Your browser doesn't support javascript.
loading
MUC1-C Dictates PBRM1-Mediated Chronic Induction of Interferon Signaling, DNA Damage Resistance, and Immunosuppression in Triple-Negative Breast Cancer.
Yamashita, Nami; Morimoto, Yoshihiro; Fushimi, Atsushi; Ahmad, Rehan; Bhattacharya, Atrayee; Daimon, Tatsuaki; Haratake, Naoki; Inoue, Yuka; Ishikawa, Satoshi; Yamamoto, Masaaki; Hata, Tsuyoshi; Akiyoshi, Sayuri; Hu, Qiang; Liu, Tao; Withers, Henry; Liu, Song; Shapiro, Geoffrey I; Yoshizumi, Tomoharu; Long, Mark D; Kufe, Donald.
Affiliation
  • Yamashita N; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Morimoto Y; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Fushimi A; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Ahmad R; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Bhattacharya A; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Daimon T; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Haratake N; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Inoue Y; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
  • Ishikawa S; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Yamamoto M; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Hata T; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Akiyoshi S; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
  • Hu Q; Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Liu T; Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Withers H; Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Liu S; Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Shapiro GI; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
  • Yoshizumi T; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
  • Long MD; Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
  • Kufe D; Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts.
Mol Cancer Res ; 21(3): 274-289, 2023 03 01.
Article in En | MEDLINE | ID: mdl-36445328
ABSTRACT
The polybromo-1 (PBRM1) chromatin-targeting subunit of the SWI/SNF PBAF chromatin remodeling complex drives DNA damage resistance and immune evasion in certain cancer cells through mechanisms that remain unclear. STAT1 and IRF1 are essential effectors of type I and II IFN pathways. Here, we report that MUC1-C is necessary for PBRM1 expression and that it forms a nuclear complex with PBRM1 in triple-negative breast cancer (TNBC) cells. Analysis of global transcriptional (RNA-seq) and chromatin accessibility (ATAC-seq) profiles further demonstrated that MUC1-C and PBRM1 drive STAT1 and IRF1 expression by increasing chromatin accessibility of promoter-like signatures (PLS) on their respective genes. We also found that MUC1-C, PBRM1, and IRF1 increase the expression and chromatin accessibility on PLSs of the (i) type II IFN pathway IDO1 and WARS genes and (ii) type I IFN pathway RIG-I, MDA5, and ISG15 genes that collectively contribute to DNA damage resistance and immune evasion. In support of these results, targeting MUC1-C in wild-type BRCA TNBC cells enhanced carboplatin-induced DNA damage and the loss of self-renewal capacity. In addition, MUC1-C was necessary for DNA damage resistance, self-renewal, and tumorigenicity in olaparib-resistant BRCA1-mutant TNBC cells. Analysis of TNBC tumors corroborated that (i) MUC1 and PBRM1 are associated with decreased responsiveness to chemotherapy and (ii) MUC1-C expression is associated with the depletion of tumor-infiltrating lymphocytes (TIL). These findings demonstrate that MUC1-C activates PBRM1, and thereby chromatin remodeling of IFN-stimulated genes that promote chronic inflammation, DNA damage resistance, and immune evasion. IMPLICATIONS MUC1-C is necessary for PBRM1-driven chromatin remodeling in chronic activation of IFN pathway genes that promote DNA damage resistance and immunosuppression.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription Factors / Mucin-1 / Triple Negative Breast Neoplasms Limits: Humans Language: En Journal: Mol Cancer Res Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2023 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription Factors / Mucin-1 / Triple Negative Breast Neoplasms Limits: Humans Language: En Journal: Mol Cancer Res Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2023 Document type: Article