Flavin monooxygenase 3, the host hepatic enzyme in the metaorganismal trimethylamine N-oxide-generating pathway, modulates platelet responsiveness and thrombosis risk

J Thromb Haemost. 2018 Sep;16(9):1857-1872. doi: 10.1111/jth.14234. Epub 2018 Aug 9.

Abstract

Essentials Microbe-dependent production of trimethylamine N-oxide (TMAO) contributes to thrombosis risk. The impact of host flavin monooxygenase 3 (FMO3) modulation on platelet function is unknown. Genetic manipulation of FMO3 in mice alters systemic TMAO levels and thrombosis potential. Genetic manipulation of FMO3 is associated with alteration of gut microbial community structure.

Summary: Background Gut microbes play a critical role in the production of trimethylamine N-oxide (TMAO), an atherogenic metabolite that impacts platelet responsiveness and thrombosis potential. Involving both microbe and host enzymatic machinery, TMAO generation utilizes a metaorganismal pathway, beginning with ingestion of trimethylamine (TMA)-containing dietary nutrients such as choline, phosphatidylcholine and carnitine, which are abundant in a Western diet. Gut microbial TMA lyases use these nutrients as substrates to produce TMA, which upon delivery to the liver via the portal circulation, is converted into TMAO by host hepatic flavin monooxygenases (FMOs). Gut microbial production of TMA is rate limiting in the metaorganismal TMAO pathway because hepatic FMO activity is typically in excess. Objectives FMO3 is the major FMO responsible for host generation of TMAO; however, a role for FMO3 in altering platelet responsiveness and thrombosis potential in vivo has not yet been explored. Methods The impact of FMO3 suppression (antisense oligonucleotide-targeting) and overexpression (as transgene) on plasma TMAO levels, platelet responsiveness and thrombosis potential was examined using a murine FeCl3 -induced carotid artery injury model. Cecal microbial composition was examined using 16S analyses. Results Modulation of FMO3 directly impacts systemic TMAO levels, platelet responsiveness and rate of thrombus formation in vivo. Microbial composition analyses reveal taxa whose proportions are associated with both plasma TMAO levels and in vivo thrombosis potential. Conclusions The present studies demonstrate that host hepatic FMO3, the terminal step in the metaorganismal TMAO pathway, participates in diet-dependent and gut microbiota-dependent changes in both platelet responsiveness and thrombosis potential in vivo.

Keywords: blood platelets; cardiovascular disease; choline; gut microbe; thrombosis.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Blood Platelets / physiology*
  • Carotid Artery Thrombosis / blood
  • Carotid Artery Thrombosis / chemically induced
  • Carotid Artery, Common
  • Chlorides / toxicity
  • Ferric Compounds / toxicity
  • Gastrointestinal Microbiome / physiology*
  • Gene Knockdown Techniques
  • Humans
  • Liver / enzymology*
  • Methylamines / metabolism*
  • Mice
  • Mice, Inbred C57BL
  • Oligonucleotides, Antisense / pharmacology
  • Oxygenases / antagonists & inhibitors
  • Oxygenases / genetics
  • Oxygenases / physiology*
  • Platelet-Rich Plasma
  • Ribotyping
  • Risk
  • Thrombophilia / enzymology*
  • Thrombophilia / microbiology
  • Transgenes

Substances

  • Chlorides
  • Ferric Compounds
  • Methylamines
  • Oligonucleotides, Antisense
  • Oxygenases
  • dimethylaniline monooxygenase (N-oxide forming)
  • trimethyloxamine
  • ferric chloride