Your browser doesn't support javascript.
loading
Cyclic-di-GMP induces inflammation and acute lung injury through direct binding to MD2.
Qian, Chenchen; Zhu, Weiwei; Wang, Jiong; Wang, Zhe; Tang, Weiyang; Liu, Xin; Jin, Bo; Xu, Yong; Zhang, Yuyang; Liang, Guang; Wang, Yi.
Affiliation
  • Qian C; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China.
  • Zhu W; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Wang J; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Wang Z; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Tang W; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China.
  • Liu X; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China.
  • Jin B; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Xu Y; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
  • Zhang Y; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China.
  • Liang G; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China.
  • Wang Y; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
Clin Transl Med ; 14(8): e1744, 2024 Aug.
Article in En | MEDLINE | ID: mdl-39166890
ABSTRACT

BACKGROUND:

Severe bacterial infections can trigger acute lung injury (ALI) and acute respiratory distress syndrome, with bacterial pathogen-associated molecular patterns (PAMPs) exacerbating the inflammatory response, particularly in COVID-19 patients. Cyclic-di-GMP (CDG), one of the PAMPs, is synthesized by various Gram-positve and Gram-negative bacteria. Previous studies mainly focused on the inflammatory responses triggered by intracellular bacteria-released CDG. However, how extracellular CDG, which is released by bacterial autolysis or rupture, activates the inflammatory response remains unclear.

METHODS:

The interaction between extracellular CDG and myeloid differentiation protein 2 (MD2) was investigated using in vivo and in vitro models. MD2 blockade was achieved using specific inhibitor and genetic knockout mice. Site-directed mutagenesis, co-immunoprecipitation, SPR and Bis-ANS displacement assays were used to identify the potential binding sites of MD2 on CDG.

RESULTS:

Our data show that extracellular CDG directly interacts with MD2, leading to activation of the TLR4 signalling pathway and lung injury. Specific inhibitors or genetic knockout of MD2 in mice significantly alleviated CDG-induced lung injury. Moreover, isoleucine residues at positions 80 and 94, along with phenylalanine at position 121, are essential for the binding of MD2 to CDG.

CONCLUSION:

These results reveal that extracellular CDG induces lung injury through direct interaction with MD2 and activation of the TLR4 signalling pathway, providing valuable insights into bacteria-induced ALI mechanisms and new therapeutic approaches for the treatment of bacterial co-infection in COVID-19 patients.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cyclic GMP / Lymphocyte Antigen 96 / Acute Lung Injury / COVID-19 Limits: Animals / Humans / Male Language: En Journal: Clin Transl Med Year: 2024 Document type: Article Affiliation country: China

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cyclic GMP / Lymphocyte Antigen 96 / Acute Lung Injury / COVID-19 Limits: Animals / Humans / Male Language: En Journal: Clin Transl Med Year: 2024 Document type: Article Affiliation country: China