ILANIT 2020

A TRAIL-TL1A paracrine network involving adipocytes, macrophages and lymphocytes induces adipose tissue dysfunction downstream of E2F1 in human obesity

Nitzan Maixner 1 Tal Pecht 1 Yulia Haim 1 Nir Goldstein 1 Tania Tarnovscki 1 Idit F. Liberty 4 Boris Kirshtein 5 Vered Chalifa-Caspi 6 Rachel Golan 2 Nava Bashan 1 Mathias Bluher 3 Assaf Rudich 1,6
1Department of Clinical Biochemistry, Ben Gurion University, Israel
2Department of Epidemiology and Preventive Medicine, Ben Gurion University, Israel
3Department of Medicine, University of Leipzig, Germany
4Diabetes research and treatment center, Soroka University medical Center, Israel
5Surgical Department A, Soroka University Medical Center, Israel
6The National Institute of Biotechnology in the Negev, Ben Gurion University, Israel

Background: In human visceral adipose-tissue, elevated levels of the transcription factor E2F1 in adipocytes were recently associated with cardio-metabolically-at-risk obesity, demonstrated in both in vitro and clinical models (Autophagy, 11:2074,2015). However, given the ubiquitous nature of E2F1 the specific pathways mediating this effect are of particular interest, and these were not fully depicted yet.

Hypothesis: We hypothesized that the E2F1-driven dysfunctional profile is mediated by a complex network of downstream effectors.

Methods: An unbiased approach utilizing RNA-sequencing was used to assess the transcriptome of omental adipose-tissue samples from age-, sex- and BMI–matched patients, whose omental E2F1 protein expression was either high (E2F1high) or low (E2F1low). Significant pathways were identified using STRING© analysis. This was completed using in-vitro molecular approaches challenging the functional significance of identified pathways.

Results: Paired analysis revealed over 1300 differentially expressed genes consisting of several functionally significant pathways. Of those, stood out a network of TNF-superfamily factors, especially TRAIL(TNFSF10), TL1A(TNFSF15) and their receptors, later validated in a second cohort. Further ex-vivo experiments unraveled a feed-forward loop in which TL1A overexpression was the product of TRAIL-exposed adipose tissue components, specifically T-lymphocytes. In-vitro, TL1A induced insulin resistance and secretory malfunction in human adipocytes, while facilitating a lipid-accumulating profile in human macrophages, generating foam-cells which further aggravated tissue dysfunction. These phenomenon were also reflected in human cohorts, correlating TL1A with clinical parameters of cardio-metabolic risk determinants.

Conclusions: We propose an intra-adipose tissue paracrine loop, consisting of E2F1-driven TNF-SF activation, ultimately impinging on adipose tissue function and contributing to the generation of a systemic dys-metabolic phenotype.









Powered by Eventact EMS