File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: APOBEC3B expression in breast cancer reflects cellular proliferation, while a deletion polymorphism is associated with immune activation

TitleAPOBEC3B expression in breast cancer reflects cellular proliferation, while a deletion polymorphism is associated with immune activation
Authors
KeywordsMutagenesis
Cancer
Cellular proliferation
Issue Date2015
Citation
Proceedings of the National Academy of Sciences of the United States of America, 2015, v. 112, n. 9, p. 2841-2846 How to Cite?
AbstractGenomic sequencing studies of breast and other cancers have identified patterns of mutations that have been attributed to the endogenous mutator activity of APOBEC3B (A3B), a member of the AID/APOBEC family of cytidine deaminases. A3B gene expression is increased in many cancers, but its upstream drivers remain undefined. Furthermore, there exists a common germ-line deletion polymorphism (A3Bdel), which has been associated with a paradoxical increase in breast cancer risk. To examine causes and consequences of A3B expression and its constitutive absence in breast cancer, we analyzed two large clinically annotated genomic datasets [The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC)]. We confirmed that A3B expression is associated with aggressive clinicopathologic characteristics and adverse outcomes and show that A3B expression is highly correlated with proliferative features (mitosis and cell cycle-related gene expression) in breast and 15 of 16 other solid tumor types. However, breast cancers arising in homozygous A3Bdel individuals with A3B absent did not differ in these features, indicating that A3B expression is a reflection rather than a direct cause of increased proliferation. Using gene set enrichment analysis (GSEA), we detected a pattern of immune activation in A3Bdel breast cancers, which seems to be related to hypermutation arising in A3Bdel carriers. Together, these results provide an explanation for A3B overexpression and its prognostic effect, giving context to additional study of this mutator as a cancer biomarker or putative drug target. In addition, although immune features of A3Bdel require additional study, these findings nominate the A3Bdel polymorphism as a potential predictor for cancer immunotherapy.
Persistent Identifierhttp://hdl.handle.net/10722/292867
ISSN
2023 Impact Factor: 9.4
2023 SCImago Journal Rankings: 3.737
PubMed Central ID
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorCescon, David W.-
dc.contributor.authorHaibe-Kains, Benjamin-
dc.contributor.authorMak, Tak W.-
dc.date.accessioned2020-11-17T14:57:23Z-
dc.date.available2020-11-17T14:57:23Z-
dc.date.issued2015-
dc.identifier.citationProceedings of the National Academy of Sciences of the United States of America, 2015, v. 112, n. 9, p. 2841-2846-
dc.identifier.issn0027-8424-
dc.identifier.urihttp://hdl.handle.net/10722/292867-
dc.description.abstractGenomic sequencing studies of breast and other cancers have identified patterns of mutations that have been attributed to the endogenous mutator activity of APOBEC3B (A3B), a member of the AID/APOBEC family of cytidine deaminases. A3B gene expression is increased in many cancers, but its upstream drivers remain undefined. Furthermore, there exists a common germ-line deletion polymorphism (A3Bdel), which has been associated with a paradoxical increase in breast cancer risk. To examine causes and consequences of A3B expression and its constitutive absence in breast cancer, we analyzed two large clinically annotated genomic datasets [The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC)]. We confirmed that A3B expression is associated with aggressive clinicopathologic characteristics and adverse outcomes and show that A3B expression is highly correlated with proliferative features (mitosis and cell cycle-related gene expression) in breast and 15 of 16 other solid tumor types. However, breast cancers arising in homozygous A3Bdel individuals with A3B absent did not differ in these features, indicating that A3B expression is a reflection rather than a direct cause of increased proliferation. Using gene set enrichment analysis (GSEA), we detected a pattern of immune activation in A3Bdel breast cancers, which seems to be related to hypermutation arising in A3Bdel carriers. Together, these results provide an explanation for A3B overexpression and its prognostic effect, giving context to additional study of this mutator as a cancer biomarker or putative drug target. In addition, although immune features of A3Bdel require additional study, these findings nominate the A3Bdel polymorphism as a potential predictor for cancer immunotherapy.-
dc.languageeng-
dc.relation.ispartofProceedings of the National Academy of Sciences of the United States of America-
dc.subjectMutagenesis-
dc.subjectCancer-
dc.subjectCellular proliferation-
dc.titleAPOBEC3B expression in breast cancer reflects cellular proliferation, while a deletion polymorphism is associated with immune activation-
dc.typeArticle-
dc.description.naturelink_to_OA_fulltext-
dc.identifier.doi10.1073/pnas.1424869112-
dc.identifier.pmid25730878-
dc.identifier.pmcidPMC4352793-
dc.identifier.scopuseid_2-s2.0-84924184255-
dc.identifier.volume112-
dc.identifier.issue9-
dc.identifier.spage2841-
dc.identifier.epage2846-
dc.identifier.eissn1091-6490-
dc.identifier.isiWOS:000350224900063-
dc.identifier.issnl0027-8424-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats