[HTML][HTML] Lipid droplets fuel SARS-CoV-2 replication and production of inflammatory mediators

SSG Dias, VC Soares, AC Ferreira… - PLoS …, 2020 - journals.plos.org
SSG Dias, VC Soares, AC Ferreira, CQ Sacramento, N Fintelman-Rodrigues, JR Temerozo
PLoS pathogens, 2020journals.plos.org
Viruses are obligate intracellular parasites that make use of the host metabolic machineries
to meet their biosynthetic needs. Thus, identifying the host pathways essential for the virus
replication may lead to potential targets for therapeutic intervention. The mechanisms and
pathways explored by SARS-CoV-2 to support its replication within host cells are not fully
known. Lipid droplets (LD) are organelles with major functions in lipid metabolism, energy
homeostasis and intracellular transport, and have multiple roles in infections and …
Viruses are obligate intracellular parasites that make use of the host metabolic machineries to meet their biosynthetic needs. Thus, identifying the host pathways essential for the virus replication may lead to potential targets for therapeutic intervention. The mechanisms and pathways explored by SARS-CoV-2 to support its replication within host cells are not fully known. Lipid droplets (LD) are organelles with major functions in lipid metabolism, energy homeostasis and intracellular transport, and have multiple roles in infections and inflammation. Here we described that monocytes from COVID-19 patients have an increased LD accumulation compared to SARS-CoV-2 negative donors. In vitro, SARS-CoV-2 infection were seen to modulate pathways of lipid synthesis and uptake as monitored by testing for CD36, SREBP-1, PPARγ, and DGAT-1 expression in monocytes and triggered LD formation in different human cell lines. LDs were found in close apposition with SARS-CoV-2 proteins and double-stranded (ds)-RNA in infected Vero cells. Electron microscopy (EM) analysis of SARS-CoV-2 infected Vero cells show viral particles colocalizing with LDs, suggestive that LDs might serve as an assembly platform. Pharmacological modulation of LD formation by inhibition of DGAT-1 with A922500 significantly inhibited SARS-CoV-2 replication as well as reduced production of mediators pro-inflammatory response. Taken together, we demonstrate the essential role of lipid metabolic reprograming and LD formation in SARS-CoV-2 replication and pathogenesis, opening new opportunities for therapeutic strategies to COVID-19.
PLOS