Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Dev Biol ; 469: 54-67, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32971120

ABSTRACT

Tie1 is a receptor tyrosine kinase expressed in endothelial cells, where it modulates Angiopoietin/Tie2 signaling. Previous studies have shown that mouse Tie1 mutants exhibit severe cardiovascular defects; however, much remains to be learned about the role of Tie1, especially during cardiac development. To further understand Tie1 function, we generated a zebrafish tie1 mutant line. Homozygous mutant embryos display reduced endothelial and endocardial cell numbers and reduced heart size. Live imaging and ultrastructural analyses at embryonic stages revealed increased cardiac jelly thickness as well as cardiomyocyte defects, including a loss of sarcomere organization and altered cell shape. Transcriptomic profiling of embryonic hearts uncovered the downregulation of tll1, which encodes a Tolloid-like protease, in tie1-/- compared with wild-type siblings. Using mRNA injections into one-cell stage embryos, we found that tll1 overexpression could partially rescue the tie1 mutant cardiac phenotypes including the endocardial and myocardial cell numbers as well as the cardiac jelly thickness. Altogether, our results indicate the importance of a Tie1-Tolloid-like 1 axis in paracrine signaling during cardiac development.


Subject(s)
Heart/embryology , Tolloid-Like Metalloproteinases/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Heart Defects, Congenital/genetics , Morphogenesis , Mutation , Myocytes, Cardiac/cytology , Receptor, TIE-1/genetics , Receptor, TIE-1/physiology , Tolloid-Like Metalloproteinases/genetics , Transcriptome , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
2.
Int J Mol Sci ; 21(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32947856

ABSTRACT

TIE1 is a cell surface protein expressed in endothelial cells. Involved in angiogenesis and lymphangiogenesis, including morphogenesis of lymphatic valves, TIE1 is important for lymphatic system functional integrity. The main purpose of this study was to identify different variants in the TIE1 gene that could be associated with lymphatic malformations or dysfunction and predisposition for lymphedema. In a cohort of 235 Italian lymphedema patients, who tested negative for variants in known lymphedema genes, we performed a further test for new candidate genes, including TIE1. Three probands carried different variants in TIE1. Two of these segregated with lymphedema or lymphatic dysfunction in familial cases. Variants in TIE1 could contribute to the onset of lymphedema. On the basis of our findings, we propose TIE1 as a candidate gene for comprehensive genetic testing of lymphedema.


Subject(s)
Lymphatic Abnormalities/genetics , Lymphedema/genetics , Receptor, TIE-1/physiology , Aged , Amino Acid Sequence , Chromosomes, Human, Pair 1/genetics , Computer Simulation , Female , Genetic Association Studies , Genetic Testing , Humans , Italy , Lymphangiogenesis/genetics , Male , Middle Aged , Models, Molecular , Mutation , Pedigree , Protein Conformation , Receptor, TIE-1/genetics , Retrospective Studies , Sequence Alignment , Young Adult
3.
Sci Rep ; 7(1): 3375, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28611395

ABSTRACT

Blood flow influences atherosclerosis by generating wall shear stress, which alters endothelial cell (EC) physiology. Low shear stress induces dedifferentiation of EC through a process termed endothelial-to-mesenchymal transition (EndMT). The mechanisms underlying shear stress-regulation of EndMT are uncertain. Here we investigated the role of the transcription factor Snail in low shear stress-induced EndMT. Studies of cultured EC exposed to flow revealed that low shear stress induced Snail expression. Using gene silencing it was demonstrated that Snail positively regulated the expression of EndMT markers (Slug, N-cadherin, α-SMA) in EC exposed to low shear stress. Gene silencing also revealed that Snail enhanced the permeability of endothelial monolayers to macromolecules by promoting EC proliferation and migration. En face staining of the murine aorta or carotid arteries modified with flow-altering cuffs demonstrated that Snail was expressed preferentially at low shear stress sites that are predisposed to atherosclerosis. Snail was also expressed in EC overlying atherosclerotic plaques in coronary arteries from patients with ischemic heart disease implying a role in human arterial disease. We conclude that Snail is an essential driver of EndMT under low shear stress conditions and may promote early atherogenesis by enhancing vascular permeability.


Subject(s)
Carotid Arteries/pathology , Endothelium, Vascular/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation , Plaque, Atherosclerotic/pathology , Snail Family Transcription Factors/metabolism , Stress, Mechanical , Animals , Aorta/metabolism , Aorta/pathology , Carotid Arteries/metabolism , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Knockout , Nuclear Proteins/physiology , Plaque, Atherosclerotic/metabolism , Receptor, TIE-1/physiology , Snail Family Transcription Factors/genetics , Swine , Twist-Related Protein 1/physiology
4.
Cell Rep ; 12(11): 1761-73, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26344773

ABSTRACT

Tie1 is a mechanistically poorly characterized endothelial cell (EC)-specific orphan receptor. Yet, Tie1 deletion is embryonic lethal and Tie1 has been implicated in critical vascular pathologies, including atherosclerosis and tumor angiogenesis. Here, we show that Tie1 does not function independently but exerts context-dependent effects on the related receptor Tie2. Tie1 was identified as an EC activation marker that is expressed during angiogenesis by a subset of angiogenic tip and remodeling stalk cells and downregulated in the adult quiescent vasculature. Functionally, Tie1 expression by angiogenic EC contributes to shaping the tip cell phenotype by negatively regulating Tie2 surface presentation. In contrast, Tie1 acts in remodeling stalk cells cooperatively to sustain Tie2 signaling. Collectively, our data support an interactive model of Tie1 and Tie2 function, in which dynamically regulated Tie1 versus Tie2 expression determines the net positive or negative effect of Tie1 on Tie2 signaling.


Subject(s)
Receptor, TIE-1/physiology , Receptor, TIE-2/physiology , Vascular Remodeling/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Endothelial Cells/enzymology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Retinal Vessels/physiology , Signal Transduction
5.
Arterioscler Thromb Vasc Biol ; 34(6): 1221-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24764452

ABSTRACT

OBJECTIVE: The genetic program underlying lymphatic development is still incompletely understood. This study aims to dissect the role of receptor tyrosine kinase with immunoglobulin-like and EGF (epidermal growth factor)-like domains 1 (Tie1) and Tie2 in lymphatic formation using genetically modified mouse models. APPROACH AND RESULTS: We generated conditional knockout mouse models targeting Tie1, Tie2, and angiopoietin-2 in this study. Tie1(ΔICD/ΔICD) mice, with its intracellular domain targeted, appeared normal at E10.5 but displayed subcutaneous edema by E13.5. Lymph sac formation occurred in Tie1(ΔICD/ΔICD) mice, but they had defects with the remodeling of primary lymphatic network to form collecting vessels and valvulogenesis. Consistently, induced deletion of Tie1-ICD postnatally using a ubiquitous Cre deleter led to abnormal lymphangiogenesis and valve formation in Tie1-ICD(iUCKO/-) mice. In comparison with the lymphatic phenotype of Tie1 mutants, we found that the diameter of lymphatic capillaries was significantly less in mice deficient of angiopoietin-2, besides the disruption of collecting lymphatic vessel formation as previously reported. There was also no lymphedema observed in Ang2(-/-) mice during embryonic development, which differs from that of Tie1(ΔICD/ΔICD) mice. We further investigated whether Tie1 exerted its function via Tie2 during lymphatic development. To our surprise, genetic deletion of Tie2 (Tie2(iUCKO/-)) in neonate mice did not affect lymphatic vessel growth and maturation. CONCLUSIONS: In contrast to the important role of Tie2 in the regulation of blood vascular development, Tie1 is crucial in the process of lymphatic remodeling and maturation, which is independent of Tie2.


