Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 217
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38742432

ABSTRACT

Development of the vascular system is regulated by multiple signaling pathways mediated by receptor tyrosine kinases. Among them, angiopoietin (Ang)/Tie signaling regulates lymphatic and blood vessel development in mammals. Of the two Tie receptors, Tie2 is well known as a key mediator of Ang/Tie signaling, but, unexpectedly, recent studies have revealed that the Tie2 locus has been lost in many vertebrate species, whereas the Tie1 gene is more commonly present. However, Tie1-driven signaling pathways, including ligands and cellular functions, are not well understood. Here, we performed comprehensive mutant analyses of angiopoietins and Tie receptors in zebrafish and found that only angpt1 and tie1 mutants show defects in trunk lymphatic vessel development. Among zebrafish angiopoietins, only Angpt1 binds to Tie1 as a ligand. We indirectly monitored Ang1/Tie1 signaling and detected Tie1 activation in sprouting endothelial cells, where Tie1 inhibits nuclear import of EGFP-Foxo1a. Angpt1/Tie1 signaling functions in endothelial cell migration and proliferation, and in lymphatic specification during early lymphangiogenesis, at least in part by modulating Vegfc/Vegfr3 signaling. Thus, we show that Angpt1/Tie1 signaling constitutes an essential signaling pathway for lymphatic development in zebrafish.


Subject(s)
Angiopoietin-1 , Lymphangiogenesis , Lymphatic Vessels , Receptor, TIE-1 , Signal Transduction , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/embryology , Angiopoietin-1/metabolism , Angiopoietin-1/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-1/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Lymphangiogenesis/genetics , Cell Movement , Endothelial Cells/metabolism , Protein Binding , Cell Proliferation , Vascular Endothelial Growth Factor Receptor-3/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Mutation/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Gene Expression Regulation, Developmental
2.
Exp Cell Res ; 439(1): 114060, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38719173

ABSTRACT

BACKGROUND: Tie1 orphan receptor has become a focus of research, Tie1 can form a polymer with Tie2, regulate the Ang/Tie2 pathway and play a vital role in pathological angiogenesis and tumor progression, the function of Tie1 has remained uncertain in the progression of cervical cancer (CC). Here, we investigated the functional influences of Tie1 overexpress on CC in vitro and in vivo. METHODS: We used Immunohistochemistry (IHC) analysis to detect the relative expression of Tie1 in CC, and we analyzed its connection with the overall survival (OS) and progression free survival (PFS)of CC patients. To prove the role of Tie1 in cell proliferation and metastatic, Tie1 expression in CC cell lines was upregulated by lentivirus. RESULTS: The high expression of Tie1 in tumor cells of cervical cancer tissues is significantly correlated with FIGO stage, differentiated tumors, tumors with diameters, deep stromal invasion. We found that cell progression was promoted in Tie1-overexpress CC cell lines in vivo and in vitro. Tie1 potentially exerts a commanding influence on the expression of markers associated with epithelial-mesenchymal transition (EMT) and the PI3K/AKT signaling pathway. CONCLUSIONS: Our research indicates that Tie1 is highly connected to CC progression as it may play a role in the EMT process through the PI3K/AKT signaling pathway.


Subject(s)
Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Receptor, TIE-1 , Signal Transduction , Uterine Cervical Neoplasms , Animals , Female , Humans , Mice , Middle Aged , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-1/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
3.
Arterioscler Thromb Vasc Biol ; 43(8): e323-e338, 2023 08.
Article in English | MEDLINE | ID: mdl-37317851

