Apposition of Fibroblasts With Metaplastic Gastric Cells Promotes Dysplastic Transition

Su Hyung Lee, Ela W. Contreras Panta, David Gibbs, Yoonkyung Won, Jimin Min, Changqing Zhang, Joseph T. Roland, Se Hoon Hong, Yoojin Sohn, Evan Krystofiak, Bogun Jang, Lorenzo Ferri, Veena Sangwan, Jiannis Ragoussis, Sophie Camilleri-Broët, Joseph Caruso, Chira Chen-Tanyolac, Michael Strasser, Philippe Gascard, Thea D. TlstySui Huang, Eunyoung Choi, James R. Goldenring

Research output: Contribution to journalArticlepeer-review

7 Scopus citations

Abstract

Background & Aims: Elements of field cancerization, including atrophic gastritis, metaplasia, and dysplasia, promote gastric cancer development in association with chronic inflammation. However, it remains unclear how stroma changes during carcinogenesis and how the stroma contributes to progression of gastric preneoplasia. Here we investigated heterogeneity of fibroblasts, one of the most important elements in the stroma, and their roles in neoplastic transformation of metaplasia. Methods: We used single-cell transcriptomics to evaluate the cellular heterogeneity of mucosal cells from patients with gastric cancer. Tissue sections from the same cohort and tissue microarrays were used to identify the geographical distribution of distinct fibroblast subsets. We further evaluated the role of fibroblasts from pathologic mucosa in dysplastic progression of metaplastic cells using patient-derived metaplastic gastroids and fibroblasts. Results: We identified 4 subsets of fibroblasts within stromal cells defined by the differential expression of PDGFRA, FBLN2, ACTA2, or PDGFRB. Each subset was distributed distinctively throughout stomach tissues with different proportions at each pathologic stage. The PDGFRα+ subset expanded in metaplasia and cancer compared with normal, maintaining a close proximity with the epithelial compartment. Co-culture of metaplasia- or cancer-derived fibroblasts with gastroids showing the characteristics of spasmolytic polypeptide-expressing metaplasia–induced disordered growth, loss of metaplastic markers, and increases in markers of dysplasia. Culture of metaplastic gastroids with conditioned media from metaplasia- or cancer-derived fibroblasts also promoted dysplastic transition. Conclusions: These findings indicate that fibroblast associations with metaplastic epithelial cells can facilitate direct transition of metaplastic spasmolytic polypeptide-expressing metaplasia cell lineages into dysplastic lineages.

Original languageEnglish (US)
Pages (from-to)374-390
Number of pages17
JournalGastroenterology
Volume165
Issue number2
DOIs
StatePublished - Aug 2023
Externally publishedYes

Keywords

  • Fibroblasts
  • Gastric Carcinogenesis
  • Metaplasia
  • PDGFRA
  • SPEM

ASJC Scopus subject areas

  • Hepatology
  • Gastroenterology

Fingerprint

Dive into the research topics of 'Apposition of Fibroblasts With Metaplastic Gastric Cells Promotes Dysplastic Transition'. Together they form a unique fingerprint.

Cite this