File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Inhibition of CAF-1 histone chaperone complex triggers cytosolic DNA and dsRNA sensing pathways and induces intrinsic immunity of hepatocellular carcinoma

TitleInhibition of CAF-1 histone chaperone complex triggers cytosolic DNA and dsRNA sensing pathways and induces intrinsic immunity of hepatocellular carcinoma
Authors
Issue Date5-Dec-2023
PublisherWolters Kluwer Health Inc.
Citation
Hepatology, 2023, v. 80, n. 2, p. 295-311 How to Cite?
Abstract

Background and Aims: 

Chromatin assembly factor 1 (CAF-1) is a replication-dependent epigenetic regulator that controls cell cycle progression and chromatin dynamics. In this study, we aim to investigate the immunomodulatory role and therapeutic potential of the CAF-1 complex in HCC.

Approach and Results: 

CAF-1 complex knockout cell lines were established using the CRISPR/Cas9 system. The effects of CAF-1 in HCC were studied in HCC cell lines, nude mice, and immunocompetent mice. RNA-sequencing, ChIP-Seq, and assay for transposase accessible chromatin with high-throughput sequencing (ATAC-Seq) were used to explore the changes in the epigenome and transcriptome. CAF-1 complex was significantly upregulated in human and mouse HCCs and was associated with poor prognosis in patients with HCC. Knockout of CAF-1 remarkably suppressed HCC growth in both in vitro and in vivo models. Mechanistically, depletion of CAF-1 induced replicative stress and chromatin instability, which eventually led to cytoplasmic DNA leakage as micronuclei. Also, chromatin immunoprecipitation sequencing analyses revealed a massive H3.3 histone variant replacement upon CAF-1 knockout. Enrichment of euchromatic H3.3 increased chromatin accessibility and activated the expression of endogenous retrovirus elements, a phenomenon known as viral mimicry. However, cytosolic micronuclei and endogenous retroviruses are recognized as ectopic elements by the stimulator of interferon genes and dsRNA viral sensing pathways, respectively. As a result, the knockout of CAF-1 activated inflammatory response and antitumor immune surveillance and thereby significantly enhanced the anticancer effect of immune checkpoint inhibitors in HCC.

Conclusions: 

Our findings suggest that CAF-1 is essential for HCC development; targeting CAF-1 may awaken the anticancer immune response and may work cooperatively with immune checkpoint inhibitor treatment in cancer therapy.


Persistent Identifierhttp://hdl.handle.net/10722/346516
ISSN
2023 Impact Factor: 12.9
2023 SCImago Journal Rankings: 5.011

 

DC FieldValueLanguage
dc.contributor.authorChan, For-Fan-
dc.contributor.authorYuen, Vincent Wai-Hin-
dc.contributor.authorShen, Jialing-
dc.contributor.authorChin, Don Wai-Ching-
dc.contributor.authorLaw, Cheuk-Ting-
dc.contributor.authorWong, Bowie Po-Yee-
dc.contributor.authorChan, Cerise Yuen-Ki-
dc.contributor.authorCheu, Jacinth Wing-Sum-
dc.contributor.authorNg, Irene Oi-Lin-
dc.contributor.authorWong, Carmen Chak-Lui-
dc.contributor.authorWong, Chun-Ming-
dc.date.accessioned2024-09-17T00:31:08Z-
dc.date.available2024-09-17T00:31:08Z-
dc.date.issued2023-12-05-
dc.identifier.citationHepatology, 2023, v. 80, n. 2, p. 295-311-
dc.identifier.issn0270-9139-
dc.identifier.urihttp://hdl.handle.net/10722/346516-
dc.description.abstract<h3>Background and Aims: </h3><p>Chromatin assembly factor 1 (CAF-1) is a replication-dependent epigenetic regulator that controls cell cycle progression and chromatin dynamics. In this study, we aim to investigate the immunomodulatory role and therapeutic potential of the CAF-1 complex in HCC.</p><h3>Approach and Results: </h3><p>CAF-1 complex knockout cell lines were established using the CRISPR/Cas9 system. The effects of CAF-1 in HCC were studied in HCC cell lines, nude mice, and immunocompetent mice. RNA-sequencing, ChIP-Seq, and assay for transposase accessible chromatin with high-throughput sequencing (ATAC-Seq) were used to explore the changes in the epigenome and transcriptome. CAF-1 complex was significantly upregulated in human and mouse HCCs and was associated with poor prognosis in patients with HCC. Knockout of CAF-1 remarkably suppressed HCC growth in both in vitro and in vivo models. Mechanistically, depletion of CAF-1 induced replicative stress and chromatin instability, which eventually led to cytoplasmic DNA leakage as micronuclei. Also, chromatin immunoprecipitation sequencing analyses revealed a massive H3.3 histone variant replacement upon CAF-1 knockout. Enrichment of euchromatic H3.3 increased chromatin accessibility and activated the expression of endogenous retrovirus elements, a phenomenon known as viral mimicry. However, cytosolic micronuclei and endogenous retroviruses are recognized as ectopic elements by the stimulator of interferon genes and dsRNA viral sensing pathways, respectively. As a result, the knockout of CAF-1 activated inflammatory response and antitumor immune surveillance and thereby significantly enhanced the anticancer effect of immune checkpoint inhibitors in HCC.</p><h3>Conclusions: </h3><p>Our findings suggest that CAF-1 is essential for HCC development; targeting CAF-1 may awaken the anticancer immune response and may work cooperatively with immune checkpoint inhibitor treatment in cancer therapy.</p>-
dc.languageeng-
dc.publisherWolters Kluwer Health Inc.-
dc.relation.ispartofHepatology-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.titleInhibition of CAF-1 histone chaperone complex triggers cytosolic DNA and dsRNA sensing pathways and induces intrinsic immunity of hepatocellular carcinoma-
dc.typeArticle-
dc.identifier.doi10.1097/HEP.0000000000000709-
dc.identifier.volume80-
dc.identifier.issue2-
dc.identifier.spage295-
dc.identifier.epage311-
dc.identifier.eissn1527-3350-
dc.identifier.issnl0270-9139-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats