1. Academic Validation
  2. A streptococcal Fic domain-containing protein disrupts blood-brain barrier integrity by activating moesin in endothelial cells

A streptococcal Fic domain-containing protein disrupts blood-brain barrier integrity by activating moesin in endothelial cells

  • PLoS Pathog. 2019 May 9;15(5):e1007737. doi: 10.1371/journal.ppat.1007737.
Zhe Ma 1 2 3 4 5 Jie Peng 1 Dandan Yu 1 Joseph S Park 2 3 6 Huixing Lin 1 4 5 Bin Xu 1 Chengping Lu 1 Hongjie Fan 1 4 5 Matthew K Waldor 2 3 6
Affiliations

Affiliations

  • 1 MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China.
  • 2 Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America.
  • 3 Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America.
  • 4 Ministry of Agriculture Key Laboratory of Animal Bacteriology, Nanjing, Jiangsu, China.
  • 5 Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China.
  • 6 Howard Hughes Medical Institute, Boston, Massachusetts, United States of America.
Abstract

Streptococcus equi subsp. zooepidemicus (SEZ) is a zoonotic pathogen capable of causing meningitis in humans. The mechanisms that enable pathogens to traverse the blood-brain barrier (BBB) are incompletely understood. Here, we investigated the role of a newly identified Fic domain-containing protein, BifA, in SEZ virulence. BifA was required for SEZ to cross the BBB and to cause meningitis in mice. BifA also enhanced SEZ translocation across human Brain Microvascular Endothelial Cell (hBMEC) monolayers. Purified BifA or its Fic domain-containing C-terminus alone were able to enter into hBMECs, leading to disruption of monolayer barrier integrity. A SILAC-based proteomic screen revealed that BifA binds moesin. BifA's Fic domain was required for its binding to this regulator of host cell cytoskeletal processes. BifA treatment of hBMECs led to moesin phosphorylation and downstream RhoA activation. Inhibition of moesin activation or moesin depletion in hBMEC monolayers abrogated BifA-mediated increases in barrier permeability and SEZ's capacity to translocate across monolayers. Thus, BifA activation of moesin appears to constitute a key mechanism by which SEZ disrupts endothelial monolayer integrity to penetrate the BBB.

Figures
Products