author_facet Liao, Lili
Alicea‐Velázquez, Nilda L.
Langbein, Lauren
Niu, Xiaohua
Cai, Weijia
Cho, Eun‐Ah
Zhang, Meiling
Greer, Celeste B.
Yan, Qin
Cosgrove, Michael S.
Yang, Haifeng
Liao, Lili
Alicea‐Velázquez, Nilda L.
Langbein, Lauren
Niu, Xiaohua
Cai, Weijia
Cho, Eun‐Ah
Zhang, Meiling
Greer, Celeste B.
Yan, Qin
Cosgrove, Michael S.
Yang, Haifeng
author Liao, Lili
Alicea‐Velázquez, Nilda L.
Langbein, Lauren
Niu, Xiaohua
Cai, Weijia
Cho, Eun‐Ah
Zhang, Meiling
Greer, Celeste B.
Yan, Qin
Cosgrove, Michael S.
Yang, Haifeng
spellingShingle Liao, Lili
Alicea‐Velázquez, Nilda L.
Langbein, Lauren
Niu, Xiaohua
Cai, Weijia
Cho, Eun‐Ah
Zhang, Meiling
Greer, Celeste B.
Yan, Qin
Cosgrove, Michael S.
Yang, Haifeng
Molecular Oncology
High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
Cancer Research
Genetics
Molecular Medicine
General Medicine
Oncology
author_sort liao, lili
spelling Liao, Lili Alicea‐Velázquez, Nilda L. Langbein, Lauren Niu, Xiaohua Cai, Weijia Cho, Eun‐Ah Zhang, Meiling Greer, Celeste B. Yan, Qin Cosgrove, Michael S. Yang, Haifeng 1574-7891 1878-0261 Wiley Cancer Research Genetics Molecular Medicine General Medicine Oncology http://dx.doi.org/10.1002/1878-0261.12434 <jats:p>Polybromo‐1 (<jats:styled-content style="fixed-case">PBRM</jats:styled-content>1) is an important tumor suppressor in kidney cancer. It contains six tandem bromodomains (<jats:styled-content style="fixed-case">BD</jats:styled-content>s), which are specialized structures that recognize acetyl‐lysine residues. While <jats:styled-content style="fixed-case">BD</jats:styled-content>2 has been found to bind acetylated histone H3 lysine 14 (H3K14ac), it is not known whether other <jats:styled-content style="fixed-case">BD</jats:styled-content>s collaborate with <jats:styled-content style="fixed-case">BD</jats:styled-content>2 to generate strong binding to H3K14ac, and the importance of H3K14ac recognition for the molecular and tumor suppressor function of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 is also unknown. We discovered that full‐length <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1, but not its individual <jats:styled-content style="fixed-case">BD</jats:styled-content>s, strongly binds H3K14ac. <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 were found to collaborate to facilitate strong binding to H3K14ac. Quantitative measurement of the interactions between purified <jats:styled-content style="fixed-case">BD</jats:styled-content> proteins and H3K14ac or nonacetylated peptides confirmed the tight and specific association of the former. Interestingly, while the structural integrity of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was found to be required for H3K14ac recognition, the conserved acetyl‐lysine binding site of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was not. Furthermore, simultaneous point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 prevented recognition of H3K14ac, altered promoter binding and gene expression, and caused <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to relocalize to the cytoplasm. In contrast, tumor‐derived point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone lowered <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1's affinity to H3K14ac and also disrupted promoter binding and gene expression without altering cellular localization. Finally, overexpression of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 variants containing point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 or <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone failed to suppress tumor growth in a xenograft model. Taken together, our study demonstrates that <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 collaborate to generate high affinity to H3K14ac and tether <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to chromatin. Mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone weaken these interactions, and this is sufficient to abolish its molecular and tumor suppressor functions.</jats:p> High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of <scp>PBRM</scp>1 Molecular Oncology
doi_str_mv 10.1002/1878-0261.12434
facet_avail Online
Free
finc_class_facet Biologie
Medizin
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi8xODc4LTAyNjEuMTI0MzQ
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi8xODc4LTAyNjEuMTI0MzQ
institution DE-105
DE-14
DE-Ch1
DE-L229
DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
imprint Wiley, 2019
imprint_str_mv Wiley, 2019
issn 1574-7891