Subject(s)
Lymphangiogenesis/physiology , Lymphatic Vessels/physiology , Receptor, TIE-1/physiology , Receptor, TIE-2/physiology , Angiopoietin-2/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, TIE-1/genetics , Receptor, TIE-2/genetics , Signal Transduction
6.
J Clin Invest ; 124(2): 824-34, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24430181

ABSTRACT

The endothelial Tie1 receptor is ligand-less, but interacts with the Tie2 receptor for angiopoietins (Angpt). Angpt2 is expressed in tumor blood vessels, and its blockade inhibits tumor angiogenesis. Here we found that Tie1 deletion from the endothelium of adult mice inhibits tumor angiogenesis and growth by decreasing endothelial cell survival in tumor vessels, without affecting normal vasculature. Treatment with VEGF or VEGFR-2 blocking antibodies similarly reduced tumor angiogenesis and growth; however, no additive inhibition was obtained by targeting both Tie1 and VEGF/VEGFR-2. In contrast, treatment of Tie1-deficient mice with a soluble form of the extracellular domain of Tie2, which blocks Angpt activity, resulted in additive inhibition of tumor growth. Notably, Tie1 deletion decreased sprouting angiogenesis and increased Notch pathway activity in the postnatal retinal vasculature, while pharmacological Notch suppression in the absence of Tie1 promoted retinal hypervasularization. Moreover, substantial additive inhibition of the retinal vascular front migration was observed when Angpt2 blocking antibodies were administered to Tie1-deficient pups. Thus, Tie1 regulates tumor angiogenesis, postnatal sprouting angiogenesis, and endothelial cell survival, which are controlled by VEGF, Angpt, and Notch signals. Our results suggest that targeting Tie1 in combination with Angpt/Tie2 has the potential to improve antiangiogenic therapy.


Subject(s)
Angiopoietin-1/antagonists & inhibitors , Endothelial Cells/cytology , Endothelium, Vascular/metabolism , Gene Deletion , Receptor, TIE-1/genetics , Retinal Vessels/pathology , Angiogenesis Inhibitors/chemistry , Angiopoietin-1/metabolism , Animals , Apoptosis , Cell Survival , Homozygote , Ligands , Mice , Mice, Transgenic , Neoplasm Transplantation , Neovascularization, Pathologic , Phenotype , Receptor, TIE-1/physiology , Receptor, TIE-2/physiology , Receptors, Notch/metabolism , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
J Biol Regul Homeost Agents ; 27(3): 827-39, 2013.
Article in English | MEDLINE | ID: mdl-24152847

ABSTRACT

The Angiopoietin/Tie system is a key regulator of vascular remodeling, maturation, angiogenesis and lymphangiogenesis. In humans there are three angiopoietins: Angiopoietin-1 (Ang1), Angiopoietin-2 (Ang2), and Angiopoietin-4 (Ang4). Ang1 and Ang2 are the best characterized angiopoietins. The angiopoietin receptor system consists of two type I tyrosine kinase receptors (Tie1 and Tie2). Tie2 binds all known angiopoietins. We sought to characterize Ang1, Ang2, Tie1 and Tie2 expression and functions in human basophils and mast cells. Basophils, LAD-2 cells and Human Lung Mast Cells (HLMCs) constitutively express Ang1 and Ang2 mRNA. Intracellular staining for Ang1 and Ang2 was stronger in basophils than in mast cells. Immunoelectron microscopy demonstrated Ang1 in cytoplasmic vesicles of basophils. The protein kinase C activators phorbol diester (PMA) and bryostatin 1 (Bryo1) stimulated basophils to rapidly release a large amount of Ang1. PMA-induced Ang1 release was inhibited by brefeldin A. Tie1 and Tie2 mRNAs were expressed in basophils, LAD-2 and HLMCs. Basophils, LAD-2 and HLMCs expressed Tie1 on the cell surface. HLMCs and LAD-2 expressed Tie2 on the cell surface, whereas basophils did not. Ang1, but not Ang2, induced migration of mast cells through the engagement of Tie2. Neither Ang1 nor Ang2 induced basophil chemotaxis. We have identified a novel mechanism of cross-talk between human basophils and mast cells mediated by the Ang1/Tie2 system that might be relevant in the orchestration of inflammatory and neoplastic angiogenesis.


