Arginine alleviates Clostridium perfringens α toxin-induced intestinal injury in vivo and in vitro via the SLC38A9/mTORC1 pathway

Front Immunol. 2024 Apr 4:15:1357072. doi: 10.3389/fimmu.2024.1357072. eCollection 2024.

Abstract

Introduction: Clostridium perfringens α toxin is a main virulence factor responsible for gut damage in animals. Arginine is a functional amino acid exhibiting significant immunoregulatory activities. However, the effects and immunoregulatory mechanisms of arginine supplementation on α toxin-induced intestinal injury remain unclear.

Methods: In vivo, 256 male Arbor Acres chickens were randomly assigned to a 2×2 factorial arrangement, involving diet treatments (with or without 0.3% arginine supplementation) and immunological stress (with or without α toxin challenge). In vitro, IEC-6 cells were treated with or without arginine in the presence or absence of α toxin. Moreover, IEC-6 cells were transfected with siRNA targeting mTOR and SLC38A9 to explore the underlying mechanisms.

Results and discussion: The results showed that in vivo, arginine supplementation significantly alleviated the α toxin-induced growth performance impairment, decreases in serum immunoglobulin (Ig)A and IgG levels, and intestinal morphology damage. Arginine supplementation also significantly reduced the α toxin-induced increase in jejunal proinflammatory cytokines interleukin (IL)-1β, IL-6 and IL-17 mRNA expression. Clostridium perfringens α toxin significantly decreased jejunal mechanistic target of rapamycin (mTOR) and solute carrier family 38 member 9 (SLC38A9) mRNA expression, while arginine supplementation significantly increased mTOR and SLC38A9 mRNA expression. In vitro, arginine pretreatment mitigated the α toxin-induced decrease in cell viability and the increase in cytotoxicity and apoptosis. Arginine pretreatment also alleviated the α toxin-induced upregulation of mRNA expression of inflammation-related cytokines IL-6, C-X-C motif chemokine ligand (CXCL)10, CXCL11 and transforming growth factor-β (TGF-β), as well as apoptosis-related genes B-cell lymphoma-2 associated X protein (Bax), B-cell lymphoma-2 (Bcl-2), B-cell lymphoma-extra large (Bcl-XL) and cysteinyl aspartate specific proteinase 3 (Caspase-3) and the ratio of Bax to Bcl-2. Arginine pretreatment significantly increased the α toxin-induced decrease in mTOR, SLC38A9, eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4EBP1) and ribosomal protein S6 kinase (S6K) mRNA expression. Knockdown SLC38A9 and mTOR largely abrogated the positive effects of arginine pretreatment on α toxin-induced intracellular changes. Furthermore, SLC38A9 silencing abolished the increased mTOR mRNA expression caused by arginine pretreatment. In conclusion, arginine administration attenuated α toxin-induced intestinal injury in vivo and in vitro, which could be associated with the downregulation of inflammation via regulating SLC38A9/mTORC1 pathway.

Keywords: Clostridium perfringens α toxin; IEC-6 cells; SLC38A9/mTORC1 pathway; arginine; broiler chickens; cell apoptosis; intestinal inflammation.

MeSH terms

  • Amino Acid Transport Systems / metabolism
  • Animals
  • Arginine* / pharmacology
  • Bacterial Toxins* / toxicity
  • Calcium-Binding Proteins*
  • Chickens / genetics
  • Inflammation
  • Interleukin-6*
  • Male
  • Mechanistic Target of Rapamycin Complex 1
  • RNA, Messenger / genetics
  • TOR Serine-Threonine Kinases / metabolism
  • Type C Phospholipases*
  • bcl-2-Associated X Protein

Substances

  • alpha toxin, Clostridium perfringens
  • Arginine
  • Bacterial Toxins
  • bcl-2-Associated X Protein
  • Calcium-Binding Proteins
  • Interleukin-6
  • Mechanistic Target of Rapamycin Complex 1
  • RNA, Messenger
  • TOR Serine-Threonine Kinases
  • Type C Phospholipases
  • Amino Acid Transport Systems

Grants and funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This project was supported by the National Natural Science Foundation of China (32102580), the Natural Science Foundation of Shandong Province (ZR2020QC181), the Doctoral Science Research Starup Funding of Qingdao Agricultural University (663/1120008), the Open Project of State Key Laboratory of Animal Nutrition and the National College Student Innovation and Entrepreneurship Training Program Project (202210435024).