Your browser doesn't support javascript.
loading
Genome-wide CRISPR screening identifies tyrosylprotein sulfotransferase-2 as a target for augmenting anti-PD1 efficacy.
Oh, Yumi; Kim, Sujeong; Kim, Yunjae; Kim, Hyun; Jang, Dongjun; Shin, Seungjae; Lee, Soo-Jin; Kim, Jiwon; Lee, Sang Eun; Oh, Jaeik; Yang, Yoojin; Kim, Dohee; Jung, Hae Rim; Kim, Sangjin; Kim, Jihui; Min, Kyungchan; Cho, Beomki; Seo, Hoseok; Han, Dohyun; Park, Hansoo; Cho, Sung-Yup.
Afiliação
  • Oh Y; Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Kim S; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Korea.
  • Kim Y; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Korea.
  • Kim H; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Korea.
  • Jang D; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Shin S; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Lee SJ; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Kim J; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Lee SE; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Oh J; Department of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Yang Y; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Kim D; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Jung HR; Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, 03080, Korea.
  • Kim S; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Korea.
  • Kim J; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Korea.
  • Min K; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Korea.
  • Cho B; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Korea.
  • Seo H; Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, 03080, Korea.
  • Han D; Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, 08826, Korea.
  • Park H; Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, 03080, Korea.
  • Cho SY; Department of Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea.
Mol Cancer ; 23(1): 155, 2024 Aug 02.
Article em En | MEDLINE | ID: mdl-39095793
ABSTRACT

BACKGROUND:

Immune checkpoint therapy (ICT) provides durable responses in select cancer patients, yet resistance remains a significant challenge, prompting the exploration of underlying molecular mechanisms. Tyrosylprotein sulfotransferase-2 (TPST2), known for its role in protein tyrosine O-sulfation, has been suggested to modulate the extracellular protein-protein interactions, but its specific role in cancer immunity remains largely unexplored.

METHODS:

To explore tumor cell-intrinsic factors influencing anti-PD1 responsiveness, we conducted a pooled loss-of-function genetic screen in humanized mice engrafted with human immune cells. The responsiveness of cancer cells to interferon-γ (IFNγ) was estimated by evaluating IFNγ-mediated induction of target genes, STAT1 phosphorylation, HLA expression, and cell growth suppression. The sulfotyrosine-modified target gene of TPST2 was identified by co-immunoprecipitation and mass spectrometry. The in vivo effects of TPST2 inhibition were evaluated using mouse syngeneic tumor models and corroborated by bulk and single-cell RNA sequencing analyses.

RESULTS:

Through in vivo genome-wide CRISPR screening, TPST2 loss-of-function emerged as a potential enhancer of anti-PD1 treatment efficacy. TPST2 suppressed IFNγ signaling by sulfating IFNγ receptor 1 at Y397 residue, while its downregulation boosted IFNγ-mediated signaling and antigen presentation. Depletion of TPST2 in cancer cells augmented anti-PD1 antibody efficacy in syngeneic mouse tumor models by enhancing tumor-infiltrating lymphocytes. RNA sequencing data revealed TPST2's inverse correlation with antigen presentation, and increased TPST2 expression is associated with poor prognosis and altered cancer immunity across cancer types.

CONCLUSIONS:

We propose TPST2's novel role as a suppressor of cancer immunity and advocate for its consideration as a therapeutic target in ICT-based treatments.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Sulfotransferases / Receptor de Morte Celular Programada 1 Limite: Animals / Humans Idioma: En Revista: Mol Cancer Assunto da revista: NEOPLASIAS Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Sulfotransferases / Receptor de Morte Celular Programada 1 Limite: Animals / Humans Idioma: En Revista: Mol Cancer Assunto da revista: NEOPLASIAS Ano de publicação: 2024 Tipo de documento: Article