Subject(s)
Angiopoietin-1/physiology , Angiopoietin-2/physiology , Basophils/physiology , Mast Cells/physiology , Receptor, TIE-1/physiology , Receptor, TIE-2/physiology , Angiopoietin-1/analysis , Angiopoietin-2/analysis , Basophils/chemistry , Cells, Cultured , Chemotaxis , Humans , Lymphangiogenesis , Mast Cells/chemistry , Neovascularization, Physiologic , Receptor, TIE-1/analysis , Receptor, TIE-2/analysis
8.
Cold Spring Harb Perspect Med ; 2(9): a006550, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22951441

ABSTRACT

The angiopoietin-Tie signaling system is a vascular-specific receptor tyrosine kinase pathway that is essential for normal vascular development. Although the basic functioning of the pathway is understood, many uncertainties remain about the role of certain members of the pathway, particularly angiopoietin-2 (Ang2), in pathological vascular remodeling and angiogenesis. We summarize the components of the angiopoietin-Tie pathway and then focus on studies that highlight the role of Ang2 in disease settings, including cancer and inflammation. The expression of Ang2 is elevated in many cancers and types of inflammation, which prompted the development of specific reagents to block its interaction with the Tie2 receptor. The application of these reagents in preclinical models of inflammation and cancer has begun to elucidate the role of Ang2 in vascular remodeling and disease pathogenesis and has led to emerging clinical tests of Ang2 inhibitors.


Subject(s)
Angiopoietin-2/physiology , Receptor, TIE-1/physiology , Receptor, TIE-2/physiology , Signal Transduction/physiology , Animals , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Humans , Ligands , Mice , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/etiology , Receptor Protein-Tyrosine Kinases/physiology , Receptor, TIE-1/agonists , Receptor, TIE-1/antagonists & inhibitors , Receptor, TIE-2/agonists , Receptor, TIE-2/antagonists & inhibitors , Up-Regulation , Vasculitis/etiology
9.
J Clin Invest ; 121(6): 2278-89, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21606590

ABSTRACT

Angiopoietin-1/Tek signaling is a critical regulator of blood vessel development, with conventional knockout of angiopoietin-1 or Tek in mice being embryonically lethal due to vascular defects. In addition, angiopoietin-1 is thought to be required for the stability of mature vessels. Using a Cre-Lox conditional gene targeting approach, we have studied the role of angiopoietin-1 in embryonic and adult vasculature. We report here that angiopoietin-1 is critical for regulating both the number and diameter of developing vessels but is not required for pericyte recruitment. Cardiac-specific knockout of angiopoietin-1 reproduced the phenotype of the conventional knockout, demonstrating that the early vascular abnormalities arise from flow-dependent defects. Strikingly, deletion in the entire embryo after day E13.5 produced no immediate vascular phenotype. However, when combined with injury or microvascular stress, angiopoietin-1 deficiency resulted in profound organ damage, accelerated angiogenesis, and fibrosis. These findings redefine our understanding of the biological roles of angiopoietin-1: it is dispensable in quiescent vessels but has a powerful ability to modulate the vascular response after injury.


Subject(s)
Angiopoietin-1/physiology , Blood Vessels/embryology , Blood Vessels/injuries , Neovascularization, Physiologic/physiology , Wound Healing/physiology , Angiopoietin-1/deficiency , Angiopoietin-1/genetics , Animals , Blood Vessels/cytology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/physiopathology , Fetal Heart/growth & development , Fetal Heart/pathology , Gene Expression Regulation, Developmental , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/pathology , Liver/blood supply , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Neovascularization, Pathologic/embryology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Pericytes/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptor, TIE-1/physiology , Receptor, TIE-2 , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology
10.
J Clin Invest ; 121(6): 2157-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21606600

ABSTRACT

Twenty years after the discovery of the vascular endothelial Tie receptor tyrosine kinases and 15 years after the discovery of the Tie2 ligand, angiopoietin-1 (Angpt1, also known as Ang1), a study published in the current issue of the JCI reveals an unexpected loss-of-function phenotype of mice conditionally deleted of the Angpt1 gene. The results suggest that Angpt1 is needed as a vascular stabilizing factor that organizes and limits the angiogenesis response and protects from pathological consequences, such as tissue fibrosis.


