Abstract

BackgroundThe blood–spinal cord barrier (BSCB) is composed of a monolayer of endothelium linked with tight junctions and extracellular matrix (ECM)-rich basement membranes and is surrounded by astrocyte foot processes. Endothelial permeability is regulated by interaction between endothelial cells and ECM proteins. Fibronectin (FN) is a principal ECM component of microvessels. Excessive FN deposition disrupts cell–cell adhesion in fibroblasts through β1 integrin ligation. To determine whether excessive FN deposition contributes to the disruption of endothelial integrity, we used an in vitro model of the endothelial monolayer to investigate whether the FN inhibitor pUR4 prevents FN deposition into the subendothelial matrix and attenuates endothelial leakage.MethodsTo correlate the effects of excessive FN accumulation in microvessels on BSCB disruption, spinal nerve ligation—which induces BSCB leakage—was applied, and FN expression in the spinal cord was evaluated through immunohistochemistry and immunoblotting. To elucidate the effects by which pUR4 modulates endothelial permeability, brain-derived endothelial (bEND.3) cells treated with tumor necrosis factor (TNF)-α were used to mimic a leaky BSCB. A bEND.3 monolayer was preincubated with pUR4 before TNF-α treatment. The transendothelial electrical resistance (TEER) measurement and transendothelial permeability assay were applied to assess the endothelial integrity of the bEND.3 monolayer. Immunofluorescence analysis and immunoblotting were performed to evaluate the inhibitory effects of pUR4 on TNF-α-induced FN deposition. To determine the mechanisms underlying pUR4-mediated endothelial permeability, cell morphology, stress fiber formation, myosin light chain (MLC) phosphorylation, and β1 integrin–mediated signaling were evaluated through immunofluorescence analysis and immunoblotting.ResultsExcessive FN was accumulated in the microvessels of the spinal cord after spinal nerve ligation; moreover, pUR4 inhibited TNF-α-induced FN deposition in the bEND.3 monolayer and maintained intact TEER and endothelial permeability. Furthermore, pUR4 reduced cell morphology alteration, actin stress fiber formation, and MLC phosphorylation, thereby attenuating paracellular gap formation. Moreover, pUR4 reduced β1 integrin activation and downstream signaling.ConclusionspUR4 reduces TNF-α-induced β1 integrin activation by depleting ECM FN, leading to a decrease in endothelial hyperpermeability and maintenance of monolayer integrity. These findings suggest therapeutic benefits of pUR4 in pathological vascular leakage treatment.

Highlights

  • The blood–spinal cord barrier (BSCB) is composed of a monolayer of endothelium linked with tight junctions and extracellular matrix (ECM)-rich basement membranes and is surrounded by astrocyte foot processes

  • Our study demonstrated that intact ECM FN is crucial for maintaining endothelial monolayer integrity and that excessive Tumor necrosis factor-α (TNF-α)-induced FN deposition enhances endothelial permeability by modulating β1 integrin–mediated stress fiber formation, actomyosin interaction, and paracellular gap formation

  • This study employed the FN inhibitor pUR4 to identify the importance of ECM FN as a mediator in tumor necrosis factor (TNF)-α-induced endothelial monolayer hyperpermeability

Read more

Summary

Introduction

The blood–spinal cord barrier (BSCB) is composed of a monolayer of endothelium linked with tight junctions and extracellular matrix (ECM)-rich basement membranes and is surrounded by astrocyte foot processes. Excessive FN deposition disrupts cell–cell adhesion in fibroblasts through β1 integrin ligation. The BSCB is composed of a monolayer of nonfenestrated capillary endothelium sealed with tight junctions, extracellular matrix (ECM)-rich basal lamina, and astrocyte foot processes [1]. The barrier function of the BSCB is strictly controlled by endothelial cell–cell tight junctions [2, 3]. Β1 integrin–ECM engagement induces integrin signaling cascades that modulate the expression of the tight junction proteins and maintain the endothelial cell integrity of the CNS microvessels [4, 16]. Under the pathological condition, ECM composition changes and β1 integrin–ECM interaction increases; endothelial integrity is disrupted [13, 17]

Methods
Results
Discussion
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