ABSTRACT

BACKGROUND: Vascular growth followed by vessel specification is crucial for the establishment of a hierarchical blood vascular network. We have shown that TIE2 is required for vein development while little is known about its homologue TIE1 (tyrosine kinase with immunoglobulin-like and EGF [epithelial growth factor]-like domains 1) in this process. METHODS: We analyzed functions of TIE1 as well as its synergy with TIE2 in the regulation of vein formation by employing genetic mouse models targeting Tie1, Tek, and Nr2f2, together with in vitro cultured endothelial cells to decipher the underlying mechanism. RESULTS: Cardinal vein growth appeared normal in TIE1-deficient mice, whereas TIE2 deficiency altered the identity of cardinal vein endothelial cells with the aberrant expression of DLL4 (delta-like canonical Notch ligand 4). Interestingly, the growth of cutaneous veins, which was initiated at approximately embryonic day 13.5, was retarded in mice lack of TIE1. TIE1 deficiency disrupted the venous integrity, displaying increased sprouting angiogenesis and vascular bleeding. Abnormal venous sprouts with defective arteriovenous alignment were also observed in the mesenteries of Tie1-deleted mice. Mechanistically, TIE1 deficiency resulted in the decreased expression of venous regulators including TIE2 and COUP-TFII (chicken ovalbumin upstream promoter transcription factor, encoded by Nr2f2, nuclear receptor subfamily 2 group F member 2) while angiogenic regulators were upregulated. The alteration of TIE2 level by TIE1 insufficiency was further confirmed by the siRNA-mediated knockdown of Tie1 in cultured endothelial cells. Interestingly, TIE2 insufficiency also reduced the expression of TIE1. Combining the endothelial deletion of Tie1 with 1 null allele of Tek resulted in a progressive increase of vein-associated angiogenesis leading to the formation of vascular tufts in retinas, whereas the loss of Tie1 alone produced a relatively mild venous defect. Furthermore, the induced deletion of endothelial Nr2f2 decreased both TIE1 and TIE2. CONCLUSIONS: Findings from this study imply that TIE1 and TIE2, together with COUP-TFII, act in a synergistic manner to restrict sprouting angiogenesis during the development of venous system.


Subject(s)
Receptor, TIE-1 , Receptor, TIE-2 , Mice , Animals , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Endothelial Cells/metabolism , Signal Transduction , Veins
4.
J Transl Med ; 21(1): 134, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36814284

ABSTRACT

BACKGROUND: Radiotherapy resistance is the main cause of treatment failure in nasopharyngeal carcinoma (NPC), which leads to poor prognosis. It is urgent to elucidate the molecular mechanisms underlying radiotherapy resistance. METHODS: RNA-seq analysis was applied to five paired progressive disease (PD) and complete response (CR) NPC tissues. Loss-and gain-of-function assays were used for oncogenic function of FLI1 both in vitro and in vivo. RNA-seq analysis, ChIP assays and dual luciferase reporter assays were performed to explore the interaction between FLI1 and TIE1. Gene expression with clinical information from tissue microarray of NPC were analyzed for associations between FLI1/TIE1 expression and NPC prognosis. RESULTS: FLI1 is a potential radiosensitivity regulator which was dramatically overexpressed in the patients with PD to radiotherapy compared to those with CR. FLI1 induced radiotherapy resistance and enhanced the ability of DNA damage repair in vitro, and promoted radiotherapy resistance in vivo. Mechanistic investigations showed that FLI1 upregulated the transcription of TIE1 by binding to its promoter, thus activated the PI3K/AKT signaling pathway. A decrease in TIE1 expression restored radiosensitivity of NPC cells. Furthermore, NPC patients with high levels of FLI1 and TIE1 were correlated with poor prognosis. CONCLUSION: Our study has revealed that FLI1 regulates radiotherapy resistance of NPC through TIE1-mediated PI3K/AKT signaling pathway, suggesting that targeting the FLI1/TIE1 signaling pathway could be a potential therapeutic strategy to enhance the efficacy of radiotherapy in NPC.


Subject(s)
Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Proto-Oncogene Protein c-fli-1 , Radiation Tolerance , Receptor, TIE-1 , Humans , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/radiotherapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Proto-Oncogene Protein c-fli-1/genetics , Radiation Tolerance/genetics , Receptor, TIE-1/genetics
5.
Arterioscler Thromb Vasc Biol ; 42(3): 348-351, 2022 03.
Article in English | MEDLINE | ID: mdl-35021855

ABSTRACT

BACKGROUND: Schlemm's canal (SC) is a large vessel residing in the iridocorneal angle and is required to regulate aqueous humor outflow. Normal SC structure and function is indispensable for maintaining normal intraocular pressure, and elevated intraocular pressure is a risk factor for development of glaucoma. Recent reports have identified a key role of the angiopoietin-Tie2 pathway for SC development and function; however, the role of the orphan receptor Tie1 has not been clarified. METHODS: We used Tie1 knock out mice to study the function of Tie1 in SC development and function. Real-time quantitative polymerase chain reaction and Western blot analyses were used to verify Tie1 deletion. High-resolution microscopy of mouse SC whole mount and cross sections were used to study SC morphology. Measurement of intraocular pressure in live mice was used to study the impact of Tie1 on SC function. RESULTS: Tie1 is highly expressed in both human and mouse SC. Tie1 knock out mice display hypomorphic SC and elevated intraocular pressure as a result of attenuated SC development. CONCLUSIONS: Tie1 is indispensable for SC development and function, supporting it as a novel target for future SC-targeted glaucoma therapies and a candidate gene for glaucoma in humans.


