Abstract

Despite their emerging relevance to fully understand disease pathogenesis, we have as yet a poor understanding as to how biomechanical signals are integrated with specific biochemical pathways to determine cell behaviour. Mesothelial-to-mesenchymal transition (MMT) markers colocalized with TGF-β1-dependent signaling and yes-associated protein (YAP) activation across biopsies from different pathologies exhibiting peritoneal fibrosis, supporting mechanotransduction as a central driving component of these class of fibrotic lesions and its crosstalk with specific signaling pathways. Transcriptome and proteome profiling of the response of mesothelial cells (MCs) to linear cyclic stretch revealed molecular changes compatible with bona fide MMT, which (i) overlapped with established YAP target gene subsets, and were largely dependent on endogenous TGF-β1 signaling. Importantly, TGF-β1 blockade blunts the transcriptional upregulation of these gene signatures, but not the mechanical activation and nuclear translocation of YAP per se. We studied the role therein of caveolin-1 (CAV1), a plasma membrane mechanotransducer. Exposure of CAV1-deficient MCs to cyclic stretch led to a robust upregulation of MMT-related gene programs, which was blunted upon TGF-β1 inhibition. Conversely, CAV1 depletion enhanced both TGF-β1 and TGFBRI expression, whereas its re-expression blunted mechanical stretching-induced MMT. CAV1 genetic deficiency exacerbated MMT and adhesion formation in an experimental murine model of peritoneal ischaemic buttons. Taken together, these results support that CAV1-YAP/TAZ fine-tune the fibrotic response through the modulation of MMT, onto which TGF-β1-dependent signaling coordinately converges. Our findings reveal a cooperation between biomechanical and biochemical signals in the triggering of MMT, representing a novel potential opportunity to intervene mechanically induced disorders coursing with peritoneal fibrosis, such as post-surgical adhesions.

Highlights

  • Biomechanical forces are increasingly recognized as major determinants of cell phenotype, and emerging drivers of numerous physiopathological processes

  • Biopsies from (i) parietal peritoneum of patients undergoing PD46, peritoneal adhesions (PAs) development after abdominal surgery, or (iii) ovarian carcinoma peritoneal metastasis, were analyzed for the expression of markers related to mechanotransduction pathways (Hippo-yesassociated protein (YAP)/TAZ and CAV1), as well as to TGF-β1 pathway as compared to healthy peritoneum samples

  • These results suggest the existence of an inter-relation between biochemical (TGF-β1-induced) and biomechanical (YAP/TAZ and CAV1-mediated) signals during the pathogenesis of peritoneal fibrotic pathologies

Read more

Summary

Introduction

Biomechanical forces are increasingly recognized as major determinants of cell phenotype, and emerging drivers of numerous physiopathological processes. Biomechanical forces can be transmitted in cells through the interaction of different adhesion receptors such as integrins, cadherins, syndecans, CD44, and dystroglycan, with other cellular receptors or extracellular matrix (ECM) proteins[1,2,3] as well as through changes in plasma membrane (PM) tension[4], that can in turn be captured through specialized structures and membrane domains, including mechanosensitive ion channels and caveolae[1,5,6]. Strippoli et al Cell Death and Disease (2020)11:647 are 60–80 nm diameter flask-shaped PM invaginations involved in numerous cellular processes including viral entry, vesicle transport, lipid metabolism, cell signaling, mechanosensing and mechanotransduction[7]. Caveolae-dependent mechanosensing orchestrates cell and tissue biomechanical adaptation, including the biochemical and mechanical remodelling of ECM11,12

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call