual DNA structures, which can occur physiologically, or can be accidentally generated during DNA repair (double-stranded DNA with mismatched tails, bimolecular G4 quartets and Holliday junctions). In addition, WRN physically interacts with components of two major systems for DNA double-strand breaks (DSBs) repair: non-homologous end-joining (NHEJ) and homologous recombination (HR). Here we demonstrated that BCR/ABL regulates the expression of WRN mRNA and protein in CML primary cells and BCR/ABL-transformed cell lines. BCR/ABL kinase-induced WRN expression is mediated by c-MYC, but not STAT5 - dependent transcription as well as by inhibition of caspases-dependent cleavage. In addition, immunoprecipitation and pull-down studies indicated that BCR/ABL interacts directly with WRN resulting in its tyrosine phosphorylation. Mutation analysis revealed that multiple domains/amino acid residues of BCR/ABL and WRN are involved in the interaction. BCR/ABL-positive leukemia cells exerted an enhanced WRN-dependent helicase activity. In addition, immunoprecipitation and double-immunofluorescence co-localization studies demonstrated an elevated interaction between WRN and RAD51 in BCR/ABL cells undergoing genotoxic stress in comparison to parental counterparts. Altogether, it is likely that WRN is involved in DSBs repair by HR in leukemia cells. More detailed studies are underway to pinpoint the role of WRN in DNA damage response in BCR/ABL-transformed cells.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    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...