Diet-Induced High Serum Levels of Trimethylamine-N-oxide Enhance the Cellular Inflammatory Response without Exacerbating Acute Intracerebral Hemorrhage Injury in Mice

Oxid Med Cell Longev. 2022 Feb 16:2022:1599747. doi: 10.1155/2022/1599747. eCollection 2022.

Abstract

Trimethylamine-N-oxide (TMAO), an intestinal flora metabolite of choline, may aggravate atherosclerosis by inducing a chronic inflammatory response and thereby promoting the occurrence of cerebrovascular diseases. Knowledge about the influence of TMAO-related inflammatory response on the pathological process of acute stroke is limited. This study was designed to explore the effects of TMAO on neuroinflammation, brain injury severity, and long-term neurologic function in mice with acute intracerebral hemorrhage (ICH). We fed mice with either a regular chow diet or a chow diet supplemented with 1.2% choline pre- and post-ICH. In this study, we measured serum levels of TMAO with ultrahigh-performance liquid chromatography-tandem mass spectrometry at 24 h and 72 h post-ICH. The expression level of P38-mitogen-protein kinase (P38-MAPK), myeloid differentiation factor 88 (MyD88), high-mobility group box1 protein (HMGB1), and interleukin-1β (IL-1β) around hematoma was examined by western blotting at 24 h. Microglial and astrocyte activation and neutrophil infiltration were examined at 72 h. The lesion was examined on days 3 and 28. Neurologic deficits were examined for 28 days. A long-term choline diet significantly increased serum levels of TMAO compared with a regular diet at 24 h and 72 h after sham operation or ICH. Choline diet-induced high serum levels of TMAO did not enhance the expression of P38-MAPK, MyD88, HMGB1, or IL-1β at 24 h. However, it did increase the number of activated microglia and astrocytes around the hematoma at 72 h. Contrary to our expectations, it did not aggravate acute or long-term histologic damage or neurologic deficits after ICH. In summary, choline diet-induced high serum levels of TMAO increased the cellular inflammatory response probably by activating microglia and astrocytes. However, it did not aggravate brain injury or worsen long-term neurologic deficits. Although TMAO might be a potential risk factor for cerebrovascular diseases, this exploratory study did not support that TMAO is a promising target for ICH therapy.

MeSH terms

  • Acute Disease
  • Animals
  • Astrocytes / metabolism*
  • Brain Injuries / blood*
  • Brain Injuries / complications*
  • Brain Injuries / microbiology
  • Cerebral Hemorrhage / blood*
  • Cerebral Hemorrhage / complications*
  • Cerebral Hemorrhage / microbiology
  • Choline / adverse effects*
  • Diet / adverse effects*
  • Disease Models, Animal
  • Gastrointestinal Microbiome
  • Inflammation / blood
  • Inflammation / chemically induced
  • Interleukin-1beta / metabolism
  • Male
  • Methylamines / blood*
  • Mice
  • Mice, Inbred C57BL
  • Microglia / metabolism*
  • Neutrophil Infiltration / drug effects
  • Neutrophils / immunology
  • Signal Transduction / drug effects*
  • p38 Mitogen-Activated Protein Kinases / metabolism

Substances

  • IL1B protein, mouse
  • Interleukin-1beta
  • Methylamines
  • p38 Mitogen-Activated Protein Kinases
  • trimethyloxamine
  • Choline