1. Academic Validation
  2. Cancer-derived exosomal HSPC111 promotes colorectal cancer liver metastasis by reprogramming lipid metabolism in cancer-associated fibroblasts

Cancer-derived exosomal HSPC111 promotes colorectal cancer liver metastasis by reprogramming lipid metabolism in cancer-associated fibroblasts

  • Cell Death Dis. 2022 Jan 13;13(1):57. doi: 10.1038/s41419-022-04506-4.
Chong Zhang  # 1 2 Xiang-Yu Wang  # 1 2 Peng Zhang  # 1 2 Tao-Chen He 1 2 Jia-Hao Han 1 2 Rui Zhang 1 2 Jing Lin 1 2 Jie Fan 3 Lu Lu 1 2 Wen-Wei Zhu 1 2 Hu-Liang Jia 1 2 Ju-Bo Zhang 4 Jin-Hong Chen 5 6
Affiliations

Affiliations

  • 1 Department of General Surgery, Huashan Hospital, Fudan University, 12 Wulumuqi Road (M), Shanghai, 200040, China.
  • 2 Institute of Cancer Metastasis, Fudan University, Shanghai, China.
  • 3 Department of Pathology, Huashan Hospital, Fudan University, 12 Wulumuqi Road (M), Shanghai, 200040, China.
  • 4 Department of Infectious Diseases, Huashan Hospital, Fudan University, 12 Wulumuqi Road (M), Shanghai, 200040, China.
  • 5 Department of General Surgery, Huashan Hospital, Fudan University, 12 Wulumuqi Road (M), Shanghai, 200040, China. jinhongchen@fudan.edu.cn.
  • 6 Institute of Cancer Metastasis, Fudan University, Shanghai, China. jinhongchen@fudan.edu.cn.
  • # Contributed equally.
Abstract

Tumor metastasis is a hallmark of Cancer. The communication between cancer-derived exosomes and stroma plays an irreplaceable role in facilitating pre-metastatic niche formation and Cancer metastasis. However, the mechanisms underlying exosome-mediated pre-metastatic niche formation during colorectal Cancer (CRC) liver metastasis remain incompletely understood. Here we identified HSPC111 was the leading upregulated gene in hepatic stellate cells (HSCs) incubated with CRC cell-derived exosomes. In xenograft mouse model, CRC cell-derived exosomal HSPC111 facilitated pre-metastatic niche formation and CRC liver metastases (CRLM). Consistently, CRC patients with liver metastasis had higher level of HSPC111 in serum exosomes, primary tumors and cancer-associated fibroblasts (CAFs) in liver metastasis than those without. Mechanistically, HSPC111 altered lipid metabolism of CAFs by phosphorylating ATP-citrate lyase (ACLY), which upregulated the level of acetyl-CoA. The accumulation of acetyl-CoA further promoted CXCL5 expression and secretion by increasing H3K27 acetylation in CAFs. Moreover, CXCL5-CXCR2 axis reinforced exosomal HSPC111 excretion from CRC cells and promoted liver metastasis. These results uncovered that CRC cell-derived exosomal HSPC111 promotes pre-metastatic niche formation and CRLM via reprogramming lipid metabolism in CAFs, and implicate HSPC111 may be a potential therapeutic target for preventing CRLM.

Figures
Products
Inhibitors & Agonists
Other Products