author_facet Daskalakis, Michael
Brocks, David
Schmidt, Christopher
Li, Daofeng
Li, Jing
Jang, Hyo S.
Zhang, Bo
Lipka, Daniel B
Schott, Johanna
Bierhoff, Holger
Assenov, Yassen
Helf, Monika
Ressnerova, Alzbeta
Lindroth, Anders
Haas, Simon
Essers, Marieke A. G.
Imbusch, Charles D.
Brors, Benedikt
Oehme, Ina
Witt, Olaf
Lübbert, Michael
Stoecklin, Georg
Gerhäuser, Clarissa
Oakes, Christopher C
Wang, Ting
Plass, Christoph
Daskalakis, Michael
Brocks, David
Schmidt, Christopher
Li, Daofeng
Li, Jing
Jang, Hyo S.
Zhang, Bo
Lipka, Daniel B
Schott, Johanna
Bierhoff, Holger
Assenov, Yassen
Helf, Monika
Ressnerova, Alzbeta
Lindroth, Anders
Haas, Simon
Essers, Marieke A. G.
Imbusch, Charles D.
Brors, Benedikt
Oehme, Ina
Witt, Olaf
Lübbert, Michael
Stoecklin, Georg
Gerhäuser, Clarissa
Oakes, Christopher C
Wang, Ting
Plass, Christoph
author Daskalakis, Michael
Brocks, David
Schmidt, Christopher
Li, Daofeng
Li, Jing
Jang, Hyo S.
Zhang, Bo
Lipka, Daniel B
Schott, Johanna
Bierhoff, Holger
Assenov, Yassen
Helf, Monika
Ressnerova, Alzbeta
Lindroth, Anders
Haas, Simon
Essers, Marieke A. G.
Imbusch, Charles D.
Brors, Benedikt
Oehme, Ina
Witt, Olaf
Lübbert, Michael
Stoecklin, Georg
Gerhäuser, Clarissa
Oakes, Christopher C
Wang, Ting
Plass, Christoph
spellingShingle Daskalakis, Michael
Brocks, David
Schmidt, Christopher
Li, Daofeng
Li, Jing
Jang, Hyo S.
Zhang, Bo
Lipka, Daniel B
Schott, Johanna
Bierhoff, Holger
Assenov, Yassen
Helf, Monika
Ressnerova, Alzbeta
Lindroth, Anders
Haas, Simon
Essers, Marieke A. G.
Imbusch, Charles D.
Brors, Benedikt
Oehme, Ina
Witt, Olaf
Lübbert, Michael
Stoecklin, Georg
Gerhäuser, Clarissa
Oakes, Christopher C
Wang, Ting
Plass, Christoph
Blood
Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
Cell Biology
Hematology
Immunology
Biochemistry
author_sort daskalakis, michael
spelling Daskalakis, Michael Brocks, David Schmidt, Christopher Li, Daofeng Li, Jing Jang, Hyo S. Zhang, Bo Lipka, Daniel B Schott, Johanna Bierhoff, Holger Assenov, Yassen Helf, Monika Ressnerova, Alzbeta Lindroth, Anders Haas, Simon Essers, Marieke A. G. Imbusch, Charles D. Brors, Benedikt Oehme, Ina Witt, Olaf Lübbert, Michael Stoecklin, Georg Gerhäuser, Clarissa Oakes, Christopher C Wang, Ting Plass, Christoph 0006-4971 1528-0020 American Society of Hematology Cell Biology Hematology Immunology Biochemistry http://dx.doi.org/10.1182/blood.v128.22.3931.3931 <jats:title>Abstract</jats:title> <jats:p>Epigenetic drugs are currently used for the treatment of several hematologic malignancies, but their mechanism of action remains poorly understood. By using a previously described reporter cell line for epigenetic reactivation of the DAPK1 locus, we have shown that epigenetic treatment causes transcription from uncharacterized intronic transcription start sites (TSSs), thereby generating DAPK1 mRNA with novel first exons. Based on these findings, we analyzed whether inhibition of DNA-Methyltransferases (DNMTs), Histone deacetylases (HDACs), or both resulted in the genome-wide induction of non-canonical TSSs. While epigenetic treatment altered expression of known promoter sites, we observed that both HDAC- and DNMT-inhibitors predominantly induced de novo transcription from cryptic promoters encoded in long-terminal repeat (LTR) retrotransposons. These LTR-associated 'treatment induced, not-annotated TSS' (TINATs) are currently not annotated and normally silenced in almost all cell types with the exception of testicular und thymic tissue. In the majority of cases, these TINATs arose most commonly from LTR12 elements, particularly LTR12C (which apparently provides 50% of all TINATs). TINAT activation after DNMT-inhibitors (DNMTi) coincided with DNA hypomethylation and gain in H3K4me3, H3K9ac, and H3K27ac histone marks. In contrast, HDAC-inhibitors (HDACi) induced only canonical TSSs in association with histone acetylation, but TINATs via a yet unknown mechanism. Nevertheless, both inhibitors convergently induced unidirectional transcription from identical TINAT sites. Moreover, we found a consensus GATA2 binding motif which strongly distinguished LTR12Cs with TINATs from LTR12Cs without TINATs, supporting that GATA2 is likely the upstream transcription factor responsible for TINAT activation. TINATs originating from non-canonical TSSs located within introns of protein-coding genes frequently spliced into downstream exons thereby creating LTR/non-LTR fusion transcripts that harbor novel in place of canonical exon