1878-0261
issn_str_mv 1574-7891
1878-0261
language English
mega_collection Wiley (CrossRef)
match_str liao2019highaffinitybindingofh3k14acthroughcollaborationofbromodomains24and5iscriticalforthemolecularandtumorsuppressorfunctionsofpbrm1
publishDateSort 2019
publisher Wiley
recordtype ai
record_format ai
series Molecular Oncology
source_id 49
title High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_unstemmed High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_full High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_fullStr High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_full_unstemmed High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_short High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_sort high affinity binding of h3k14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of <scp>pbrm</scp>1
topic Cancer Research
Genetics
Molecular Medicine
General Medicine
Oncology
url http://dx.doi.org/10.1002/1878-0261.12434
publishDate 2019
physical 811-828
description <jats:p>Polybromo‐1 (<jats:styled-content style="fixed-case">PBRM</jats:styled-content>1) is an important tumor suppressor in kidney cancer. It contains six tandem bromodomains (<jats:styled-content style="fixed-case">BD</jats:styled-content>s), which are specialized structures that recognize acetyl‐lysine residues. While <jats:styled-content style="fixed-case">BD</jats:styled-content>2 has been found to bind acetylated histone H3 lysine 14 (H3K14ac), it is not known whether other <jats:styled-content style="fixed-case">BD</jats:styled-content>s collaborate with <jats:styled-content style="fixed-case">BD</jats:styled-content>2 to generate strong binding to H3K14ac, and the importance of H3K14ac recognition for the molecular and tumor suppressor function of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 is also unknown. We discovered that full‐length <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1, but not its individual <jats:styled-content style="fixed-case">BD</jats:styled-content>s, strongly binds H3K14ac. <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 were found to collaborate to facilitate strong binding to H3K14ac. Quantitative measurement of the interactions between purified <jats:styled-content style="fixed-case">BD</jats:styled-content> proteins and H3K14ac or nonacetylated peptides confirmed the tight and specific association of the former. Interestingly, while the structural integrity of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was found to be required for H3K14ac recognition, the conserved acetyl‐lysine binding site of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was not. Furthermore, simultaneous point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 prevented recognition of H3K14ac, altered promoter binding and gene expression, and caused <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to relocalize to the cytoplasm. In contrast, tumor‐derived point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone lowered <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1's affinity to H3K14ac and also disrupted promoter binding and gene expression without altering cellular localization. Finally, overexpression of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 variants containing point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 or <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone failed to suppress tumor growth in a xenograft model. Taken together, our study demonstrates that <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 collaborate to generate high affinity to H3K14ac and tether <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to chromatin. Mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone weaken these interactions, and this is sufficient to abolish its molecular and tumor suppressor functions.</jats:p>
container_issue 4
container_start_page 811
container_title Molecular Oncology
container_volume 13
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_ 1792342371870965762
geogr_code not assigned
last_indexed 2024-03-01T16:34:42.727Z
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=High+affinity+binding+of+H3K14ac+through+collaboration+of+bromodomains+2%2C+4+and+5+is+critical+for+the+molecular+and+tumor+suppressor+functions+of+PBRM1&rft.date=2019-04-01&genre=article&issn=1878-0261&volume=13&issue=4&spage=811&epage=828&pages=811-828&jtitle=Molecular+Oncology&atitle=High+affinity+binding+of+H3K14ac+through+collaboration+of+bromodomains+2%2C+4+and+5+is+critical+for+the+molecular+and+tumor+suppressor+functions+of+%3Cscp%3EPBRM%3C%2Fscp%3E1&aulast=Yang&aufirst=Haifeng&rft_id=info%3Adoi%2F10.1002%2F1878-0261.12434&rft.language%5B0%5D=eng