Subject(s)
Anterior Chamber/enzymology , Anterior Chamber/growth & development , Endothelium, Corneal/enzymology , Receptor, TIE-1/metabolism , Animals , Aqueous Humor/physiology , Glaucoma/etiology , Humans , Intraocular Pressure/physiology , Lymphatic Vessels/abnormalities , Lymphatic Vessels/enzymology , Lymphatic Vessels/physiology , Mice , Mice, Knockout , Models, Animal , Receptor, TIE-1/deficiency , Receptor, TIE-1/genetics
6.
Cell Mol Life Sci ; 79(6): 312, 2022 May 23.
Article in English | MEDLINE | ID: mdl-35604495

ABSTRACT

The human signaling molecules Tie1 and Tie2 receptor tyrosine kinases (RTKs) play important pathophysiological roles in many diseases, including different cancers. The activity of Tie1 is mediated mainly through the downstream angiopoietin-1 (Ang1)-dependent activation of Tie2, rendering both Tie 1 and the Tie1/Tie2/Ang1 axis attractive putative targets for therapeutic intervention. However, the development of inhibitors that target Tie1 and an understanding of their effect on Tie2 and on the Tie1/Tie2/Ang1 axis remain unfulfilled tasks, due, largely, to the facts that Tie1 is an orphan receptor and is difficult to produce and use in the quantities required for immune antibody library screens. In a search for a selective inhibitor of this orphan receptor, we sought to exploit the advantages (e.g., small size that allows binding to hidden epitopes) of non-immune nanobodies and to simultaneously overcome their limitations (i.e., low expression and stability). We thus performed expression, stability, and affinity screens of yeast-surface-displayed naïve and predesigned synthetic (non-immune) nanobody libraries against the Tie1 extracellular domain. The screens yielded a nanobody with high expression and good affinity and specificity for Tie1, thereby yielding preferential binding for Tie1 over Tie2. The stability, selectivity, potency, and therapeutic potential of this synthetic nanobody were profiled using in vitro and cell-based assays. The nanobody triggered Tie1-dependent inhibition of RTK (Tie2, Akt, and Fak) phosphorylation and angiogenesis in endothelial cells, as well as suppression of human glioblastoma cell viability and migration. This study opens the way to developing nanobodies as therapeutics for different cancers associated with Tie1 activation.


Subject(s)
Neoplasms , Single-Domain Antibodies , Angiopoietin-1 , Endothelial Cells/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation , Receptor, TIE-1/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Single-Domain Antibodies/pharmacology
7.
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
8.
Dev Biol ; 455(1): 73-84, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31319059

ABSTRACT

The mechanisms regulating endothelial cell response to hemodynamic forces required for heart valve development, especially valve remodeling, remain elusive. Tie1, an endothelial specific receptor tyrosine kinase, is up-regulated by oscillating shear stress and is required for lymphatic valve development. In this study, we demonstrate that valvular endothelial Tie1 is differentially expressed in a dynamic pattern predicted by disturbed flow during valve remodeling. Following valvular endocardial specific deletion of Tie1 in mice, we observed enlarged aortic valve leaflets, decreased valve stiffness and valvular insufficiency. Valve abnormalities were only detected in late gestation and early postnatal mutant animals and worsened with age. The mutant mice developed perturbed extracellular matrix (ECM) deposition and remodeling characterized by increased glycosaminoglycan and decreased collagen content, as well as increased valve interstitial cell expression of Sox9, a transcription factor essential for normal ECM maturation during heart valve development. This study provides the first evidence that Tie1 is involved in modulation of late valve remodeling and suggests that an important Tie1-Sox9 signaling axis exists through which disturbed flows are converted by endocardial cells to paracrine Sox9 signals to modulate normal matrix remodeling of the aortic valve.


