EHA Library - The official digital education library of European Hematology Association (EHA)

LOW MYBL2 EXPRESSION OBSERVED IN MYELODYSPLASTIC SYNDROME PATIENTS WITH WORSE PROGNOSIS IS ASSOCIATED WITH ALTERED DNA REPAIR MECHANISMS IN HAEMATOPOETIC STEM CELLS
Author(s): ,
Rachel Bayley
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
,
Laila Cancian
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
,
Carl Ward
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
,
Giacomo Volpe
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
,
Stephanie Dumon
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
,
Jidnyasa Gujar
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
,
Grant Stewart
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
,
Eva Petermann
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
Paloma Garcia
Affiliations:
Institute of Cancer and Genomic Science,University of Birmingham,Birmingham,United Kingdom
(Abstract release date: 05/18/17) EHA Library. Garcia P. 06/23/17; 181406; S119
Paloma Garcia
Paloma Garcia
Contributions
Abstract

Abstract: S119

Type: Oral Presentation

Presentation during EHA22: On Friday, June 23, 2017 from 11:30 - 11:45

Location: Hall E

Background

MYBL2 is a transcription factor with roles in the cell cycle and genome integrity. MYBL2 is located on chromosome 20, within a region commonly deleted in human blood disorders (del20q). Our published data shows that reduced levels of MYBL2 predispose to development of myelodysplastic syndrome (MDS)-like disease in mouse models during ageing, indicating that MYBL2 could be acting as a tumour suppressor gene within del20q abnormality. Moreover, our previous work demonstrated that regardless of del20q deletion, MYBL2 expression is reduced in CD34+ bone marrow cells from MDS patients with worse prognosis. Because it has been shown that the cell of origin of MDS is the haematopoietic stem cell (HSC) and given the role of MYBL2 in DNA replication fork progression and maintenance of genome integrity, we hypothesised that low MYBL2 levels in HSC could contribute to elevated somatic mutations through changes in DNA repair pathways and drive disease development.

Aims

The aim of this study was to determine if low MYBL2 levels affect the double strand break (DSB) DNA repair damage response in HSC. 

Methods

In this study we used our mouse model in which animals express ~50% normal levels of MYBL2 (Mybl2+/∆). We characterised the ability of HSCs from young (7 weeks) and old (70 weeks) animals to respond to in vivo ionising radiation (2Gy) in terms of proliferation, apoptosis and colony forming ability. We measured the activation of the two main DNA repair pathways operating in the cells to deal with DSB: the error prone non-homologous-end-joining (NHEJ) and the error-free homologous recombination (HR) by assessing 53BP1 and Rad51 recruitment by immunofluorescence, respectively. Finally, we analysed the frequency of chromosome abnormalities present in the progeny of Mybl2+/∆ HSC that have previously been irradiated to determine the long term effects of changes in DNA repair. 

Results

We observed that Mybl2+/∆ HSCs had limited proliferative potential and displayed an increased sensitivity to ionizing radiation which increased during ageing. Mybl2+/∆ HSCs also displayed altered kinetics of 53BP1 and Rad51 recruitment and clearance, including retention of 53BP1 foci at later time points following irradiation and decreased levels of Rad51 foci when compared to Mybl2+/+ HSCs. Using plasmid functional assays, we showed that Mybl2+/∆ HSCs repair quite efficiently by NHEJ, but this efficiency is disrupted when cells are challenged with ionising radiation. Furthermore, Mybl2+/∆ HSCs have increased sensitivity to inhibition of DNA-PKC (required for NHEJ) but not ATM (required by HR). We also observed that after ionizing irradiation Mybl2+/∆ HSCs progeny displayed an increased percentage of chromatids with fragile telomeres. Moreover, by making use of publically available RNA-seq datasets from MDS patients, we have identified a clear association between low MYBL2 levels and low expression of DNA-repair genes in patients with worse prognosis. 

Conclusion
In summary, we have shown that decreased expression of MYBL2 leads to an imbalance in the DSB DNA-repair pathway choice, ultimately resulting in increased genomic instability of the blood cell progeny. These findings are supported by a signature of deregulated DNA-repair genes which strongly associates with low MYBL2 levels in MDS patient samples, providing a mechanistic understanding for the progression of blood disorders occurring during ageing. This study demonstrates a novel role for MYBL2 in DSB repair in HSCs and suggests that low levels of MYBL2 in human MDS could contribute to the emergence of further genetic abnormalities by deregulation of DNA-repair pathways.