SOLR
_version_ 1792342371870965762
author Liao, Lili, Alicea‐Velázquez, Nilda L., Langbein, Lauren, Niu, Xiaohua, Cai, Weijia, Cho, Eun‐Ah, Zhang, Meiling, Greer, Celeste B., Yan, Qin, Cosgrove, Michael S., Yang, Haifeng
author_facet Liao, Lili, Alicea‐Velázquez, Nilda L., Langbein, Lauren, Niu, Xiaohua, Cai, Weijia, Cho, Eun‐Ah, Zhang, Meiling, Greer, Celeste B., Yan, Qin, Cosgrove, Michael S., Yang, Haifeng, Liao, Lili, Alicea‐Velázquez, Nilda L., Langbein, Lauren, Niu, Xiaohua, Cai, Weijia, Cho, Eun‐Ah, Zhang, Meiling, Greer, Celeste B., Yan, Qin, Cosgrove, Michael S., Yang, Haifeng
author_sort liao, lili
container_issue 4
container_start_page 811
container_title Molecular Oncology
container_volume 13
description <jats:p>Polybromo‐1 (<jats:styled-content style="fixed-case">PBRM</jats:styled-content>1) is an important tumor suppressor in kidney cancer. It contains six tandem bromodomains (<jats:styled-content style="fixed-case">BD</jats:styled-content>s), which are specialized structures that recognize acetyl‐lysine residues. While <jats:styled-content style="fixed-case">BD</jats:styled-content>2 has been found to bind acetylated histone H3 lysine 14 (H3K14ac), it is not known whether other <jats:styled-content style="fixed-case">BD</jats:styled-content>s collaborate with <jats:styled-content style="fixed-case">BD</jats:styled-content>2 to generate strong binding to H3K14ac, and the importance of H3K14ac recognition for the molecular and tumor suppressor function of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 is also unknown. We discovered that full‐length <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1, but not its individual <jats:styled-content style="fixed-case">BD</jats:styled-content>s, strongly binds H3K14ac. <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 were found to collaborate to facilitate strong binding to H3K14ac. Quantitative measurement of the interactions between purified <jats:styled-content style="fixed-case">BD</jats:styled-content> proteins and H3K14ac or nonacetylated peptides confirmed the tight and specific association of the former. Interestingly, while the structural integrity of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was found to be required for H3K14ac recognition, the conserved acetyl‐lysine binding site of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was not. Furthermore, simultaneous point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 prevented recognition of H3K14ac, altered promoter binding and gene expression, and caused <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to relocalize to the cytoplasm. In contrast, tumor‐derived point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone lowered <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1's affinity to H3K14ac and also disrupted promoter binding and gene expression without altering cellular localization. Finally, overexpression of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 variants containing point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 or <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone failed to suppress tumor growth in a xenograft model. Taken together, our study demonstrates that <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 collaborate to generate high affinity to H3K14ac and tether <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to chromatin. Mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone weaken these interactions, and this is sufficient to abolish its molecular and tumor suppressor functions.</jats:p>
doi_str_mv 10.1002/1878-0261.12434
facet_avail Online, Free
finc_class_facet Biologie, Medizin
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-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTAwMi8xODc4LTAyNjEuMTI0MzQ
imprint Wiley, 2019
imprint_str_mv Wiley, 2019
institution DE-105, DE-14, DE-Ch1, DE-L229, DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161, DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1
issn 1574-7891, 1878-0261
issn_str_mv 1574-7891, 1878-0261
language English
last_indexed 2024-03-01T16:34:42.727Z
match_str liao2019highaffinitybindingofh3k14acthroughcollaborationofbromodomains24and5iscriticalforthemolecularandtumorsuppressorfunctionsofpbrm1
mega_collection Wiley (CrossRef)
physical 811-828
publishDate 2019
publishDateSort 2019
publisher Wiley