Subject(s)
Aortic Valve/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Organogenesis/genetics , Receptor, TIE-1/genetics , Animals , Aortic Valve/embryology , Aortic Valve/growth & development , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Female , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptor, TIE-1/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Vascular Remodeling/genetics
9.
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
10.
BMC Cancer ; 19(1): 732, 2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31340773

ABSTRACT

BACKGROUND: Angiopoietin growth factors (Angs) regulate angiogenesis and lymphangiogenesis by binding to the endothelial Tie2 receptor. Ang2 expression is elevated in tissue hypoxia and inflammation, which also induce cleavage of the extracellular domain of the orphan Tie1 receptor. Here we have examined if the concentrations of Ang2 and the soluble extracellular domain of Tie1 in patient plasma are associated with the prognosis of patients with metastatic breast cancer. METHODS: Plasma Tie1 and Ang2 levels were measured in metastatic breast cancer patients treated in a phase II trial with a taxane-bevacizumab combination chemotherapy in the first-line treatment setting. They were analyzed before treatment, after 6 weeks and 6 months of treatment, and at the final study visit. Using the median concentrations as cutoffs, Tie1 and Ang2 data were dichotomized into low and high concentration groups. Additionally, we analyzed Tie1 concentrations in plasma from 10 healthy women participating in a breast cancer primary prevention study. RESULTS: Plasma samples were available from 58 (89%) of the 65 patients treated in the trial. The baseline Tie1 levels of the healthy controls were significantly lower than those of the metastatic patients (p < 0.001). The overall survival of the patients with a high baseline Tie1 level was significantly shorter (multivariate HR 3.07, 95% CI 1.39-6.79, p = 0.005). Additionally, the progression-free survival was shorter for patients with a high baseline Tie1 level (multivariate HR 3.78, 95% CI 1.57-9.09, p = 0.003). In contrast, the baseline Ang2 levels had no prognostic impact in a multivariate Cox proportional hazard regression analysis. The combined analysis of baseline Tie1 and Ang2 levels revealed that patients with both high Tie1 and high Ang2 baseline levels had a significantly shorter overall survival than the patients with low baseline levels of both markers (multivariate HR for overall survival 4.32, 95% CI 1.44-12.94, p = 0.009). CONCLUSIONS: This is the first study to demonstrate the prognostic value of baseline Tie1 plasma concentration in patients with metastatic breast cancer. Combined with the results of the Ang2 analyses, the patients with both high Tie1 and Ang2 levels before treatment had the poorest survival. TRIAL REGISTRATION: Clinicaltrials.gov: NCT00979641, registration date 19-DEC-2008. The regional Ethics Committee: R08142M, registration date 18-NOV-2008.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biomarkers, Tumor/blood , Breast Neoplasms/drug therapy , Receptor, TIE-1/metabolism , Adult , Aged , Angiopoietin-2/blood , Bevacizumab/administration & dosage , Breast/pathology , Breast Neoplasms/blood , Breast Neoplasms/mortality , Docetaxel/administration & dosage , Drug Administration Schedule , Female , Humans , Middle Aged , Prognosis , Progression-Free Survival , Prospective Studies , Survival Analysis
11.
Circ Res ; 120(8): 1276-1288, 2017 Apr 14.
Article in English | MEDLINE | ID: mdl-28179430

ABSTRACT

RATIONALE: Lymphatic vasculature constitutes a second vascular system essential for immune surveillance and tissue fluid homeostasis. Maturation of the hierarchical vascular structure, with a highly branched network of capillaries and ducts, is crucial for its function. Environmental cues mediate the remodeling process, but the mechanism that underlies this process is largely unknown. OBJECTIVE: Polydom (also called Svep1) is an extracellular matrix protein identified as a high-affinity ligand for integrin α9ß1. However, its physiological function is unclear. Here, we investigated the role of Polydom in lymphatic development. METHODS AND RESULTS: We generated Polydom-deficient mice. Polydom-/- mice showed severe edema and died immediately after birth because of respiratory failure. We found that although a primitive lymphatic plexus was formed, it failed to undergo remodeling in Polydom-/- embryos, including sprouting of new capillaries and formation of collecting lymphatic vessels. Impaired lymphatic development was also observed after knockdown/knockout of polydom in zebrafish. Polydom was deposited around lymphatic vessels, but secreted from surrounding mesenchymal cells. Expression of Foxc2 (forkhead box protein c2), a transcription factor involved in lymphatic remodeling, was decreased in Polydom-/- mice. Polydom bound to the lymphangiogenic factor Ang-2 (angiopoietin-2), which was found to upregulate Foxc2 expression in cultured lymphatic endothelial cells. Expressions of Tie1/Tie2 receptors for angiopoietins were also decreased in Polydom-/- mice. CONCLUSIONS: Polydom affects remodeling of lymphatic vessels in both mouse and zebrafish. Polydom deposited around lymphatic vessels seems to ensure Foxc2 upregulation in lymphatic endothelial cells, possibly via the Ang-2 and Tie1/Tie2 receptor system.