Subject(s)
Angiopoietin-1/physiology , Neovascularization, Physiologic/physiology , Angiopoietin-1/deficiency , Angiopoietin-1/genetics , Angiopoietin-2/physiology , Animals , Blood Vessels/embryology , Capillaries/cytology , Capillaries/growth & development , Capillary Permeability , Cell Adhesion , Cell Survival , Endothelial Cells/cytology , Fibrosis , Humans , Mice , Mice, Knockout , Models, Cardiovascular , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Neoplasms/blood supply , Neovascularization, Pathologic/embryology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/genetics , Pericytes/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptor, TIE-1/physiology , Receptor, TIE-2
11.
J Clin Invest ; 121(4): 1624-35, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21383501

ABSTRACT

Although the response of endothelial cells to the disturbed blood flow in the vicinity of atherosclerotic lesions is known to be distinct from that elicited by nonatherogenic laminar flow, the mechanisms involved are poorly understood. Our initial studies confirmed that expression of the endothelial receptor tyrosine kinase Tie1 was evident at regions of atherogenic flow in mature animals. We therefore hypothesized that Tie1 plays a role in the endothelial response to atherogenic shear stress. Consistent with this, we found that Tie1+/- mice bred to the apoE-deficient background displayed a 35% reduction in atherosclerosis relative to Tie1+/+;Apoe-/- mice. Since deletion of Tie1 results in embryonic lethality secondary to vascular dysfunction, we used conditional and inducible mutagenesis to study the effect of endothelial-specific Tie1 attenuation on atherogenesis in Apoe-/- mice and found a dose-dependent decrease in atherosclerotic lesions. Analysis of primary aortic endothelial cells indicated that atheroprotective laminar flow decreased Tie1 expression in vitro. Attenuation of Tie1 was associated with an increase in eNOS expression and Tie2 phosphorylation. In addition, Tie1 attenuation increased IkBα expression while decreasing ICAM levels. In summary, we have found that shear stress conditions that modulate atherogenic events also regulate Tie1 expression. Therefore, Tie1 may play a novel proinflammatory role in atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Receptor, TIE-1/deficiency , Receptor, TIE-1/genetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Base Sequence , Cell Adhesion Molecules/genetics , DNA Primers/genetics , Disease Models, Animal , Endothelial Cells/physiology , Female , Gene Expression , Hemorheology , Lac Operon , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type III/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, TIE-1/physiology , Signal Transduction , Stress, Mechanical , rho-Associated Kinases/genetics
12.
Trends Cardiovasc Med ; 21(4): 118-23, 2011 May.
Article in English | MEDLINE | ID: mdl-22681967

ABSTRACT

Atherosclerotic plaques develop in a nonrandom manner along the vasculature following a hemodynamically determined distribution profile. The pathogenesis of shear stress-induced inflammation and atherosclerotic lesion formation has led to discussions about personalized strategies in prevention and treatment. Recent discoveries involving the tyrosine kinase receptor Tie1 in (1) mechanotransduction, (2) inflammation, and (3) neovascularization have invigorated these efforts. In this review, we present the current understanding on Tie1 and its role in these key components of atherogenesis.


Subject(s)
Atherosclerosis/enzymology , Receptor, TIE-1/physiology , Stress, Mechanical , Animals , Forecasting , Humans , Inflammation/enzymology , Mechanotransduction, Cellular/physiology , Mice , Neovascularization, Pathologic/enzymology
13.
FEBS Lett ; 583(6): 1023-8, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19236867

ABSTRACT

Tie-1 is an endothelial specific receptor tyrosine kinase that is upregulated in diseases such as atherosclerosis and rheumatoid arthritis. We recently demonstrated that Tie-1 induced a proinflammatory response when overexpressed in endothelial cells. Here, we used a complementary approach and suppressed endogenous Tie-1 expression in endothelial cells to examine its function by microarray analysis. Tie-1 appeared to govern expression of many genes involved in inflammation. Expression knockdown of Tie-1 significantly reduced endothelial conditioned medium ability to stimulate MCP-1 production in U937 cells. Collectively, our results support the notion that Tie-1 has an inflammatory function in endothelial cells.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Inflammation/genetics , RNA, Small Interfering/pharmacology , Receptor, TIE-1/antagonists & inhibitors , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Profiling , Genome, Human , Humans , Monocytes/drug effects , Monocytes/physiology , Oligonucleotide Array Sequence Analysis , RNA Interference/physiology , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-1/physiology , U937 Cells
14.
Circ Res ; 101(3): 268-76, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17585070

