Determination of the autophagic flux in murine and human peripheral blood mononuclear cells

Front Cell Dev Biol. 2023 Mar 13:11:1122998. doi: 10.3389/fcell.2023.1122998. eCollection 2023.

Abstract

The autophagy lysosomal system (ALS) is crucial for cellular homeostasis, contributing to maintain whole body health and alterations are associated with diseases like cancer or cardiovascular diseases. For determining the autophagic flux, inhibition of lysosomal degradation is mandatory, highly complicating autophagy measurement in vivo. To overcome this, herein blood cells were used as they are easy and routinely to isolate. Within this study we provide detailed protocols for determination of the autophagic flux in peripheral blood mononuclear cells (PBMCs) isolated from human and, to our knowledge the first time, also from murine whole blood, extensively discussing advantages and disadvantages of both methods. Isolation of PBMCs was performed using density gradient centrifugation. To minimize changes on the autophagic flux through experimental conditions, cells were directly treated with concanamycin A (ConA) for 2 h at 37°C in their serum or for murine cells in serum filled up with NaCl. ConA treatment decreased lysosomal cathepsins activity and increased Sequestosome 1 (SQSTM1) protein and LC3A/B-II:LC3A/B-I ratio in murine PBMCs, while transcription factor EB was not altered yet. Aging further enhanced ConA-associated increase in SQSTM1 protein in murine PBMCs but not in cardiomyocytes, indicating tissue-specific differences in autophagic flux. In human PBMCs, ConA treatment also decreased lysosomal activity and increased LC3A/B-II protein levels, demonstrating successful autophagic flux detection in human subjects. In summary, both protocols are suitable to determine the autophagic flux in murine and human samples and may facilitate a better mechanistic understanding of altered autophagy in aging and disease models and to further develop novel treatment strategies.

Keywords: LC3; NZO; TFEB; aging; cardiomyocytes; concanamycin A; lysosomes; sequestome-1.

Grants and funding

CO is funded by the DZHK (German Center for Cardiovascular Research; 81Z2100502). SW is funded by DFG-funded research group TraceAge (FOR 2558). The human study is funded by the German Science Foundation (DFG RA 3340/3-1, OP-R). Publication costs are funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) - 491394008.