Session topic: 9. Myelodysplastic syndromes - Biology

Keyword(s): Aging, MDS, Hematopoietic stem and progenitor cells, DNA repair

Abstract: S119

Type: Oral Presentation

Presentation during EHA22: On Friday, June 23, 2017 from 11:30 - 11:45

Location: Hall E

Background

MYBL2 is a transcription factor with roles in the cell cycle and genome integrity. MYBL2 is located on chromosome 20, within a region commonly deleted in human blood disorders (del20q). Our published data shows that reduced levels of MYBL2 predispose to development of myelodysplastic syndrome (MDS)-like disease in mouse models during ageing, indicating that MYBL2 could be acting as a tumour suppressor gene within del20q abnormality. Moreover, our previous work demonstrated that regardless of del20q deletion, MYBL2 expression is reduced in CD34+ bone marrow cells from MDS patients with worse prognosis. Because it has been shown that the cell of origin of MDS is the haematopoietic stem cell (HSC) and given the role of MYBL2 in DNA replication fork progression and maintenance of genome integrity, we hypothesised that low MYBL2 levels in HSC could contribute to elevated somatic mutations through changes in DNA repair pathways and drive disease development.

Aims

The aim of this study was to determine if low MYBL2 levels affect the double strand break (DSB) DNA repair damage response in HSC. 

Methods

In this study we used our mouse model in which animals express ~50% normal levels of MYBL2 (Mybl2+/∆). We characterised the ability of HSCs from young (7 weeks) and old (70 weeks) animals to respond to in vivo ionising radiation (2Gy) in terms of proliferation, apoptosis and colony forming ability. We measured the activation of the two main DNA repair pathways operating in the cells to deal with DSB: the error prone non-homologous-end-joining (NHEJ) and the error-free homologous recombination (HR) by assessing 53BP1 and Rad51 recruitment by immunofluorescence, respectively. Finally, we analysed the frequency of chromosome abnormalities present in the progeny of Mybl2+/∆ HSC that have previously been irradiated to determine the long term effects of changes in DNA repair. 

Results

We observed that Mybl2+/∆ HSCs had limited proliferative potential and displayed an increased sensitivity to ionizing radiation which increased during ageing. Mybl2+/∆ HSCs also displayed altered kinetics of 53BP1 and Rad51 recruitment and clearance, including retention of 53BP1 foci at later time points following irradiation and decreased levels of Rad51 foci when compared to Mybl2+/+ HSCs. Using plasmid functional assays, we showed that Mybl2+/∆ HSCs repair quite efficiently by NHEJ, but this efficiency is disrupted when cells are challenged with ionising radiation. Furthermore, Mybl2+/∆ HSCs have increased sensitivity to inhibition of DNA-PKC (required for NHEJ) but not ATM (required by HR). We also observed that after ionizing irradiation Mybl2+/∆ HSCs progeny displayed an increased percentage of chromatids with fragile telomeres. Moreover, by making use of publically available RNA-seq datasets from MDS patients, we have identified a clear association between low MYBL2 levels and low expression of DNA-repair genes in patients with worse prognosis. 

Conclusion
In summary, we have shown that decreased expression of MYBL2 leads to an imbalance in the DSB DNA-repair pathway choice, ultimately resulting in increased genomic instability of the blood cell progeny. These findings are supported by a signature of deregulated DNA-repair genes which strongly associates with low MYBL2 levels in MDS patient samples, providing a mechanistic understanding for the progression of blood disorders occurring during ageing. This study demonstrates a novel role for MYBL2 in DSB repair in HSCs and suggests that low levels of MYBL2 in human MDS could contribute to the emergence of further genetic abnormalities by deregulation of DNA-repair pathways.

Session topic: 9. Myelodysplastic syndromes - Biology

Keyword(s): Aging, MDS, Hematopoietic stem and progenitor cells, DNA repair

By clicking “Accept Terms & all Cookies” or by continuing to browse, you agree to the storing of third-party cookies on your device to enhance your user experience and agree to the user terms and conditions of this learning management system (LMS).

Cookie Settings
Accept Terms & all Cookies