PT - JOURNAL ARTICLE AU - Patrick J. Ferrara AU - Xin Rong AU - J. Alan Maschek AU - Anthony R.P. Verkerke AU - Piyarat Siripoksup AU - Haowei Song AU - Karthickeyan C. Krishnan AU - Jordan M. Johnson AU - John Turk AU - Joseph A. Houmard AU - Aldons J. Lusis AU - James E. Cox AU - Saame Raza Shaikh AU - Peter Tontonoz AU - Katsuhiko Funai TI - The Lands cycle modulates plasma membrane lipid organization and insulin sensitivity in skeletal muscle AID - 10.1101/2019.12.23.887232 DP - 2019 Jan 01 TA - bioRxiv PG - 2019.12.23.887232 4099 - http://biorxiv.org/content/early/2019/12/23/2019.12.23.887232.short 4100 - http://biorxiv.org/content/early/2019/12/23/2019.12.23.887232.full AB - Aberrant lipid metabolism promotes the development of skeletal muscle insulin resistance, but the exact identity of lipid-mediated mechanisms relevant to human obesity remains unclear. A comprehensive lipidomic analyses of primary myocytes from lean insulin-sensitive (LN) and obese insulin-resistant (OB) individuals revealed several species of lysophospholipids (lyso-PL) that were differentially-abundant. These changes coincided with greater expression of lysophosphatidylcholine acyltransferase 3 (LPCAT3), an enzyme involved in phospholipid transacylation (Lands cycle). Strikingly, mice with skeletal muscle-specific knockout of LPCAT3 (LPCAT3-MKO) exhibited greater muscle lyso-PC/PC, concomitant with greater insulin sensitivity in vivo and insulin-stimulated skeletal muscle glucose uptake ex vivo. Absence of LPCAT3 reduced phospholipid packing of the cellular membranes and increased plasma membrane lipid clustering, suggesting that LPCAT3 affects insulin receptor phosphorylation by modulating plasma membrane lipid organization. In conclusion, obesity accelerates the skeletal muscle Lands cycle, whose consequence might induce the disruption of plasma membrane organization that suppresses muscle insulin action.