ABSTRACT

Nuclear factor kappa B (NF-kappaB) participates in hypertension-induced vascular and target-organ damage. We tested whether or not endothelial cell-specific NF-kappaB suppression would be ameliorative. We generated Cre/lox transgenic mice with endothelial cell-restricted NF-kappaB super-repressor IkappaBalphaDeltaN (Tie-1-DeltaN mice) overexpression. We confirmed cell-specific IkappaBalphaDeltaN expression and reduced NF-kappaB activity after TNF-alpha stimulation in primary endothelial cell culture. To induce hypertension with target-organ damage, we fed mice a high-salt diet and N(omega)-nitro-l-arginine-methyl-ester (L-NAME) and infused angiotensin (Ang) II. This treatment caused a 40-mm Hg blood pressure increase in both Tie-1-DeltaN and control mice. In contrast to control mice, Tie-1-DeltaN mice developed a milder renal injury, reduced inflammation, and less albuminuria. RT-PCR showed significantly reduced expression of the NF-kappaB targets VCAM-1 and ICAM-1, compared with control mice. Thus, the data demonstrate a causal link between endothelial NF-kappaB activation and hypertension-induced renal damage. We conclude that in vivo NF-kappaB suppression in endothelial cells stops a signaling cascade leading to reduced hypertension-induced renal damage despite high blood pressure.


Subject(s)
Endothelial Cells/metabolism , Hypertension/complications , Kidney Diseases/prevention & control , NF-kappa B/antagonists & inhibitors , Albuminuria/etiology , Albuminuria/prevention & control , Angiotensin II/toxicity , Animals , Atherosclerosis/physiopathology , Cells, Cultured , Gene Expression Regulation , Humans , Hypertension/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/physiology , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Kidney Diseases/etiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Cardiovascular , NF-KappaB Inhibitor alpha , NF-kappa B/physiology , NG-Nitroarginine Methyl Ester/toxicity , Nephritis/prevention & control , Nitric Oxide/physiology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Organ Specificity , Receptor, TIE-1/physiology , Recombinant Fusion Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sodium Chloride, Dietary/toxicity , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
16.
J Biol Chem ; 280(3): 2361-9, 2005 Jan 21.
Article in English | MEDLINE | ID: mdl-15536067

ABSTRACT

Here, we demonstrate the expression of carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1) in angiogenic sprouts but not in large mother blood vessels within tumor tissue. Correspondingly, only human microvascular endothelial cells involved in in vitro tube formation exhibit CEACAM1. CEACAM1-overexpressing versus CEACAM1-silenced human microvascular endothelial cells were used in migration and tube formation assays. CEACAM1-overexpressing microvascular endothelial cells showed prolonged survival and increased tube formation when they were stimulated with vascular endothelial growth factor (VEGF), whereas CEACAM1 silencing via small interfering RNA blocks these effects. Gene array and LightCycler analyses show an up-regulation of angiogenic factors such as VEGF, VEGF receptor 2, angiopoietin-1, angiopoietin-2, tie-2, angiogenin, and interleukin-8 but a down-regulation of collagen XVIII/endostatin and Tie-1 in CEACAM1-overexpressing microvascular endothelial cells. Western blot analyses confirm these results for VEGF and endostatin at the protein level. These results suggest that constitutive expression of CEACAM1 in microvascular endothelial cells switches them to an angiogenic phenotype, whereas CEACAM1 silencing apparently abrogates the VEGF-induced morphogenetic effects during capillary formation. Thus, strategies targeting the endothelial up-regulation of CEACAM1 might be promising for antiangiogenic tumor therapy.