sequence at their 5' end. The resulting transcripts encode truncated or chimeric open reading frames which translated into currently uncharacterized protein isoforms with predicted abnormal functions or immunogenic potential, the last one based on their foreign sequence and capability of being presented on MHC-class I molecules. In summary, we could show that DNMTi and/or HDACi do not predominantly alter the expression of canonical genes, but induce de novo transcription of LTRs especially of the LTR12 family, resulting in numerous fusion transcripts that encode novel protein isoforms which might have the potential to influence cell proliferation or might be an elegant explanation for the priming effect of epigenetic therapy. Ongoing experiments are investigating the functional mechanisms of TINAT reactivation upon epigenetic drug treatment and future proteomic approaches combined with T-cell cytotoxicity assays will further shed light on the interaction between epigenetic and immune therapy and the role of ERV-derived antigen presentation.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>Lübbert: Janssen-Cilag: Other: Travel Funding, Research Funding; Ratiopharm: Other: Study drug valproic acid; Celgene: Other: Travel Funding.</jats:p> </jats:sec> Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats Blood
doi_str_mv 10.1182/blood.v128.22.3931.3931
facet_avail Online
Free
finc_class_facet Biologie
Medizin
Chemie und Pharmazie
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE4Mi9ibG9vZC52MTI4LjIyLjM5MzEuMzkzMQ
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE4Mi9ibG9vZC52MTI4LjIyLjM5MzEuMzkzMQ
institution DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
DE-L229
imprint American Society of Hematology, 2016
imprint_str_mv American Society of Hematology, 2016
issn 0006-4971
1528-0020
issn_str_mv 0006-4971
1528-0020
language English
mega_collection American Society of Hematology (CrossRef)
match_str daskalakis2016epigeneticdrugtreatmentgloballyinducescryptictranscriptionstartsitesencodedinlongterminalrepeats
publishDateSort 2016
publisher American Society of Hematology
recordtype ai
record_format ai
series Blood
source_id 49
title Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_unstemmed Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_full Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_fullStr Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_full_unstemmed Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_short Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_sort epigenetic drug treatment globally induces cryptic transcription start sites encoded in long terminal repeats
topic Cell Biology
Hematology
Immunology
Biochemistry
url http://dx.doi.org/10.1182/blood.v128.22.3931.3931
publishDate 2016
physical 3931-3931
description <jats:title>Abstract</jats:title> <jats:p>Epigenetic drugs are currently used for the treatment of several hematologic malignancies, but their mechanism of action remains poorly understood. By using a previously described reporter cell line for epigenetic reactivation of the DAPK1 locus, we have shown that epigenetic treatment causes transcription from uncharacterized intronic transcription start sites (TSSs), thereby generating DAPK1 mRNA with novel first exons. Based on these findings, we analyzed whether inhibition of DNA-Methyltransferases (DNMTs), Histone deacetylases (HDACs), or both resulted in the genome-wide induction of non-canonical TSSs. While epigenetic treatment altered expression of known promoter sites, we observed that both HDAC- and DNMT-inhibitors predominantly induced de novo transcription from cryptic promoters encoded in long-terminal repeat (LTR) retrotransposons. These LTR-associated 'treatment induced, not-annotated TSS' (TINATs) are currently not annotated and normally silenced in almost all cell types with the exception of testicular und thymic tissue. In the majority of cases, these TINATs arose most commonly from LTR12 elements, particularly LTR12C (which apparently provides 50% of all TINATs). TINAT activation after DNMT-inhibitors (DNMTi) coincided with DNA hypomethylation and gain in H3K4me3, H3K9ac, and H3K27ac histone marks. In contrast, HDAC-inhibitors (HDACi) induced only canonical TSSs in association with histone acetylation, but TINATs via a yet unknown mechanism. Nevertheless, both inhibitors convergently induced unidirectional transcription from identical TINAT sites. Moreover, we