4.8 Article

Activation of the GPR35 pathway drives angiogenesis in the tumour microenvironment

Journal

GUT
Volume 71, Issue 3, Pages 509-+

Publisher

BMJ PUBLISHING GROUP
DOI: 10.1136/gutjnl-2020-323363

Keywords

-

Funding

  1. Wellcome Trust [NK 216630/Z/19/Z, NK 103077/Z/13/Z, 106260/Z/14/Z, UNS59491]
  2. European Research Council under the European Community [648 889, 609 020]
  3. Addenbrooke's Charitable Trust [ACT 25/16A]
  4. UniNA
  5. Compagnia Di San Paolo 'STAR programme for young researchers' fellowship
  6. NIHR Cambridge BRC
  7. Wellcome Trust [103077/Z/13/Z, 106260/Z/14/Z, 216630/Z/19/Z] Funding Source: Wellcome Trust

Ask authors/readers for more resources

The study demonstrates that GPR35 on macrophages can enhance tumor growth by stimulating neoangiogenesis and tumor tissue remodeling. Deletion of GPR35 significantly reduces tumor growth caused by the mutant tumor suppressor adenomatous polyposis coli in inflammation-associated and spontaneous tumor models.
Objective Primary sclerosing cholangitis (PSC) is in 70% of cases associated with inflammatory bowel disease. The hypermorphic T108M variant of the orphan G protein-coupled receptor GPR35 increases risk for PSC and ulcerative colitis (UC), conditions strongly predisposing for inflammation-associated liver and colon cancer. Lack of GPR35 reduces tumour numbers in mouse models of spontaneous and colitis associated cancer. The tumour microenvironment substantially determines tumour growth, and tumour-associated macrophages are crucial for neovascularisation. We aim to understand the role of the GPR35 pathway in the tumour microenvironment of spontaneous and colitis-associated colon cancers. Design Mice lacking GPR35 on their macrophages underwent models of spontaneous colon cancer or colitis-associated cancer. The role of tumour-associated macrophages was then assessed in biochemical and functional assays. Results Here, we show that GPR35 on macrophages is a potent amplifier of tumour growth by stimulating neoangiogenesis and tumour tissue remodelling. Deletion of Gpr35 in macrophages profoundly reduces tumour growth in inflammation-associated and spontaneous tumour models caused by mutant tumour suppressor adenomatous polyposis coli. Neoangiogenesis and matrix metalloproteinase activity is promoted by GPR35 via Na/K-ATPase-dependent ion pumping and Src activation, and is selectively inhibited by a GPR35-specific pepducin. Supernatants from human inducible-pluripotent-stem-cell derived macrophages carrying the UC and PSC risk variant stimulate tube formation by enhancing the release of angiogenic factors. Conclusions Activation of the GPR35 pathway promotes tumour growth via two separate routes, by directly augmenting proliferation in epithelial cells that express the receptor, and by coordinating macrophages' ability to create a tumour-permissive environment.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available