Subject(s)
Antigens, CD/physiology , Antigens, Differentiation/physiology , Endothelium, Vascular/physiology , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Base Sequence , Cell Adhesion Molecules , Cell Survival/physiology , Cells, Cultured , Collagen Type XVIII/physiology , DNA Primers , Down-Regulation/physiology , Endostatins/physiology , Endothelium, Vascular/cytology , Humans , Receptor, TIE-1/physiology
18.
Circ Res ; 94(3): 394-401, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14670840

ABSTRACT

Nonlaminar flow is a major predisposing factor to atherosclerosis. Yet little is known regarding hemodynamic gene regulation in disease-prone areas of the vascular tree in vivo. We have determined spatial patterns of expression of endothelial cell receptors in the arterial tree and of reporter gene constructs in transgenic animals. In this study we show that the endothelial cell-specific receptor Tie1 is induced by disturbed flow in atherogenic vascular niches. Specifically, tie1 expression in the adult is upregulated in vascular bifurcations and branching points along the arterial tree. It is often confined to a single ring of endothelial cells functioning as sphincters and hence experiencing the steepest gradient in shear stress. In aortic valves, tie1 is asymmetrically induced only in endothelial cells encountering changes in flow direction. Disturbance of laminar flow by a surgical interposition of a vein into an artery led to induction of tie1, specifically in the region where the differently sized vessels adjoin. In pathological settings, tie1 expression is specifically induced in areas of disturbed flow because of the emergence of aneurysms and, importantly, in endothelial cells precisely overlying atherosclerotic plaques. Hemodynamic features of atherosclerotic lesion-prone regions, recreated in vitro with the aid of a flow chamber with a built-in step, corroborated an upregulated tie1 promoter activity only in cells residing where flow separation and recirculation take place. These defined promoter elements might be harnessed for targeting gene expression to atherosclerotic lesions.


Subject(s)
Arteriosclerosis/physiopathology , Endothelial Cells/metabolism , Promoter Regions, Genetic/genetics , Receptor, TIE-1/genetics , Animals , Animals, Genetically Modified , Aortic Aneurysm, Abdominal/genetics , Aortic Valve/metabolism , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Female , Gene Expression Regulation , In Situ Hybridization , Lac Operon/genetics , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Valve/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, TIE-1/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regional Blood Flow , Retina/metabolism , Rheology , Stress, Mechanical , Veins/physiopathology , Veins/transplantation
19.
Clin Cancer Res ; 9(10 Pt 2): 3821S-6S, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14506178

ABSTRACT

PURPOSE: Tie-1 is a transmembrane tyrosine kinase expressed in vascular endothelial cells during angiogenic processes and vasculogenesis. Here we evaluate targeting of rebuilding endothelium with (125)I-labeled Tie-1 monoclonal antibodies (mAbs) in mice. EXPERIMENTAL DESIGN: At first, activity of Tie-1 kinase during reforming of blood vessels was evaluated in melanoma allografts in transgenic mice with 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside staining of the Tie-1 promoter gene. Subsequently, in vivo targeting of the healing wound was evaluated with iodinated Tie-1 mAbs in mice, and finally, after confirming the specificities for targeting, we evaluated the biodistribution of Tie-1 mAbs in a melanoma model. RESULTS: Tie-1 mAbs target epithelial skin wounds in mice. Biokinetics of (125)I-Tie-1 mAbs demonstrate a stabilized equilibrium between the blood and wound over 3 days. The accumulation in wound is 21% injected dose/gram (ID/g) and 17% ID/g at 48 h with the two clones of Tie-1 mAbs, 3c4c7 and 10f11g6. Tie-1 promoter is active in the melanoma model in mice. In melanomas, the tumor accumulation is 5% and 4.4% ID/g, and the tumor:liver values are 2.7 and 4.4, respectively, for the two clones of Tie-1 mAbs, 3c4c7 and 10f11g6. The clearance of (125)I-Tie-1 antibodies is slow when compared with that of the iodinated control antibody. CONCLUSIONS: Targeting to wound is demonstrated with the Tie-1 mAbs. Accordingly, tumor targeting of melanoma is expertized with the same antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Endothelium, Vascular/cytology , Receptor, TIE-1/immunology , Animals , Antibodies, Monoclonal/chemistry , Cell Division/drug effects , Cell Line, Tumor , Chromogenic Compounds/pharmacology , Galactosides/pharmacology , Humans , Indoles/pharmacology , Kinetics , Lac Operon , Melanoma/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Neovascularization, Pathologic , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptor, TIE-1/physiology , Time Factors , Tissue Distribution , Wound Healing , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...