found a consensus GATA2 binding motif which strongly distinguished LTR12Cs with TINATs from LTR12Cs without TINATs, supporting that GATA2 is likely the upstream transcription factor responsible for TINAT activation. TINATs originating from non-canonical TSSs located within introns of protein-coding genes frequently spliced into downstream exons thereby creating LTR/non-LTR fusion transcripts that harbor novel in place of canonical exon sequence at their 5' end. The resulting transcripts encode truncated or chimeric open reading frames which translated into currently uncharacterized protein isoforms with predicted abnormal functions or immunogenic potential, the last one based on their foreign sequence and capability of being presented on MHC-class I molecules. In summary, we could show that DNMTi and/or HDACi do not predominantly alter the expression of canonical genes, but induce de novo transcription of LTRs especially of the LTR12 family, resulting in numerous fusion transcripts that encode novel protein isoforms which might have the potential to influence cell proliferation or might be an elegant explanation for the priming effect of epigenetic therapy. Ongoing experiments are investigating the functional mechanisms of TINAT reactivation upon epigenetic drug treatment and future proteomic approaches combined with T-cell cytotoxicity assays will further shed light on the interaction between epigenetic and immune therapy and the role of ERV-derived antigen presentation.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>Lübbert: Janssen-Cilag: Other: Travel Funding, Research Funding; Ratiopharm: Other: Study drug valproic acid; Celgene: Other: Travel Funding.</jats:p> </jats:sec>
container_issue 22
container_start_page 3931
container_title Blood
container_volume 128
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792324450972073993
geogr_code not assigned
last_indexed 2024-03-01T11:49:55.444Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=Epigenetic+Drug+Treatment+Globally+Induces+Cryptic+Transcription+Start+Sites+Encoded+in+Long+Terminal+Repeats&rft.date=2016-12-02&genre=article&issn=1528-0020&volume=128&issue=22&spage=3931&epage=3931&pages=3931-3931&jtitle=Blood&atitle=Epigenetic+Drug+Treatment+Globally+Induces+Cryptic+Transcription+Start+Sites+Encoded+in+Long+Terminal+Repeats&aulast=Plass&aufirst=Christoph&rft_id=info%3Adoi%2F10.1182%2Fblood.v128.22.3931.3931&rft.language%5B0%5D=eng
SOLR
_version_ 1792324450972073993
author Daskalakis, Michael, Brocks, David, Schmidt, Christopher, Li, Daofeng, Li, Jing, Jang, Hyo S., Zhang, Bo, Lipka, Daniel B, Schott, Johanna, Bierhoff, Holger, Assenov, Yassen, Helf, Monika, Ressnerova, Alzbeta, Lindroth, Anders, Haas, Simon, Essers, Marieke A. G., Imbusch, Charles D., Brors, Benedikt, Oehme, Ina, Witt, Olaf, Lübbert, Michael, Stoecklin, Georg, Gerhäuser, Clarissa, Oakes, Christopher C, Wang, Ting, Plass, Christoph
author_facet Daskalakis, Michael, Brocks, David, Schmidt, Christopher, Li, Daofeng, Li, Jing, Jang, Hyo S., Zhang, Bo, Lipka, Daniel B, Schott, Johanna, Bierhoff, Holger, Assenov, Yassen, Helf, Monika, Ressnerova, Alzbeta, Lindroth, Anders, Haas, Simon, Essers, Marieke A. G., Imbusch, Charles D., Brors, Benedikt, Oehme, Ina, Witt, Olaf, Lübbert, Michael, Stoecklin, Georg, Gerhäuser, Clarissa, Oakes, Christopher C, Wang, Ting, Plass, Christoph, Daskalakis, Michael, Brocks, David, Schmidt, Christopher, Li, Daofeng, Li, Jing, Jang, Hyo S., Zhang, Bo, Lipka, Daniel B, Schott, Johanna, Bierhoff, Holger, Assenov, Yassen, Helf, Monika, Ressnerova, Alzbeta, Lindroth, Anders, Haas, Simon, Essers, Marieke A. G., Imbusch, Charles D., Brors, Benedikt, Oehme, Ina, Witt, Olaf, Lübbert, Michael, Stoecklin, Georg, Gerhäuser, Clarissa, Oakes, Christopher C, Wang, Ting, Plass, Christoph
author_sort daskalakis, michael
container_issue 22
container_start_page 3931
container_title Blood
container_volume 128
