Your browser doesn't support javascript.
loading
A heterochromatin inducing protein differentially recognizes self versus foreign genomes.
Burton, Eric M; Akinyemi, Ibukun A; Frey, Tiffany R; Xu, Huanzhou; Li, Xiaofan; Su, Lai Jing; Zhi, Jizu; McIntosh, Michael T; Bhaduri-McIntosh, Sumita.
Afiliação
  • Burton EM; Dept. of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, United States of America.
  • Akinyemi IA; Child Health Research Institute, Dept. of Pediatrics, University of Florida, Gainesville, Florida, United States of America.
  • Frey TR; Child Health Research Institute, Dept. of Pediatrics, University of Florida, Gainesville, Florida, United States of America.
  • Xu H; Division of Infectious Disease, Dept. of Pediatrics, University of Florida, Gainesville, Florida, United States of America.
  • Li X; Division of Infectious Disease, Dept. of Pediatrics, University of Florida, Gainesville, Florida, United States of America.
  • Su LJ; Child Health Research Institute, Dept. of Pediatrics, University of Florida, Gainesville, Florida, United States of America.
  • Zhi J; Dept of Pathology, Stony Brook University, Stony Brook, New York, United States of America.
  • McIntosh MT; Child Health Research Institute, Depts. of Pediatrics and of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America.
  • Bhaduri-McIntosh S; Division of Infectious Disease, Depts. of Pediatrics and of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America.
PLoS Pathog ; 17(3): e1009447, 2021 03.
Article em En | MEDLINE | ID: mdl-33730092
ABSTRACT
Krüppel-associated box-domain zinc finger protein (KRAB-ZFP) transcriptional repressors recruit TRIM28/KAP1 to heterochromatinize the mammalian genome while also guarding the host by silencing invading foreign genomes. However, how a KRAB-ZFP recognizes target sequences in the natural context of its own or foreign genomes is unclear. Our studies on B-lymphocytes permanently harboring the cancer-causing Epstein-Barr virus (EBV) have shown that SZF1, a KRAB-ZFP, binds to several lytic/replicative phase genes to silence them, thereby promoting the latent/quiescent phase of the virus. As a result, unless SZF1 and its binding partners are displaced from target regions on the viral genome, EBV remains dormant, i.e. refractory to lytic phase-inducing triggers. As SZF1 also heterochromatinizes the cellular genome, we performed in situ footprint mapping on both viral and host genomes in physically separated B-lymphocytes bearing latent or replicative/active EBV genomes. By analyzing footprints, we learned that SZF1 recognizes the host genome through a repeat sequence-bearing motif near centromeres. Remarkably, SZF1 does not use this motif to recognize the EBV genome. Instead, it uses distinct binding sites that lack obvious similarities to each other or the above motif, to silence the viral genome. Virus mutagenesis studies show that these distinct binding sites are not only key to maintaining the established latent phase but also silencing the lytic phase in newly-infected cells, thus enabling the virus to establish latency and transform cells. Notably, these binding sites on the viral genome, when also present on the human genome, are not used by SZF1 to silence host genes during latency. This differential approach towards target site recognition may reflect a strategy by which the host silences and regulates genomes of persistent invaders without jeopardizing its own homeostasis.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas Repressoras / Latência Viral / Infecções por Vírus Epstein-Barr / Inativação Gênica Limite: Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas Repressoras / Latência Viral / Infecções por Vírus Epstein-Barr / Inativação Gênica Limite: Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article