Herpes Simplex Virus Type 1 Infects Enteric Neurons and Triggers Gut Dysfunction via Macrophage Recruitment

Front Cell Infect Microbiol. 2018 Mar 15:8:74. doi: 10.3389/fcimb.2018.00074. eCollection 2018.

Abstract

Herpes Simplex Virus type 1 (HSV-1), a neurotropic pathogen widespread in human population, infects the enteric nervous system (ENS) in humans and rodents and causes intestinal neuromuscular dysfunction in rats. Although infiltration of inflammatory cells in the myenteric plexus and neurodegeneration of enteric nerves are common features of patients suffering from functional intestinal disorders, the proof of a pathogenic link with HSV-1 is still unsettled mainly because the underlying mechanisms are largely unknown. In this study we demonstrated that following intragastrical administration HSV-1 infects neurons within the myenteric plexus resulting in functional and structural alterations of the ENS. By infecting mice with HSV-1 replication-defective strain we revealed that gastrointestinal neuromuscular anomalies were however independent of viral replication. Indeed, enteric neurons exposed to UV-inactivated HSV-1 produced monocyte chemoattractant protein-1 (MCP-1/CCL2) to recruit activated macrophages in the longitudinal muscle myenteric plexus. Infiltrating macrophages produced reactive oxygen and nitrogen species and directly harmed enteric neurons resulting in gastrointestinal dysmotility. In HSV-1 infected mice intestinal neuromuscular dysfunctions were ameliorated by in vivo administration of (i) liposomes containing dichloromethylene bisphosphonic acid (clodronate) to deplete tissue macrophages, (ii) CCR2 chemokine receptor antagonist RS504393 to block the CCL2/CCR2 pathway, (iii) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME) and AR-C 102222 to quench production of nitrogen reactive species produced via iNOS. Overall these data demonstrate that HSV-1 infection makes enteric neurons recruit macrophages via production of a specific chemoattractant factor. The resulting inflammatory reaction is mandatory for intestinal dysmotility. These findings provide insights into the neuro-immune communication that occurs in the ENS following HSV-1 infection and allow recognition of an original pathophysiologic mechanism underlying gastrointestinal diseases as well as identification of novel therapeutic targets.

Keywords: enteric neuropathies; inflammation; macrophage recruitment; neuromuscular dysfunction; neurotropic virus.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adaptive Immunity
  • Animals
  • Arginine / analogs & derivatives
  • Arginine / metabolism
  • Chemokine CCL2 / metabolism
  • Clodronic Acid
  • Disease Models, Animal
  • Enteric Nervous System / drug effects*
  • Enteric Nervous System / metabolism
  • Enteric Nervous System / pathology
  • Enteric Nervous System / virology*
  • Gastrointestinal Motility / drug effects*
  • Herpes Simplex / immunology
  • Herpes Simplex / metabolism*
  • Herpes Simplex / pathology
  • Herpes Simplex / virology
  • Herpesvirus 1, Human / pathogenicity*
  • Ileum / immunology
  • Ileum / pathology
  • Ileum / virology
  • Inflammation / metabolism
  • Liposomes / metabolism
  • Macrophages / metabolism*
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Myenteric Plexus / drug effects
  • Myenteric Plexus / metabolism
  • Myenteric Plexus / pathology
  • Myenteric Plexus / virology
  • NG-Nitroarginine Methyl Ester / metabolism
  • Neurons / drug effects*
  • Neurons / virology
  • Rats
  • Reactive Nitrogen Species / metabolism
  • Reactive Nitrogen Species / toxicity
  • Reactive Oxygen Species / metabolism
  • Reactive Oxygen Species / toxicity
  • Receptors, Chemokine
  • Virus Internalization
  • Virus Replication

Substances

  • Chemokine CCL2
  • Liposomes
  • Reactive Nitrogen Species
  • Reactive Oxygen Species
  • Receptors, Chemokine
  • Clodronic Acid
  • arginine methyl ester
  • Arginine
  • NG-Nitroarginine Methyl Ester