description <jats:title>Abstract</jats:title> <jats:p>Epigenetic drugs are currently used for the treatment of several hematologic malignancies, but their mechanism of action remains poorly understood. By using a previously described reporter cell line for epigenetic reactivation of the DAPK1 locus, we have shown that epigenetic treatment causes transcription from uncharacterized intronic transcription start sites (TSSs), thereby generating DAPK1 mRNA with novel first exons. Based on these findings, we analyzed whether inhibition of DNA-Methyltransferases (DNMTs), Histone deacetylases (HDACs), or both resulted in the genome-wide induction of non-canonical TSSs. While epigenetic treatment altered expression of known promoter sites, we observed that both HDAC- and DNMT-inhibitors predominantly induced de novo transcription from cryptic promoters encoded in long-terminal repeat (LTR) retrotransposons. These LTR-associated 'treatment induced, not-annotated TSS' (TINATs) are currently not annotated and normally silenced in almost all cell types with the exception of testicular und thymic tissue. In the majority of cases, these TINATs arose most commonly from LTR12 elements, particularly LTR12C (which apparently provides 50% of all TINATs). TINAT activation after DNMT-inhibitors (DNMTi) coincided with DNA hypomethylation and gain in H3K4me3, H3K9ac, and H3K27ac histone marks. In contrast, HDAC-inhibitors (HDACi) induced only canonical TSSs in association with histone acetylation, but TINATs via a yet unknown mechanism. Nevertheless, both inhibitors convergently induced unidirectional transcription from identical TINAT sites. Moreover, we found a consensus GATA2 binding motif which strongly distinguished LTR12Cs with TINATs from LTR12Cs without TINATs, supporting that GATA2 is likely the upstream transcription factor responsible for TINAT activation. TINATs originating from non-canonical TSSs located within introns of protein-coding genes frequently spliced into downstream exons thereby creating LTR/non-LTR fusion transcripts that harbor novel in place of canonical exon sequence at their 5' end. The resulting transcripts encode truncated or chimeric open reading frames which translated into currently uncharacterized protein isoforms with predicted abnormal functions or immunogenic potential, the last one based on their foreign sequence and capability of being presented on MHC-class I molecules. In summary, we could show that DNMTi and/or HDACi do not predominantly alter the expression of canonical genes, but induce de novo transcription of LTRs especially of the LTR12 family, resulting in numerous fusion transcripts that encode novel protein isoforms which might have the potential to influence cell proliferation or might be an elegant explanation for the priming effect of epigenetic therapy. Ongoing experiments are investigating the functional mechanisms of TINAT reactivation upon epigenetic drug treatment and future proteomic approaches combined with T-cell cytotoxicity assays will further shed light on the interaction between epigenetic and immune therapy and the role of ERV-derived antigen presentation.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>Lübbert: Janssen-Cilag: Other: Travel Funding, Research Funding; Ratiopharm: Other: Study drug valproic acid; Celgene: Other: Travel Funding.</jats:p> </jats:sec>
doi_str_mv 10.1182/blood.v128.22.3931.3931
facet_avail Online, Free
finc_class_facet Biologie, Medizin, Chemie und Pharmazie
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE4Mi9ibG9vZC52MTI4LjIyLjM5MzEuMzkzMQ
imprint American Society of Hematology, 2016
imprint_str_mv American Society of Hematology, 2016
institution DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161, DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1, DE-L229
issn 0006-4971, 1528-0020
issn_str_mv 0006-4971, 1528-0020
language English
last_indexed 2024-03-01T11:49:55.444Z
match_str daskalakis2016epigeneticdrugtreatmentgloballyinducescryptictranscriptionstartsitesencodedinlongterminalrepeats
mega_collection American Society of Hematology (CrossRef)
physical 3931-3931
publishDate 2016
publishDateSort 2016
publisher American Society of Hematology
record_format ai
recordtype ai
series Blood
source_id 49
spelling Daskalakis, Michael Brocks, David Schmidt, Christopher Li, Daofeng Li, Jing Jang, Hyo S. Zhang, Bo Lipka, Daniel B Schott, Johanna Bierhoff, Holger Assenov, Yassen Helf, Monika Ressnerova, Alzbeta Lindroth, Anders Haas, Simon Essers, Marieke A. G. Imbusch, Charles D. Brors, Benedikt Oehme, Ina Witt, Olaf Lübbert, Michael Stoecklin, Georg Gerhäuser, Clarissa Oakes, Christopher C Wang, Ting Plass, Christoph 0006-4971 1528-0020 American Society of Hematology Cell Biology Hematology Immunology Biochemistry http://dx.doi.org/10.1182/blood.v128.22.3931.3931 <jats:title>Abstract</jats:title> <jats:p>Epigenetic drugs are currently used for the treatment of several hematologic malignancies, but their mechanism of action remains poorly