Ammonia transporter RhBG initiates downstream signaling and functional responses by activating NFκB

Transceptors, solute transporters that facilitate intracellular entry of molecules and also initiate intracellular signaling events, have been primarily studied in lower-order species. Ammonia, a cytotoxic endogenous metabolite, is converted to urea in hepatocytes for urinary excretion in mammals. D...

Full description

Saved in:
Bibliographic Details
Published inProceedings of the National Academy of Sciences - PNAS Vol. 121; no. 31; p. e2314760121
Main Authors Mishra, Saurabh, Welch, Nicole, Singh, Shashi Shekhar, Singh, Khuraijam Dhanachandra, Bellar, Annette, Kumar, Avinash, Deutz, Lars N, Hanlon, Maxmillian D, Kant, Sashi, Dastidar, Sumitava, Patel, Hailee, Agrawal, Vandana, Attaway, Amy H, Musich, Ryan, Stark, George R, Tedesco, Francesco Saverio, Truskey, George A, Weiner, I David, Karnik, Sadashiva S, Dasarathy, Srinivasan
Format Journal Article
LanguageEnglish
Published United States National Academy of Sciences 30.07.2024
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Transceptors, solute transporters that facilitate intracellular entry of molecules and also initiate intracellular signaling events, have been primarily studied in lower-order species. Ammonia, a cytotoxic endogenous metabolite, is converted to urea in hepatocytes for urinary excretion in mammals. During hyperammonemia, when hepatic metabolism is impaired, nonureagenic ammonia disposal occurs primarily in skeletal muscle. Increased ammonia uptake in skeletal muscle is mediated by a membrane-bound, 12 transmembrane domain solute transporter, Rhesus blood group-associated B glycoprotein (RhBG). We show that in addition to its transport function, RhBG interacts with myeloid differentiation primary response-88 (MyD88) to initiate an intracellular signaling cascade that culminates in activation of NFκB. We also show that ammonia-induced MyD88 signaling is independent of the canonical toll-like receptor-initiated mechanism of MyD88-dependent NFκB activation. In silico, in vitro, and in situ experiments show that the conserved cytosolic J-domain of the RhBG protein interacts with the Toll-interleukin-1 receptor (TIR) domain of MyD88. In skeletal muscle from human patients, human-induced pluripotent stem cell-derived myotubes, and myobundles show an interaction of RhBG-MyD88 during hyperammonemia. Using complementary experimental and multiomics analyses in murine myotubes and mice with muscle-specific RhBG or MyD88 deletion, we show that the RhBG-MyD88 interaction is essential for the activation of NFkB but not ammonia transport. Our studies show a paradigm of substrate-dependent regulation of transceptor function with the potential for modulation of cellular responses in mammalian systems by decoupling transport and signaling functions of transceptors.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:0027-8424
1091-6490
1091-6490
DOI:10.1073/pnas.2314760121