The recombinant lectin-like domain of thrombomodulin inhibits angiogenesis through interaction with Lewis Y antigen

Blood ◽  
2012 ◽  
Vol 119 (5) ◽  
pp. 1302-1313 ◽  
Author(s):  
Cheng-Hsiang Kuo ◽  
Po-Ku Chen ◽  
Bi-Ing Chang ◽  
Meng-Chen Sung ◽  
Chung-Sheng Shi ◽  
...  

AbstractLewis Y Ag (LeY) is a cell-surface tetrasaccharide that participates in angiogenesis. Recently, we demonstrated that LeY is a specific ligand of the recombinant lectin-like domain of thrombomodulin (TM). However, the biologic function of interaction between LeY and TM in endothelial cells has never been investigated. Therefore, the role of LeY in tube formation and the role of the recombinant lectin-like domain of TM—TM domain 1 (rTMD1)—in antiangiogenesis were investigated. The recombinant TM ectodomain exhibited lower angiogenic activity than did the recombinant TM domains 2 and 3. rTMD1 interacted with soluble LeY and membrane-bound LeY and inhibited soluble LeY-mediated chemotaxis of endothelial cells. LeY was highly expressed on membrane ruffles and protrusions during tube formation on Matrigel. Blockade of LeY with rTMD1 or Ab against LeY inhibited endothelial tube formation in vitro. Epidermal growth factor (EGF) receptor in HUVECs was LeY modified. rTMD1 inhibited EGF receptor signaling, chemotaxis, and tube formation in vitro, and EGF-mediated angiogenesis and tumor angiogenesis in vivo. We concluded that LeY is involved in vascular endothelial tube formation and rTMD1 inhibits angiogenesis via interaction with LeY. Administration of rTMD1 or recombinant adeno-associated virus vector carrying TMD1 could be a promising antiangiogenesis strategy.

Molecules ◽  
2020 ◽  
Vol 25 (22) ◽  
pp. 5343
Author(s):  
Michał Otręba ◽  
Leon Kośmider ◽  
Jerzy Stojko ◽  
Anna Rzepecka-Stojko

Polyphenols have recently gained popularity among the general public as products and diets classified as healthy and containing naturally occurring phenols. Many polyphenolic extracts are available on the market as dietary supplements, functional foods, or cosmetics, taking advantage of clients’ desire to live a healthier and longer life. However, due to the difficulty of discovering the in vivo functions of polyphenols, most of the research focuses on in vitro studies. In this review, we focused on the cardioprotective activity of different polyphenols as possible candidates for use in cardiovascular disease therapy and for improving the quality of life of patients. Thus, the studies, which were mainly based on endothelial cells, aortic cells, and some in vivo studies, were analyzed. Based on the reviewed articles, polyphenols have a few points of action, including inhibition of acetylcholinesterase, decrease in reactive oxygen species production and endothelial tube formation, stimulation of acetylcholine-induced endothelium-derived mediator release, and others, which lead to their cardio- and/or vasoprotective effects on endothelial cells. The obtained results suggest positive effects of polyphenols, but more long-term in vivo studies demonstrating effects on mechanism of action, sensitivity, and specificity or efficacy are needed before legal health claims can be made.


2004 ◽  
Vol 286 (6) ◽  
pp. G1059-G1068 ◽  
Author(s):  
Jan Heidemann ◽  
Hitoshi Ogawa ◽  
Parvaneh Rafiee ◽  
Norbert Lügering ◽  
Christian Maaser ◽  
...  