understood. By using a previously described reporter cell line for epigenetic reactivation of the DAPK1 locus, we have shown that epigenetic treatment causes transcription from uncharacterized intronic transcription start sites (TSSs), thereby generating DAPK1 mRNA with novel first exons. Based on these findings, we analyzed whether inhibition of DNA-Methyltransferases (DNMTs), Histone deacetylases (HDACs), or both resulted in the genome-wide induction of non-canonical TSSs. While epigenetic treatment altered expression of known promoter sites, we observed that both HDAC- and DNMT-inhibitors predominantly induced de novo transcription from cryptic promoters encoded in long-terminal repeat (LTR) retrotransposons. These LTR-associated 'treatment induced, not-annotated TSS' (TINATs) are currently not annotated and normally silenced in almost all cell types with the exception of testicular und thymic tissue. In the majority of cases, these TINATs arose most commonly from LTR12 elements, particularly LTR12C (which apparently provides 50% of all TINATs). TINAT activation after DNMT-inhibitors (DNMTi) coincided with DNA hypomethylation and gain in H3K4me3, H3K9ac, and H3K27ac histone marks. In contrast, HDAC-inhibitors (HDACi) induced only canonical TSSs in association with histone acetylation, but TINATs via a yet unknown mechanism. Nevertheless, both inhibitors convergently induced unidirectional transcription from identical TINAT sites. Moreover, we found a consensus GATA2 binding motif which strongly distinguished LTR12Cs with TINATs from LTR12Cs without TINATs, supporting that GATA2 is likely the upstream transcription factor responsible for TINAT activation. TINATs originating from non-canonical TSSs located within introns of protein-coding genes frequently spliced into downstream exons thereby creating LTR/non-LTR fusion transcripts that harbor novel in place of canonical exon sequence at their 5' end. The resulting transcripts encode truncated or chimeric open reading frames which translated into currently uncharacterized protein isoforms with predicted abnormal functions or immunogenic potential, the last one based on their foreign sequence and capability of being presented on MHC-class I molecules. In summary, we could show that DNMTi and/or HDACi do not predominantly alter the expression of canonical genes, but induce de novo transcription of LTRs especially of the LTR12 family, resulting in numerous fusion transcripts that encode novel protein isoforms which might have the potential to influence cell proliferation or might be an elegant explanation for the priming effect of epigenetic therapy. Ongoing experiments are investigating the functional mechanisms of TINAT reactivation upon epigenetic drug treatment and future proteomic approaches combined with T-cell cytotoxicity assays will further shed light on the interaction between epigenetic and immune therapy and the role of ERV-derived antigen presentation.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>Lübbert: Janssen-Cilag: Other: Travel Funding, Research Funding; Ratiopharm: Other: Study drug valproic acid; Celgene: Other: Travel Funding.</jats:p> </jats:sec> Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats Blood
spellingShingle Daskalakis, Michael, Brocks, David, Schmidt, Christopher, Li, Daofeng, Li, Jing, Jang, Hyo S., Zhang, Bo, Lipka, Daniel B, Schott, Johanna, Bierhoff, Holger, Assenov, Yassen, Helf, Monika, Ressnerova, Alzbeta, Lindroth, Anders, Haas, Simon, Essers, Marieke A. G., Imbusch, Charles D., Brors, Benedikt, Oehme, Ina, Witt, Olaf, Lübbert, Michael, Stoecklin, Georg, Gerhäuser, Clarissa, Oakes, Christopher C, Wang, Ting, Plass, Christoph, Blood, Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats, Cell Biology, Hematology, Immunology, Biochemistry
title Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_full Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_fullStr Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_full_unstemmed Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_short Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
title_sort epigenetic drug treatment globally induces cryptic transcription start sites encoded in long terminal repeats
title_unstemmed Epigenetic Drug Treatment Globally Induces Cryptic Transcription Start Sites Encoded in Long Terminal Repeats
topic Cell Biology, Hematology, Immunology, Biochemistry
url http://dx.doi.org/10.1182/blood.v128.22.3931.3931