Annals of the Rheumatic Diseases | 2021

OP0312\u2005METABOLIC REPROGRAMMING IN MEMORY CD4+ T CELLS IS ASSOCIATED WITH REACTIVE OXYGEN INDUCED IMMUNE CELL DYSFUNCTION DURING AGING

 
 
 
 
 
 
 
 
 
 
 

Abstract


Inflamm-aging is a chronic, sterile, low-grade inflammatory status, characterized by an increase of proinflammatory cytokines which participate in the development of most age-related diseases such as cancer, Alzheimer’s disease, type 2 diabetes mellitus, stroke, cardiovascular diseases, and rheumatoid arthritis (RA). As cellular metabolism modulates T cell function, it can be assumed that metabolic changes accompany the differentiation of memory CD4+ T cells into senescent CD4+ T cell and contribute to memory CD4+ T cells dysfunction during aging.Therefore, we hypothesized that metabolic reprograming in memory CD4+ T cells might represent an essential factor promoting immune cell dysfunction during aging, thereby fuelling to the pathogenesis of age-related diseases including RATo this end, we analysed memory CD4+ T cells isolated from PBMCs of young donors (20-32 years) and old donors (52-67 years) by using MACSTM technology. Ex vivo memory CD4+ T cells were analysed by SeahorseTM Technology to determine proton efflux rate (PER) as a measure of glycolysis (glycPER) and oxygen consumption rate (OCR) as a measure of mitochondrial respiration (mitoOCR). Cytokine expression and secretion was measured by flow cytometry and multiplex assay with and without Mitotempo an inhibitor of reactive oxygen species (ROS). Finally, TCR-stimulated memory CD4+ T cell proliferation was determined using CSFE and Ki-67 after 3 days and 4 days by flow cytometry. ROS and mitochondrial activity were analysed after 24 h using DCF-DA and CellROX Deep Red and Mitotracker by flow cytometry.In a quiescent state, memory CD4+ T cells from elderly individuals demonstrated a decrease in basal glycolysis and compensatory glycolysis, and an increase in the ratio of basal mitochondrial oxygen consumption rate (mitoOCR) to glycolytic proton efflux rate (glycoPER) while their mitochondrial profile was equivalent to that of young donors while the amount of mitochondria was higher with no increase in steady-state ATP level. In this line and in comparison to the younger reference group, memory CD4+ T cells from aged donors presented a greater spare respiratory capacity after TCR-activation and a marked increase in intracellular ROS production. Interestingly, we did not observe an impact of aging on memory CD4+ T cell proliferation as determined by CFSE and Ki-67. Although the capacity of intracellular cytokine expression did not differ between the compared groups, the levels of secreted IFN-γ, IP-10, IL-6, IL-9, and MCAF were significantly higher in the supernatants of memory CD4+ T cells taken from aged donors but were sensitive to ROS inhibition. .These findings suggest that metabolic reprogramming in human memory CD4+ T cells during aging results in an increased expression of proinflammatory cytokines as a result of ROS production and mitochondrial dysfunction. This process may culminate in T cell dysfunction and thus contribute to the pathogenesis of inflamm-aging and the development of age-related diseases such as rheumatoid arthritis (RA).None declared.

Volume 80
Pages None
DOI 10.1136/ANNRHEUMDIS-2021-EULAR.3937
Language English
Journal Annals of the Rheumatic Diseases

Full Text