1. Academic Validation
  2. Neutrophils resist ferroptosis and promote breast cancer metastasis through aconitate decarboxylase 1

Neutrophils resist ferroptosis and promote breast cancer metastasis through aconitate decarboxylase 1

  • Cell Metab. 2023 Oct 3;35(10):1688-1703.e10. doi: 10.1016/j.cmet.2023.09.004.
Yun Zhao 1 Zhongshun Liu 1 Guoqiang Liu 2 Yuting Zhang 2 Sheng Liu 3 Dailin Gan 4 Wennan Chang 5 Xiaoxia Peng 1 Eun Suh Sung 1 Keegan Gilbert 1 Yini Zhu 2 Xuechun Wang 1 Ziyu Zeng 1 Hope Baldwin 1 Guanzhu Ren 1 Jessica Weaver 1 Anna Huron 1 Toni Mayberry 6 Qingfei Wang 7 Yujue Wang 8 Maria Elena Diaz-Rubio 8 Xiaoyang Su 9 M Sharon Stack 10 Siyuan Zhang 11 Xuemin Lu 1 Ryan D Sheldon 12 Jun Li 4 Chi Zhang 5 Jun Wan 13 Xin Lu 14
Affiliations

Affiliations

  • 1 Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA.
  • 2 Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA.
  • 3 Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
  • 4 Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA.
  • 5 Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Electrical and Computer Engineering, Purdue University, Indianapolis, IN 46202, USA.
  • 6 Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA.
  • 7 Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
  • 8 Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA.
  • 9 Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
  • 10 Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
  • 11 Department of Pathology, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
  • 12 Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI, USA.
  • 13 Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; School of Informatics and Computing, Indiana University - Purdue University at Indianapolis, Indianapolis, IN 46202, USA.
  • 14 Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA; Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA. Electronic address: xlu@nd.edu.
Abstract

Metastasis causes breast cancer-related mortality. Tumor-infiltrating neutrophils (TINs) inflict immunosuppression and promote metastasis. Therapeutic debilitation of TINs may enhance immunotherapy, yet it remains a challenge to identify therapeutic targets highly expressed and functionally essential in TINs but under-expressed in extra-tumoral neutrophils. Here, using single-cell RNA sequencing to compare TINs and circulating neutrophils in murine mammary tumor models, we identified aconitate decarboxylase 1 (Acod1) as the most upregulated metabolic Enzyme in mouse TINs and validated high Acod1 expression in human TINs. Activated through the GM-CSF-JAK/STAT5-C/EBPβ pathway, Acod1 produces itaconate, which mediates Nrf2-dependent defense against Ferroptosis and upholds the persistence of TINs. Acod1 ablation abates TIN infiltration, constrains metastasis (but not primary tumors), bolsters antitumor T cell immunity, and boosts the efficacy of immune checkpoint blockade. Our findings reveal how TINs escape from Ferroptosis through the Acod1-dependent immunometabolism switch and establish Acod1 as a target to offset immunosuppression and improve immunotherapy against metastasis.

Keywords

Acod1; MDSC; breast cancer; ferroptosis; immune checkpoint blockade; immune metabolism; itaconate; metastasis; neutrophil; single-cell RNA sequencing.

Figures
Products