Matrix metalloproteinase activity is required for activity-induced angiogenesis in rat skeletal muscle

2000 ◽  
Vol 279 (4) ◽  
pp. H1540-H1547 ◽  
Author(s):  
T. L. Haas ◽  
M. Milkiewicz ◽  
S. J. Davis ◽  
A. L. Zhou ◽  
S. Egginton ◽  
...  

Proteolysis of the capillary basement membrane is a hallmark of inflammation-mediated angiogenesis, but it is undetermined whether proteolysis plays a critical role in the process of activity-induced angiogenesis. Matrix metalloproteinases (MMPs) constitute the major class of proteases responsible for degradation of basement membrane proteins. We observed significant elevations of mRNA and protein levels of both MMP-2 and membrane type 1 (MT1)-MMP (2.9 ± 0.7- and 1.5 ± 0.1-fold above control, respectively) after 3 days of chronic electrical stimulation of rat skeletal muscle. Inhibition of MMP activity via the inhibitor GM-6001 prevented the growth of new capillaries as assessed by the capillary-to-fiber ratio (1.34 ± 0.08 in GM-6001-treated muscles compared with 1.69 ± 0.03 in control 7-day-stimulated muscles). This inhibition correlated with a significant reduction in the number of capillaries with observable breaks in the basement membrane, as assessed by electron microscopy (0.27 ± 0.27% in GM-6001-treated muscles compared with 3.72 ± 0.65% in control stimulated muscles). Proliferation of capillary-associated cells was significantly elevated by 2 days and remained elevated throughout 14 days of stimulation. Capillary-associated cell proliferation during muscle stimulation was not affected by MMP inhibition (80.3 ± 9.3 nuclei in control and 63.5 ± 8.5 nuclei in GM-6001-treated animals). We conclude that MMP proteolysis of capillary basement membrane proteins is a critical component of physiological angiogenesis, and we postulate that capillary-associated proliferation precedes and occurs independently of endothelial cell sprout formation.

Cytoskeleton ◽  
2019 ◽  
Vol 76 (6) ◽  
pp. 371-382 ◽  
Author(s):  
Jacob W. Fleming ◽  
Andrew J. Capel ◽  
Rowan P. Rimington ◽  
Darren J. Player ◽  
Alexandra Stolzing ◽  
...  

2008 ◽  
Vol 49 (3) ◽  
pp. 137-141 ◽  
Author(s):  
Nemesha Desai ◽  
Jill Allen ◽  
Iaisha Ali ◽  
Vanessa Venning ◽  
Fenella Wojnarowska

1986 ◽  
Vol 63 (3) ◽  
pp. 571-580 ◽  
Author(s):  
Meeri Apaja-Sarkkinen ◽  
Helena Autio-Harmainen ◽  
Martti Alavaikko ◽  
Juha Risteli ◽  
Leila Risteli

2017 ◽  
Vol 114 (45) ◽  
pp. E9559-E9568 ◽  
Author(s):  
Qing He ◽  
Richard Bouley ◽  
Zun Liu ◽  
Marc N. Wein ◽  
Yan Zhu ◽  
...  

Alterations in the activity/levels of the extralarge G protein α-subunit (XLαs) are implicated in various human disorders, such as perinatal growth retardation. Encoded by GNAS, XLαs is partly identical to the α-subunit of the stimulatory G protein (Gsα), but the cellular actions of XLαs remain poorly defined. Following an initial proteomic screen, we identified sorting nexin-9 (SNX9) and dynamins, key components of clathrin-mediated endocytosis, as binding partners of XLαs. Overexpression of XLαs in HEK293 cells inhibited internalization of transferrin, a process that depends on clathrin-mediated endocytosis, while its ablation by CRISPR/Cas9 in an osteocyte-like cell line (Ocy454) enhanced it. Similarly, primary cardiomyocytes derived from XLαs knockout (XLKO) pups showed enhanced transferrin internalization. Early postnatal XLKO mice showed a significantly higher degree of cardiac iron uptake than wild-type littermates following iron dextran injection. In XLKO neonates, iron and ferritin levels were elevated in heart and skeletal muscle, where XLαs is normally expressed abundantly. XLKO heart and skeletal muscle, as well as XLKO Ocy454 cells, showed elevated SNX9 protein levels, and siRNA-mediated knockdown of SNX9 in XLKO Ocy454 cells prevented enhanced transferrin internalization. In transfected cells, XLαs also inhibited internalization of the parathyroid hormone and type 2 vasopressin receptors. Internalization of transferrin and these G protein-coupled receptors was also inhibited in cells expressing an XLαs mutant missing the Gα portion, but not Gsα or an N-terminally truncated XLαs mutant unable to interact with SNX9 or dynamin. Thus, XLαs restricts clathrin-mediated endocytosis and plays a critical role in iron/transferrin uptake in vivo.


Sign in / Sign up

Export Citation Format

Share Document