record_format ai
recordtype ai
series Molecular Oncology
source_id 49
spelling Liao, Lili Alicea‐Velázquez, Nilda L. Langbein, Lauren Niu, Xiaohua Cai, Weijia Cho, Eun‐Ah Zhang, Meiling Greer, Celeste B. Yan, Qin Cosgrove, Michael S. Yang, Haifeng 1574-7891 1878-0261 Wiley Cancer Research Genetics Molecular Medicine General Medicine Oncology http://dx.doi.org/10.1002/1878-0261.12434 <jats:p>Polybromo‐1 (<jats:styled-content style="fixed-case">PBRM</jats:styled-content>1) is an important tumor suppressor in kidney cancer. It contains six tandem bromodomains (<jats:styled-content style="fixed-case">BD</jats:styled-content>s), which are specialized structures that recognize acetyl‐lysine residues. While <jats:styled-content style="fixed-case">BD</jats:styled-content>2 has been found to bind acetylated histone H3 lysine 14 (H3K14ac), it is not known whether other <jats:styled-content style="fixed-case">BD</jats:styled-content>s collaborate with <jats:styled-content style="fixed-case">BD</jats:styled-content>2 to generate strong binding to H3K14ac, and the importance of H3K14ac recognition for the molecular and tumor suppressor function of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 is also unknown. We discovered that full‐length <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1, but not its individual <jats:styled-content style="fixed-case">BD</jats:styled-content>s, strongly binds H3K14ac. <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 were found to collaborate to facilitate strong binding to H3K14ac. Quantitative measurement of the interactions between purified <jats:styled-content style="fixed-case">BD</jats:styled-content> proteins and H3K14ac or nonacetylated peptides confirmed the tight and specific association of the former. Interestingly, while the structural integrity of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was found to be required for H3K14ac recognition, the conserved acetyl‐lysine binding site of <jats:styled-content style="fixed-case">BD</jats:styled-content>4 was not. Furthermore, simultaneous point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 prevented recognition of H3K14ac, altered promoter binding and gene expression, and caused <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to relocalize to the cytoplasm. In contrast, tumor‐derived point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone lowered <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1's affinity to H3K14ac and also disrupted promoter binding and gene expression without altering cellular localization. Finally, overexpression of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 variants containing point mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 or <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone failed to suppress tumor growth in a xenograft model. Taken together, our study demonstrates that <jats:styled-content style="fixed-case">BD</jats:styled-content>s 2, 4, and 5 of <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 collaborate to generate high affinity to H3K14ac and tether <jats:styled-content style="fixed-case">PBRM</jats:styled-content>1 to chromatin. Mutations in <jats:styled-content style="fixed-case">BD</jats:styled-content>2 alone weaken these interactions, and this is sufficient to abolish its molecular and tumor suppressor functions.</jats:p> High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of <scp>PBRM</scp>1 Molecular Oncology
spellingShingle Liao, Lili, Alicea‐Velázquez, Nilda L., Langbein, Lauren, Niu, Xiaohua, Cai, Weijia, Cho, Eun‐Ah, Zhang, Meiling, Greer, Celeste B., Yan, Qin, Cosgrove, Michael S., Yang, Haifeng, Molecular Oncology, High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1, Cancer Research, Genetics, Molecular Medicine, General Medicine, Oncology
title High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_full High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_fullStr High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_full_unstemmed High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_short High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
title_sort high affinity binding of h3k14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of <scp>pbrm</scp>1
title_unstemmed High affinity binding of H3K14ac through collaboration of bromodomains 2, 4 and 5 is critical for the molecular and tumor suppressor functions of PBRM1
topic Cancer Research, Genetics, Molecular Medicine, General Medicine, Oncology
url http://dx.doi.org/10.1002/1878-0261.12434