Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 41(11): 2756-2769, 2021 11.
Article in English | MEDLINE | ID: mdl-34551592

ABSTRACT

Objective: Angiogenic factor AGGF1 (angiogenic factor with G-patch and FHA [Forkhead-associated] domain 1) promotes angiogenesis as potently as VEGFA (vascular endothelial growth factor A) and regulates endothelial cell (EC) proliferation, migration, specification of multipotent hemangioblasts and venous ECs, hematopoiesis, and vascular development and causes vascular disease Klippel-Trenaunay syndrome when mutated. However, the receptor for AGGF1 and the underlying molecular mechanisms remain to be defined. Approach and Results: Using functional blocking studies with neutralizing antibodies, we identified [alpha]5[beta]1 as the receptor for AGGF1 on ECs. AGGF1 interacts with [alpha]5[beta]1 and activates FAK (focal adhesion kinase), Src (proto-oncogene tyrosine-protein kinase), and AKT (protein kinase B). Functional analysis of 12 serial N-terminal deletions and 13 C-terminal deletions by every 50 amino acids mapped the angiogenic domain of AGGF1 to a domain between amino acids 604-613 (FQRDDAPAS). The angiogenic domain is required for EC adhesion and migration, capillary tube formation, and AKT activation. The deletion of the angiogenic domain eliminated the effects of AGGF1 on therapeutic angiogenesis and increased blood flow in a mouse model for peripheral artery disease. A 40-mer or 15-mer peptide containing the angiogenic domain blocks AGGF1 function, however, a 15-mer peptide containing a single amino acid mutation from -RDD- to -RGD- (a classical RGD integrin-binding motif) failed to block AGGF1 function. Conclusions: We have identified integrin [alpha]5[beta]1 as an EC receptor for AGGF1 and a novel AGGF1-mediated signaling pathway of [alpha]5[beta]1-FAK-Src-AKT for angiogenesis. Our results identify an FQRDDAPAS angiogenic domain of AGGF1 crucial for its interaction with [alpha]5[beta]1 and signaling.


Subject(s)
Angiogenic Proteins/metabolism , Endothelial Cells/metabolism , Hindlimb/blood supply , Integrin alpha5beta1/metabolism , Ischemia/metabolism , Neovascularization, Physiologic , 3T3-L1 Cells , Angiogenesis Inducing Agents/pharmacology , Angiogenic Proteins/genetics , Angiogenic Proteins/pharmacology , Animals , Disease Models, Animal , Endothelial Cells/drug effects , Female , Focal Adhesion Kinase 1/metabolism , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Integrin alpha5beta1/genetics , Ischemia/drug therapy , Ischemia/genetics , Ischemia/physiopathology , Ligands , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Phosphorylation , Protein Interaction Domains and Motifs , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , src-Family Kinases/metabolism
2.
Cell Signal ; 35: 231-241, 2017 07.
Article in English | MEDLINE | ID: mdl-28431986

ABSTRACT

Previous genetic studies suggested that variants in NINJ2 (encode ninjurin2) confer risk to ischemic stroke or large artery atherosclerotic stroke. However, the underlying mechanisms of NINJ2 in ischemic stroke or atherosclerosis are still unknown. In this study, we hypothesized that NINJ2 may play a role in endothelial inflammation and activation, and regulate the process of atherosclerosis. Here, we demonstrated that NINJ2 can regulate the expression of a panel of genes that are associated with inflammation and atherosclerosis (e.g. IL-1ß, TNF-α, IL-8, IL-6, ICAM-1 and E-selectin) in human vascular endothelial cells (HUVECs). Moreover, we found the expression of ninjurin2 is upregulated in LPS stimulated HUVECs and mouse aorta, and it can regulate LPS-induced endothelial activation and the adhesion of monocytes to endothelial cells. We also found that NINJ2 can regulate NF-κB and c-jun through interacting with TLR4. In conclusion, our study suggests that ninjurin2 is a novel regulator of endothelia inflammation and activation through TLR4 signaling pathways, and these data provided new insights into the mechanisms between NINJ2 and atherosclerosis.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Coronary Artery Disease/genetics , Stroke/genetics , Toll-Like Receptor 4/genetics , Animals , Cell Adhesion/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Coronary Artery Disease/pathology , Endothelium/metabolism , Endothelium/pathology , Gene Expression Regulation/genetics , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/genetics , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Monocytes/metabolism , Monocytes/pathology , NF-kappa B/genetics , Signal Transduction/genetics , Stroke/pathology , Toll-Like Receptor 4/metabolism
3.
Hum Mol Genet ; 25(23): 5094-5110, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27522498

ABSTRACT

Aggf1 is the first gene identified for Klippel-Trenaunay syndrome (KTS), and encodes an angiogenic factor. However, the in vivo roles of Aggf1 are incompletely defined. Here we demonstrate that Aggf1 is essential for both physiological angiogenesis and pathological tumour angiogenesis in vivo. Two lines of Aggf1 knockout (KO) mice showed a particularly severe phenotype as no homozygous embryos were observed and heterozygous mice also showed embryonic lethality (haploinsufficient lethality) observed only for Vegfa and Dll4. Aggf1+/- KO caused defective angiogenesis in yolk sacs and embryos. Survived adult heterozygous mice exhibit frequent haemorrhages and increased vascular permeability due to increased phosphorylation and reduced membrane localization of VE-cadherin. AGGF1 inhibits VE-cadherin phosphorylation, increases plasma membrane VE-cadherin in ECs and in mice, blocks vascular permeability induced by ischaemia-reperfusion (IR), restores depressed cardiac function and contraction, reduces infarct sizes, cardiac fibrosis and necrosis, haemorrhages, edema, and macrophage density associated with IR. Mechanistically, AGGF1 promotes angiogenesis by activating catalytic p110α subunit and p85α regulatory subunit of PI3K, leading to activation of AKT, GSK3ß and p70S6K. AKT activation is significantly reduced in heterozygous KO mice and isolated KO ECs, which can be rescued by exogenous AGGF1. ECs from KO mice show reduced capillary angiogenesis, which is rescued by AGGF1 and AKT. Tumour growth/angiogenesis is reduced in heterozygous mice, which was associated with reduced activation of p110α, p85α and AKT. Together with recent identification of somatic mutations in p110α (encoded by PIK3CA), our data establish a potential mechanistic link between AGGF1 and PIK3CA, the two genes identified for KTS.


Subject(s)
Angiogenic Proteins/genetics , Antigens, CD/genetics , Cadherins/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Klippel-Trenaunay-Weber Syndrome/genetics , Neovascularization, Pathologic/genetics , Angiogenic Proteins/biosynthesis , Animals , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Class I Phosphatidylinositol 3-Kinases/biosynthesis , Embryonic Development/genetics , Haploinsufficiency/genetics , Humans , Klippel-Trenaunay-Weber Syndrome/physiopathology , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...