PARP2 downregulation in T cells ameliorates lipopolysaccharide-induced inflammation of the large intestine

Front Immunol. 2023 Jun 30:14:1135410. doi: 10.3389/fimmu.2023.1135410. eCollection 2023.

Abstract

Introduction: T cell-dependent inflammatory response with the upregulation of helper 17 T cells (Th17) and the downregulation of regulatory T cells (Treg) accompanied by the increased production of tumor necrosis alpha (TNFa) is characteristic of inflammatory bowel diseases (IBD). Modulation of T cell response may alleviate the inflammation thus reduce intestinal damage. Poly(ADP-ribose) polymerase-2 (PARP2) plays role in the development, differentiation and reactivity of T cell subpopulations. Our aim was to investigate the potential beneficial effect of T cell-specific PARP2 downregulation in the lipopolysaccharide (LPS) induced inflammatory response of the cecum and the colon.

Methods: Low-dose LPS was injected intraperitoneally to induce local inflammatory response, characterized by increased TNFa production, in control (CD4Cre; PARP2+/+) and T cell-specific conditional PARP2 knockout (CD4Cre; PARP2f/f) mice. TNFa, IL-1b, IL-17 levels were measured by ELISA, oxidative-nitrative stress was estimated by immunohistochemistry, while PARP1 activity, p38 MAPK and ERK phosphorylation, and NF-kB expression in large intestine tissue samples were examined by Western-blot. Systemic & local T cell subpopulation; Th17 and Treg alterations were also investigated using flowcytometry and immunohistochemistry.

Results: In control animals, LPS induced intestinal inflammation with increased TNFa production, while no significant elevation of TNFa production was observed in T cell-specific PARP2 knockout animals. The absence of LPS-induced elevation in TNFa levels was accompanied by the absence of IL-1b elevation and the suppression of IL-17 production, showing markedly reduced inflammatory response. The increase in oxidative-nitrative stress and PARP1-activation was also absent in these tissues together with altered ERK and NF-kB activation. An increase in the number of the anti-inflammatory Treg cells in the intestinal mucosa was observed in these animals, together with the reduction of Treg count in the peripheral circulation.

Discussion: Our results confirmed that T cell-specific PARP2 downregulation ameliorated LPS-induced colitis. The dampened TNFa production, decreased IL-17 production and the increased intestinal regulatory T cell number after LPS treatment may be also beneficial during inflammatory processes seen in IBD. By reducing oxidative-nitrative stress and PARP1 activation, T cell-specific PARP2 downregulation may also alleviate intestinal tissue damage.

Keywords: IBD - inflammatory bowel disease; PARP (poly(ADP-ribose) polymerase; PARP2; T lymphocyte; colitis; regulatory (Treg) cell.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Colon / pathology
  • Down-Regulation
  • Inflammation / chemically induced
  • Inflammation / pathology
  • Inflammatory Bowel Diseases* / pathology
  • Interleukin-17 / metabolism
  • Lipopolysaccharides* / toxicity
  • Mice
  • NF-kappa B / metabolism
  • Poly(ADP-ribose) Polymerases / metabolism
  • T-Lymphocytes, Regulatory / metabolism

Substances

  • Lipopolysaccharides
  • Interleukin-17
  • NF-kappa B
  • Poly(ADP-ribose) Polymerases

Grants and funding

This work was supported by the Hungarian National Research, Development and Innovation Office NKFIH-FK129206 (EH), NKFIH-FK128376 (RC-K), TKP2021-EGA-24 (RC-K) and Semmelweis University (EH:STIA-18).