Your browser doesn't support javascript.
loading
Ex vivo model of breast cancer cell invasion in live lymph node tissue.
Morgaenko, Katerina; Arneja, Abhinav; Ball, Alexander G; Putelo, Audrey M; Munson, Jennifer M; Rutkowski, Melanie R; Pompano, Rebecca R.
Afiliação
  • Morgaenko K; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.
  • Arneja A; Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States.
  • Ball AG; Department of Pathology, University of Virginia, Charlottesville, VA, United States.
  • Putelo AM; Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States.
  • Munson JM; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States.
  • Rutkowski MR; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States.
  • Pompano RR; Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA, United States.
bioRxiv ; 2024 Jul 22.
Article em En | MEDLINE | ID: mdl-39091774
ABSTRACT
Lymph nodes (LNs) are common sites of metastatic invasion in breast cancer, often preceding spread to distant organs and serving as key indicators of clinical disease progression. However, the mechanisms of cancer cell invasion into LNs are not well understood. Existing in vivo models struggle to isolate the specific impacts of the tumor-draining lymph node (TDLN) milieu on cancer cell invasion due to the co-evolving relationship between TDLNs and the upstream tumor. To address these limitations, we used live ex vivo LN tissue slices with intact chemotactic function to model cancer cell spread within a spatially organized microenvironment. After showing that BRPKp110 breast cancer cells were chemoattracted to factors secreted by naïve LN tissue in a 3D migration assay, we demonstrated that ex vivo LN slices could support cancer cell seeding, invasion, and spread. This novel approach revealed dynamic, preferential cancer cell invasion within specific anatomical regions of LNs, particularly the subcapsular sinus (SCS) and cortex, as well as chemokine-rich domains of immobilized CXCL13 and CCL1. While CXCR5 was necessary for a portion of BRPKp110 invasion into naïve LNs, disruption of CXCR5/CXCL13 signaling alone was insufficient to prevent invasion towards CXCL13-rich domains. Finally, we extended this system to pre-metastatic TDLNs, where the ex vivo model predicted a lower invasion of cancer cells. The reduced invasion was not due to diminished chemokine secretion, but it correlated with elevated intranodal IL-21. In summary, this innovative ex vivo model of cancer cell spread in live LN slices provides a platform to investigate cancer invasion within the intricate tissue microenvironment, supporting time-course analysis and parallel read-outs. We anticipate that this system will enable further research into cancer-immune interactions and allow isolation of specific factors that make TDLNs resistant to cancer cell invasion, which are challenging to dissect in vivo.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article