PUBLICATION

Zebrafish Xenograft Model for Studying Pancreatic Cancer-Instructed Innate Immune Microenvironment

Authors
Wang, X., Li, W., Jiang, H., Ma, C., Huang, M., Wei, X., Wang, W., Jing, L.
ID
ZDB-PUB-220625-13
Date
2022
Source
International Journal of Molecular Sciences   23(12): (Journal)
Registered Authors
Keywords
innate immune microenvironment, pancreatic cancer, tumor associated myeloid cells, zebrafish xenograft
MeSH Terms
  • Animals
  • Carcinoma, Pancreatic Ductal*/pathology
  • Cell Line, Tumor
  • Disease Models, Animal
  • Heterografts
  • Humans
  • Immunity, Innate
  • Pancreatic Neoplasms*/pathology
  • Tumor Microenvironment
  • Zebrafish
PubMed
35742884 Full text @ Int. J. Mol. Sci.
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has up to half the tumor mass of tumor-associated myeloid cells. Myeloid innate immune cells play important roles in regulating cancer cell recognition and tumor growth. PDAC cells often mold myeloid cells into pro-tumoral state to fuel cancer growth and induce immune suppression. However, how tumor cells educate the innate immune responses remains largely unknown. In this study, we used four different human PDAC cell lines (PANC1, BxPC3, AsPC1, and CFPAC1) to establish the zebrafish xenograft model and investigated the interaction between pancreatic cancer and innate immune cells. The primary tumor-derived cancer cells PANC1 and BxPC3 activated innate immune anti-tumoral responses efficiently, while cancer cells from metastatic tissues AsPC1 and CFPAC1 induced an innate immune suppression and educated innate immune cells towards pro-tumoral state. Chemical conversion of innate immune cells to anti-tumoral state inhibited tumor growth for AsPC1 and CFPAC1. Moreover, genetic and pharmacological inhibition of macrophages also significantly reduced tumor growth, supporting the important roles of macrophages in innate immune suppression. REG4 expression is high in AsPC1 and CFPAC1. Knockdown of REG4 induced innate immune activation and reduced tumor growth in the xenografts, indicating that REG4 is a beneficial target for PDAC therapy. Our study provides a fast in-vivo model to study PDAC-innate immune interaction and their plasticity that could be used to study the related mechanism as well as identify new drugs to enhance immunotherapy.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping