4.7 Article

Loss of host tissue transglutaminase boosts antitumor T cell immunity by altering STAT1/STAT3 phosphorylation in ovarian cancer

Journal

JOURNAL FOR IMMUNOTHERAPY OF CANCER
Volume 9, Issue 9, Pages -

Publisher

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2021-002682

Keywords

tumor microenvironment; CD8-positive T-lymphocytes; lymphocyte activation; lymphocytes; tumor-infiltrating; genital neoplasms; female

Funding

  1. US Department of Veterans Affairs [I01 BX000792-06]
  2. Robert H. Comprehensive Cancer Center
  3. Walter S. and Lucienne Driskill Immunotherapy Research fund
  4. UEFISCDI [PN-III-P1-1.1-TE-2019-0670]
  5. Cancer Center Support Grant NCI [CA060553]

Ask authors/readers for more resources

The study showed that host TG2 deficiency delayed tumor growth, increased infiltration of CD8(+) T cells, and decreased myeloid cell numbers in the peritoneal fluid. Loss of TG2 enhanced tumor antigen-specific CD8(+) T cell cytotoxic responses and reduced immune-suppressive signals in the tumor microenvironment, leading to decreased tumor burden.
Background Tissue transglutaminase (TG2), an enzyme overexpressed in cancer cells, promotes metastasis and resistance to chemotherapy. Its distinct effects in cancer versus the host compartments have not been elucidated. Methods Here, by using a TG2(-/-) syngeneic ovarian cancer mouse model, we assessed the effects of TG2 deficiency in the host tissues on antitumor immunity and tumor progression. Multicolor flow cytometry was used to phenotype immune cell populations in the peritoneal environment. Cancer cells recovered from malignant ascites were characterized by RNA sequencing, proliferation, and apoptosis assays. Results We observed that host TG2 loss delayed tumor growth and ascites accumulation and caused increased infiltration of CD8(+) T cells and decreased numbers of myeloid cells in the peritoneal fluid. Tumor antigen-specific CD8(+) T cell cytotoxic responses were enhanced in ascites from TG2(-/-) versus TG2(+/+) mice and CD8(+) T cell depletion caused accelerated ascites accumulation in TG2(-/-) mice. CD8(+) T cells from tumor-bearing TG2(-/-) mice displayed an effector T cell phenotype, differentiated toward effector memory (T-em). Mechanistically, absence of TG2 augmented signals promoting T cell activation, such as increased cytokine-induced STAT1 and attenuated STAT3 phosphorylation in T cells. Additionally, immune-suppressive myeloid cell populations were reduced in the peritoneal milieu of TG2(-/-) tumor-bearing mice. In response to the more robust immune response caused by loss of TG2, cancer cells growing intraperitoneally exhibited an interferon-gamma (IFN-gamma) responsive gene signature and underwent apoptosis. In human specimens, stromal, not tumor, TG2 expression correlated indirectly with numbers of tumor-infiltrating lymphocytes. Conclusions Collectively, our data demonstrate decreased tumor burden, increased activation and effector function of T cells, and loss of immunosuppressive signals in the tumor microenvironment of TG2(-/-) mice. We propose that TG2 acts as an attenuator of antitumor T cell immunity and is a new immunomodulatory target.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available