Mice genetically deficient in the chemokine receptor CXCR4 or its ligand stromal cell-derived factor (SDF)-1/CXCL12 die perinatally with marked defects in vascularization of the gastrointestinal tract. The aim of this study was to define the expression and angiogenic functions of microvascular CXCR4 and SDF-1/CXCL12 in the human intestinal tract. Studies of human colonic mucosa in vivo and primary cultures of human intestinal microvascular endothelial cells (HIMEC) in vitro showed that the intestinal microvasculature expresses CXCR4 and its cognate ligand SDF-1/CXCL12. Moreover, SDF-1/CXCL12 stimulation of HIMEC triggers CXCR4-linked G proteins, phosphorylates ERK1/2, and activates proliferative and chemotactic responses. Pharmacological studies indicate SDF-1/CXCL12 evokes HIMEC chemotaxis via activation of ERK1/2 and phosphoinositide 3-kinase signaling pathways. Consistent with chemotaxis and proliferation, endothelial tube formation was inhibited by neutralizing CXCR4 or SDF-1/CXCL12 antibodies, as well as the ERK1/2 inhibitor PD-98059. Taken together, these data demonstrate an important mechanistic role for CXCR4 and SDF-1/CXCL12 in regulating angiogenesis within the human intestinal mucosa.


2001 ◽  
Vol 152 (6) ◽  
pp. 1247-1254 ◽  
Author(s):  
Boris Troyanovsky ◽  
Tetyana Levchenko ◽  
Göran Månsson ◽  
Olga Matvijenko ◽  
Lars Holmgren

Angiostatin, a circulating inhibitor of angiogenesis, was identified by its ability to maintain dormancy of established metastases in vivo. In vitro, angiostatin inhibits endothelial cell migration, proliferation, and tube formation, and induces apoptosis in a cell type–specific manner. We have used a construct encoding the kringle domains 1–4 of angiostatin to screen a placenta yeast two-hybrid cDNA library for angiostatin-binding peptides. Here we report the identification of angiomotin, a novel protein that mediates angiostatin inhibition of migration and tube formation of endothelial cells. In vivo, angiomotin is expressed in the endothelial cells of capillaries as well as larger vessels of the human placenta. Upon expression of angiomotin in HeLa cells, angiomotin bound and internalized fluorescein-labeled angiostatin. Transfected angiomotin as well as endogenous angiomotin protein were localized to the leading edge of migrating endothelial cells. Expression of angiomotin in endothelial cells resulted in increased cell migration, suggesting a stimulatory role of angiomotin in cell motility. However, treatment with angiostatin inhibited migration and tube formation in angiomotin-expressing cells but not in control cells. These findings indicate that angiostatin inhibits cell migration by interfering with angiomotin activity in endothelial cells.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3934-3934
Author(s):  
Abdel Kareem Azab ◽  
Feda Azab ◽  
Phong Quang ◽  
Patricia Maiso ◽  
Hai T Ngo ◽  
...  

