4.8 Article

SLC15A4 mediates M1-prone metabolic shifts in macrophages and guards immune cells from metabolic stress

Publisher

NATL ACAD SCIENCES
DOI: 10.1073/pnas.2100295118

Keywords

innate immune cell; immunometabolism; amino acid transporter; cytokine; macrophage

Funding

  1. Ministry of Education, Culture, Sports, Science and Technology of Japan [17H04070, 20H05354, 18K07056, 18K16162]
  2. National Center for Global Health and Medicine [23S001]
  3. Japan Science and Technology Agency Exploratory Research for Advanced Technology Suematsu Gas Biology Project (FY2010-FY2015)
  4. Grants-in-Aid for Scientific Research [17H04070, 18K07056, 20H05354, 18K16162] Funding Source: KAKEN

Ask authors/readers for more resources

The amino acid and oligopeptide transporter SLC15A4 plays a critical role in regulating inflammatory responses and cytokine production in immune cells. Loss of SLC15A4 leads to metabolic imbalances, impacting glutamine metabolism and the tricarboxylic acid cycle. Additionally, SLC15A4 is essential for AMPK activation and maintaining respiratory homeostasis in macrophages.
The amino acid and oligopeptide transporter Solute carrier family 15 member A4 (SLC15A4), which resides in lysosomes and is preferentially expressed in immune cells, plays critical roles in the pathogenesis of lupus and colitis in murine models. Toll-like receptor (TLR) 7/9-and nucleotide-binding oligomerization domain-containing protein 1 (NOD1)-mediated inflammatory responses require SLC15A4 function for regulating the mechanistic target of rapamycin complex 1 (mTORC1) or transporting L-Ala-gamma-D-Glu-meso-diaminopimelic acid, IL-12: interleukin-12 (Tri-DAP), respectively. Here, we further investigated the mechanism of how SLC15A4 directs inflammatory responses. Proximity-dependent biotin identification revealed glycolysis as highly enriched gene ontology terms. Fluxome analyses in macrophages indicated that SLC15A4 loss causes insufficient biotransformation of pyruvate to the tricarboxylic acid cycle, while increasing glutaminolysis to the cycle. Furthermore, SLC15A4 was required for M1-prone metabolic change and inflammatory IL-12 cytokine productions after TLR9 stimulation. SLC15A4 could be in close proximity to AMP-activated protein kinase (AMPK) and mTOR, and SLC15A4 deficiency impaired TLR-mediated AMPK activation. Interestingly, SLC15A4-intact but not SLC15A4-deficient macrophages became resistant to fluctuations in environmental nutrient levels by limiting the use of the glutamine source; thus, SLC15A4 was critical for macrophage's respiratory homeostasis. Our findings reveal a mechanism of metabolic regulation in which an amino acid transporter acts as a gatekeeper that protects immune cells' ability to acquire an M1-prone metabolic phenotype in inflammatory tissues by mitigating metabolic stress.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available