Mutation analysis of the DNA-damage checkpoint gene CHK2 in myelodysplastic syndromes and acute myeloid leukemias

2001 ◽  
Vol 25 (4) ◽  
pp. 333-338 ◽  
Author(s):  
Wolf-K. Hofmann ◽  
Carl W. Miller ◽  
Kunihiro Tsukasaki ◽  
Sigal Tavor ◽  
Takayuki Ikezoe ◽  
...  
Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5226-5226
Author(s):  
Henning D Popp ◽  
Susanne Brendel ◽  
Nicole Naumann ◽  
Thomas Henzler ◽  
Wolf-Karsten Hofmann ◽  
...  

Abstract Purpose: Increased DNA damage and alteration of the DNA damage response (DDR) are critical features of genetic instability presumably implicated in pathogenesis of myelodysplastic syndromes (MDS) and acute myeloid leukemias (AML). Materials and methods: We performed combined γH2AX/53BP1 immunofluorescent focus staining of DNA double-strand breaks (DSB) in MDS and AML cell lines, in CD34+ selected cells of normal and MDS bone marrow (including three cases of chronic myelomonocytic leukemias) and in blasts of AML bone marrow. In addition, we screened for activation of DDR by immunoblotting of p-ATM, p-ATR, p-CHK1, p-CHK2 and p-TP53. Results: Compared to γH2AX foci levels in normal bone marrow samples (0.18 focus per CD34+ ± 0.6), increased levels of γH2AX foci were detected in 1/1 MDS cell line (6.4 foci per cell ± 0.0), 6/6 AML cell lines (12.0 foci per cell ± 0.6), 12/12 MDS bone marrow samples (2.8 foci per CD34+ ± 0.7) as well as 12/12 AML bone marrow samples (6.0 foci per blast ± 0.6). γH2AX and 53BP1 co-localized in all tested samples forming diffuse, clustered and marginal patterns. DDR proteins were expressed heterogeneously suggesting impairment of DDR. Conclusions: Our results provide evidence for a continuous increase of constitutive DSB across the spectrum from MDS to AML in the context of an impaired DDR. γH2AX and 53BP1 co-localize in unique patterns in nuclei of MDS and AML presumably owing to a non-random spatial organization of the genome and foci formation implies promotion of ineffective nonhomologous end-joining repair mechanisms at sites of constitutive DSB. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 57 ◽  
pp. 112-118 ◽  
Author(s):  
Henning D. Popp ◽  
Nicole Naumann ◽  
Susanne Brendel ◽  
Thomas Henzler ◽  
Christel Weiss ◽  
...  

2002 ◽  
Vol 132 (2) ◽  
pp. 156-158 ◽  
Author(s):  
Jin-Yeong Han ◽  
Kyeong-Hee Kim ◽  
Hyuk-Chan Kwon ◽  
Jae-Seok Kim ◽  
Hyo-Jin Kim ◽  
...  

2013 ◽  
Vol 92 (11) ◽  
pp. 1543-1552 ◽  
Author(s):  
María Abáigar ◽  
Fernando Ramos ◽  
Rocío Benito ◽  
María Díez-Campelo ◽  
Javier Sánchez-del-Real ◽  
...  

2005 ◽  
Vol 157 (2) ◽  
pp. 118-126 ◽  
Author(s):  
Emmanuelle Barouk-Simonet ◽  
Valérie Soenen-Cornu ◽  
Christophe Roumier ◽  
Alain Cosson ◽  
Jean-Luc Laï ◽  
...  

2019 ◽  
Vol 11 (1) ◽  
Author(s):  
Jing Ye ◽  
Jie Zha ◽  
Yuanfei Shi ◽  
Yin Li ◽  
Delin Yuan ◽  
...  

Abstract While the aberrant translocation of the mixed-lineage leukemia (MLL) gene drives pathogenesis of acute myeloid leukemia (AML), it represents an independent predictor for poor prognosis of adult AML patients. Thus, small molecule inhibitors targeting menin-MLL fusion protein interaction have been emerging for the treatment of MLL-rearranged AML. As both inhibitors of histone deacetylase (HDAC) and menin-MLL interaction target the transcription-regulatory machinery involving epigenetic regulation of chromatin remodeling that governs the expression of genes involved in tumorigenesis, we hypothesized that these two classes of agents might interact to kill MLL-rearranged (MLL-r) AML cells. Here, we report that the combination treatment with subtoxic doses of the HDAC inhibitor chidamide and the menin-MLL interaction inhibitor MI-3 displayed a highly synergistic anti-tumor activity against human MLL-r AML cells in vitro and in vivo, but not those without this genetic aberration. Mechanistically, co-exposure to chidamide and MI-3 led to robust apoptosis in MLL-r AML cells, in association with loss of mitochondrial membrane potential and a sharp increase in ROS generation. Combined treatment also disrupted DNA damage checkpoint at the level of CHK1 and CHK2 kinases, rather than their upstream kinases (ATR and ATM), as well as DNA repair likely via homologous recombination (HR), but not non-homologous end joining (NHEJ). Genome-wide RNAseq revealed gene expression alterations involving several potential signaling pathways (e.g., cell cycle, DNA repair, MAPK, NF-κB) that might account for or contribute to the mechanisms of action underlying anti-leukemia activity of chidamide and MI-3 as a single agent and particularly in combination in MLL-r AML. Collectively, these findings provide a preclinical basis for further clinical investigation of this novel targeted strategy combining HDAC and Menin-MLL interaction inhibitors to improve therapeutic outcomes in a subset of patients with poor-prognostic MLL-r leukemia.


Sign in / Sign up

Export Citation Format

Share Document