Abstract Abstract 3934 INTRODUCTION: The interaction of multiple myeloma (MM) cells with the bone marrow (BM) microenvironment plays a crucial role in MM pathogenesis. The BM microenvironment in MM is characterized by an increased micro-vessel density and increased secretion of angiogenic factors. CXCR7 is a G-protein coupled receptor shown to play a major role in the adhesion, migration and angiogenesis of endothelial cells (ECs). Our interest is in the role of CXCR7 in cell trafficking of ECs and EPCs in MM. Thus we characterized ECs and EPCs from MM patients and MM animal models and examined the contribution of CXCR7 to the cell trafficking using in vitro and in vivo assays and using CXCR7-selective compound. METHODS AND RESULTS: We used flow cytometry to detect the frequency of ECs and EPCs in the BM and peripheral blood (PB) of 5 MM patients and 5 normal subjects. ECs were detected as VEGFR+ CD133- cells, while EPCs were detected as VEGFR+ CD133+ cells. MM patients had significantly higher numbers of ECs and EPCs compared to healthy donors in both the BM and the PB. These results were confirmed in a mouse model of MM in which MM cells or vehicle were injected to SCID mice and the frequency of ECs and EPCs in the BM and the PB was determined 4 weeks after injection. We found that in mice with MM significantly higher numbers of ECs and EPCs could be detected in both the BM and the PB than in control mice. CXCR7 was expressed on both ECs and EPCs isolated from MM patients, healthy donors, and control mice. The expression of CXCR7 on EPCs was higher than the expression on ECs. The expression of CXCR7 on ECs and EPCs isolated from the BM was higher than the expression on ECs and EPCs isolated from the PB, respectively. Therefore, to test the role of CXCR7 in cell-trafficking of ECs and EPCs, we injected 10mg/kg of CXCR7 inhibitor POL6926, a potent and selective CXCR7 antagonist based on the Protein Epitope Mimetics (PEM) Technology (Polyphor, Switzerland), to BALB/c mice and tested the frequency of ECs and EPC in the PB and BM of the mice at 0, 2, 4 and 24 hours after the injection. We found a 3-fold increase in ECs and 6-fold increase in EPCs in the PB; 2 hrs post the injection of the CXCR7 antagonist. The levels of EPCs in the PB returned to baseline at 4 and 24 hrs, while the level of ECs was maintained at 4hrs and went back to baseline at 24 hrs. No significant differences were found in the frequency of ECs and EPCs in the BM after the injection of the CXCR7 antagonist. To investigate the function of CXCR7 in ECs in vitro we used human umbilical vein endothelial cells (HUVECs) as a model for ECs. CXCR7 was highly expressed on HUVECs. We could demonstrate that in vitro tube formation was promoted by either co-culture of MM cells or by conditioned medium from MM cell cultures. Furthermore, migration of HUVEC cells was facilitated by conditioned medium from MM cell cultures. These data suggest that MM cells may secrete factors promoting migration of endothelial cells and pro-angiogenic factors promoting angiogenesis. In addition, we could show that in vitro tube formation is inhibited by POL6926 suggesting that CXCR7 expression on HUVECs is required for tube formation. At the test concentrations POL6926 was not cytotoxic to HUVECs since cell proliferation was unaffected. CONCLUSION: We have shown that the level of ECs and EPCs was elevated in the PB and BM of MM patients compared to normal subjects, a finding which was confirmed in a MM mouse model in which CXCR7 was highly expressed on these cells. Injection of PEM CXCR7 antagonist increased the numbers of ECs and EPCs in the PB. These results suggest that CXCR7 may play a role in the cell-trafficking and recruitment of ECs and EPCs in MM. To investigate this hypothesis, using in vitro tube formation and migration assays, we have shown that MM cells secrete factors that promote migration and angiogenesis of HUVECs and the PEM CXCR7 antagonist inhibits these processes. In subsequent studies POL6926 will be tested in vivo in animal models of MM to determine the contribution of CXCR7 in EPC trafficking and its contribution to angiogenesis progression in MM. Disclosures: Zimmermann: Polyphor: Employment. Patel:Polyphor: Employment. Romagnoli:Polyphor: Employment. Roccaro:Roche:. Ghobrial:Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Research Funding; Noxxon: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding.


Author(s):  
Alessio Biagioni ◽  
Anna Laurenzana ◽  
Beatrice Menicacci ◽  
Silvia Peppicelli ◽  
Elena Andreucci ◽  
...  

AbstractExosomes (Exos) have been reported to promote pre-metastatic niche formation, proliferation, angiogenesis and metastasis. We have investigated the role of uPAR in melanoma cell lines-derived Exos and their pro-angiogenic effects on human microvascular endothelial cells (HMVECs) and endothelial colony-forming cells (ECFCs). Melanoma Exos were isolated from conditioned media of A375 and M6 cells by differential centrifugation and filtration. Tunable Resistive Pulse Sensing (TRPS) and Nanoparticle tracking analysis were performed to analyze dimension and concentration of Exos. The CRISPR–Cas 9 technology was exploited to obtain a robust uPAR knockout. uPAR is expressed in melanoma Exos that are internalized by HMVECs and ECFCs, enhancing VE-Cadherin, EGFR and uPAR expression in endothelial cells that undergo a complete angiogenic program, including proliferation, migration and tube formation. uPAR loss reduced the pro-angiogenic effects of melanoma Exos in vitro and in vivo by inhibition of VE-Cadherin, EGFR and uPAR expression and of ERK1,2 signaling in endothelial cells. A similar effect was obtained with a peptide that inhibits uPAR–EGFR interaction and with the EGFR inhibitor Gefitinib, which also inhibited melanoma Exos-dependent EGFR phosphorylation. This study suggests that uPAR is required for the pro-angiogenic activity of melanoma Exos. We propose the identification of uPAR-expressing Exos as a potentially useful biomarker for assessing pro-angiogenic propensity and eventually monitoring the response to treatment in metastatic melanoma patients.


