4.7 Article

DOCK4 Regulation of Rho GTPases Mediates Pulmonary Vascular Barrier Function

Journal

ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY
Volume 42, Issue 7, Pages 886-902

Publisher

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/ATVBAHA.122.317565

Keywords

actin cytoskeleton; animals; endothelial cells; lung; permeability; sphingosine-1-phospate receptor modulators; thrombin

Funding

  1. National Institutes of Health [R01HL065095, R01DK099507, P01HL036028, K01AR054984, R01HD040895, R35HL135821, R01EY031715]
  2. National Center for Advancing Translational Sciences, National Institutes of Health Award [UL 1TR002541]
  3. Harvard University and its affiliated academic health care centers

Ask authors/readers for more resources

The study reveals that DOCK4, an atypical Rho family GTPase GEF, plays a critical role in lung vascular barrier function by regulating AJs. DOCK4 deficiency leads to lung hemorrhage, increased microvascular permeability, and impaired response to S1P-induced permeability changes. Additionally, S1P may act as a potential upstream regulator of DOCK4 activity.
Background: The vascular endothelium maintains tissue-fluid homeostasis by controlling the passage of large molecules and fluid between the blood and interstitial space. The interaction of catenins and the actin cytoskeleton with VE-cadherin (vascular endothelial cadherin) is the primary mechanism for stabilizing AJs (adherens junctions), thereby preventing lung vascular barrier disruption. Members of the Rho (Ras homology) family of GTPases and conventional GEFs (guanine exchange factors) of these GTPases have been demonstrated to play important roles in regulating endothelial permeability. Here, we evaluated the role of DOCK4 (dedicator of cytokinesis 4)-an unconventional Rho family GTPase GEF in vascular function. Methods: We generated mice deficient in DOCK4, used DOCK4 silencing and reconstitution approaches in human pulmonary artery endothelial cells, used assays to evaluate protein localization, endothelial cell permeability, and small GTPase activation. Results: Our data show that DOCK4-deficient mice are viable. However, these mice have hemorrhage selectively in the lung, incomplete smooth muscle cell coverage in pulmonary vessels, increased basal microvascular permeability, and impaired response to S1P (sphingosine-1-phosphate)-induced reversal of thrombin-induced permeability. Consistent with this, DOCK4 rapidly translocates to the cell periphery and associates with the detergent-insoluble fraction following S1P treatment, and its absence prevents S1P-induced Rac-1 activation and enhancement of barrier function. Moreover, DOCK4-silenced pulmonary artery endothelial cells exhibit enhanced basal permeability in vitro that is associated with enhanced Rho GTPase activation. Conclusions: Our findings indicate that DOCK4 maintains AJs necessary for lung vascular barrier function by establishing the normal balance between RhoA (Ras homolog family member A) and Rac-1-mediated actin cytoskeleton remodeling, a previously unappreciated function for the atypical GEF family of molecules. Our studies also identify S1P as a potential upstream regulator of DOCK4 activity.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available