Author(s):  
Shuang-Shuang Dong ◽  
Dan-Dan Dong ◽  
Zhang-Fu Yang ◽  
Gui-Qi Zhu ◽  
Dong-Mei Gao ◽  
...  

BackgroundAngiogenesis is a crucial process in tumorigenesis and development. The role of exosomes derived from hepatocellular carcinoma (HCC) cells in angiogenesis has not been clearly elucidated.Methods and ResultsExosomes were isolated from HCC cell lines (HCCLM3, MHCC97L, and PLC/RFP/5) by ultracentrifugation and identified by nano transmission electron microscopy (TEM), NanoSight analysis and western blotting, respectively. In vitro and in vivo analyses showed that exosomes isolated from highly metastatic HCC cells enhanced the migration, invasion and tube formation of human umbilical vein endothelial cells (HUVECs) compared to exosomes derived from poorly metastatic HCC cells. In addition, microarray analysis of HCC-Exos was conducted to identify potential functional molecules, and miR-3682-3p expression was found to be significantly downregulated in exosomes isolated from highly metastatic HCC cells. By in vitro gain-of-function experiments, we found that HCC cells secreted exosomal miR-3682-3p, which negatively regulates angiopoietin-1 (ANGPT1), and this led to inhibition of RAS-MEK1/2-ERK1/2 signaling in endothelial cells and eventually impaired angiogenesis.ConclusionOur study elucidates that exosomal miR-3682-3p attenuates angiogenesis by targeting ANGPT1 through RAS-MEK1/2-ERK1/2 signaling and provides novel potential targets for liver cancer therapy.


Biomolecules ◽  
2021 ◽  
Vol 11 (9) ◽  
pp. 1318
Author(s):  
Aleen Al Halawani ◽  
Lea Abdulkhalek ◽  
Suzanne M. Mithieux ◽  
Anthony S. Weiss

Tropoelastin, the soluble precursor of elastin, has been used for regenerative and wound healing purposes and noted for its ability to accelerate wound repair by enhancing vascularization at the site of implantation. However, it is not clear whether these effects are directly due to the interaction of tropoelastin with endothelial cells or communicated to endothelial cells following interactions between tropoelastin and neighboring cells, such as mesenchymal stem cells (MSCs). We adapted an endothelial tube formation assay to model in vivo vascularization with the goal of exploring the stimulatory mechanism of tropoelastin. In the presence of tropoelastin, endothelial cells formed less tubes, with reduced spreading into capillary-like networks. In contrast, conditioned media from MSCs that had been cultured on tropoelastin enhanced the formation of more dense, complex, and interconnected endothelial tube networks. This pro-angiogenic effect of tropoelastin is mediated indirectly through the action of tropoelastin on co-cultured cells. We conclude that tropoelastin inhibits endothelial tube formation, and that this effect is reversed by pro-angiogenic crosstalk from tropoelastin-treated MSCs. Furthermore, we find that the known in vivo pro-angiogenic effects of tropoelastin can be modeled in vitro, highlighting the value of tropoelastin as an indirect mediator of angiogenesis.


2010 ◽  
Vol 103 (01) ◽  
pp. 188-197 ◽  
Author(s):  
Aya Barzelay ◽  
Jeremy Ben-Shoshan ◽  
Michal Entin-Meer ◽  
Sofia Maysel-Auslender ◽  
Arnon Afek ◽  
...  

SummaryThe LIM-homeobox transcription factor islet-1 (Isl1) marks a cell population which gives rise to myocardial, pacemaker, endothelial and smooth muscle cells, which are derived from the secondary heart field during heart embryogenesis. Isl1+ precursors have the potential of self-renewal and differentiation into endothelial, cardiomyocyte and smooth muscle lineages. The primary objective of this study was to determine whether retroviral gene delivery of Isl1 to endothelial cells and mesenchymal stem cells (MSCs) could promote angiogenic and vasculogenic properties. To this end, endothelial cells and rat MSCs were retrovirally transduced to express Isl1. Isl1 expression in endothelial cells resulted in enhanced proliferation and adhesion to fibronectin. In addition, increased IL-1b and VEGF secretion was evident in Isl1 transduced endothelial cells, concomitant with increased migratory and tube formation properties of the endothelial cells. Isl1 expression in MSCs promoted their vasculogenic properties and resulted in enhanced in vitro tube formation. Finally, Isl1 expressing endothelial cells induced enhanced in vivo vascularisation in C57BL/6J mice. These data suggest, for the first time, that Isl1 promotes postnatal angiogenesis and vasculogenesis by improving the angiogenic properties of endothelial cells and MSCs.


Blood ◽  
2008 ◽  
Vol 111 (7) ◽  
pp. 3498-3506 ◽  
Author(s):  
Graeme M. Birdsey ◽  
Nicola H. Dryden ◽  
Valerie Amsellem ◽  
Frank Gebhardt ◽  
Kapil Sahnan ◽  
...  

Abstract Tight regulation of the balance between apoptosis and survival is essential in angiogenesis. The ETS transcription factor Erg is required for endothelial tube formation in vitro. Inhibition of Erg expression in human umbilical vein endothelial cells (HUVECs), using antisense oligonucleotides, resulted in detachment of cell-cell contacts and increased cell death. Inhibition of Erg expression by antisense in HUVECs also lowered expression of the adhesion molecule vascular endothelial (VE)–cadherin, a key regulator of endothelial intercellular junctions and survival. Using chromatin immunoprecipitation, we showed that Erg binds to the VE-cadherin promoter. Furthermore, Erg was found to enhance VE-cadherin promoter activity in a transactivation assay. Apoptosis induced by inhibition of Erg was partly rescued by overexpression of VE-cadherin–GFP, suggesting that VE-cadherin is involved in the Erg-dependent survival signals. To show the role of Erg in angiogenesis in vivo, we used siRNA against Erg in a Matrigel plug model. Erg inhibition resulted in a significant decrease in vascularization, with increase in caspase-positive endothelial cells (ECs). These results identify a new pathway regulating angiogenesis and endothelial survival, via the transcription factor Erg and the adhesion molecule VE-cadherin.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Cheng Wang ◽  
Xiaoyan Dai ◽  
Shengnan Wu ◽  
Wenjing Xu ◽  
Ping Song ◽  
...  

AbstractFUN14 domain-containing protein 1 (FUNDC1) is an integral mitochondrial outer-membrane protein, and mediates the formation of mitochondria-associated endoplasmic reticulum membranes (MAMs). This study aims to determine the contributions of FUNDC1-mediated MAMs to angiogenesis in vitro and in vivo. In cultured endothelial cells, VEGF significantly increases the formation of MAMs and MAM-related proteins, including FUNDC1. Endothelial cell-specific deletion of FUNDC1, which disrupts MAM formation in endothelial cells, lowers VEGFR2 expression and reduces tube formation, spheroid-sprouting, and functional blood vessel formation in vitro and in vivo. Conversely, increased MAM formation using MAM linkers mimics the effects of VEGF and promotes endothelial angiogenesis. Mechanistically, increased MAMs formation led to increased levels of Ca2+ in cytosol, promoted the phosphorylation of serum response factor (SRF) and enhanced the binding of SRF to VEGFR2 promoter, resulting in increased VEGFR2 production, with consequent angiogenesis. Moreover, blocking FUNDC1-related MAM formation with a cell-penetrating inhibitory peptide significantly suppresses the expressions of downstream angiogenic genes and inhibits tumor angiogenesis. We conclude that decreased MAMs formation by silencing FUNDC1 can inhibit angiogenesis by decreasing VEGFR2 expression, and targeting FUNDC1-dependent MAMs might be a promising approach for treating human disorders characterized by defective angiogenesis.


Sign in / Sign up

Export Citation Format

Share Document