Subject(s)
Endothelial Cells/metabolism , Lymphangiogenesis , Lymphatic Vessels/metabolism , Proteins/metabolism , Angiopoietin-2/metabolism , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules , Cell Communication , Cells, Cultured , Edema/genetics , Edema/metabolism , Edema/physiopathology , Endothelial Cells/pathology , Endothelium, Lymphatic/abnormalities , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/physiopathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Genotype , Humans , Lymphatic Vessels/abnormalities , Lymphatic Vessels/physiopathology , Mesoderm/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Binding , Proteins/genetics , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Signal Transduction , Thoracic Duct/abnormalities , Thoracic Duct/metabolism , Thoracic Duct/physiopathology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Arterioscler Thromb Vasc Biol ; 38(7): 1562-1575, 2018 07.
Article in English | MEDLINE | ID: mdl-29724820

ABSTRACT

OBJECTIVE: Tie1 (tyrosine kinase containing immunoglobulin and epidermal growth factor homology 1), an endothelial and hematopoietic cell-specific receptor tyrosine kinase, is an important regulator of angiogenesis and critical for maintaining vascular integrity. The post-transcriptional regulation of tie1 mRNA expression is not understood, but it might partly explain Tie1's differential expression pattern in endothelium. Following up on our previous work that identified natural antisense transcripts from the tie1 locus-tie1 antisense (tie1AS), which regulates tie1 mRNA levels in zebrafish-we attempted to identify the mechanism of this regulation. APPROACH AND RESULTS: Through in vitro and in vivo ribonucleoprotein binding studies, we demonstrated that tie1AS long noncoding RNA interacts with an RNA binding protein-embryonic lethal and abnormal vision Drosophila-like 1 (Elavl1)-that regulates tie1 mRNA levels. When we disrupted the interaction between tie1AS and Elavl1 by using constitutively active antisense morpholino oligonucleotides or photoactivatable morpholino oligonucleotides, tie1 mRNA levels increased between 26 and 31 hours post-fertilization, particularly in the head. This increase correlated with dilation of primordial midbrain channels, smaller eyes, and reduced ventricular space. We also observed these phenotypes when we used CRISPR (clustered regularly interspaced short palindromic repeats)-mediated CRISPRi (CRISPR-mediated interference) to knock down tie1AS. Treatment of the morpholino oligonucleotide-injected embryos with a small molecule that decreased tie1 mRNA levels rescued all 3 abnormal phenotypes. CONCLUSIONS: We identified a novel mode of temporal and spatial post-transcriptional regulation of tie1 mRNA. It involves long noncoding RNA, tie1AS, and Elavl1 (an interactor of tie1AS).


Subject(s)
Blood Vessels/enzymology , Brain/blood supply , Neovascularization, Physiologic/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Blood Vessels/embryology , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Time Factors , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
13.
Angiogenesis ; 21(4): 823-836, 2018 11.
Article in English | MEDLINE | ID: mdl-29951828

ABSTRACT

Tip cells, the leading cells of angiogenic sprouts, were identified in cultures of human umbilical vein endothelial cells (HUVECs) by using CD34 as a marker. Here, we show that tip cells are also present in primary human microvascular endothelial cells (hMVECs), a more relevant endothelial cell type for angiogenesis. By means of flow cytometry, immunocytochemistry, and qPCR, it is shown that endothelial cell cultures contain a dynamic population of CD34+ cells with many hallmarks of tip cells, including filopodia-like extensions, elevated mRNA levels of known tip cell genes, and responsiveness to stimulation with VEGF and inhibition by DLL4. Furthermore, we demonstrate that our in vitro tip cell model can be exploited to investigate cellular and molecular mechanisms in tip cells and to discover novel targets for anti-angiogenesis therapy in patients. Small interfering RNA (siRNA) was used to knockdown gene expression of the known tip cell genes angiopoietin 2 (ANGPT2) and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), which resulted in similar effects on tip cells and sprouting as compared to inhibition of tip cells in vivo. Finally, we identified two novel tip cell-specific genes in CD34+ tip cells in vitro: insulin-like growth factor 2 (IGF2) and IGF-1-receptor (IGF1R). Knockdown of these genes resulted in a significant decrease in the fraction of tip cells and in the extent of sprouting in vitro and in vivo. In conclusion, this study shows that by using our in vitro tip cell model, two novel essential tip cells genes are identified.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Insulin-Like Growth Factor II/metabolism , Microvessels/metabolism , Receptors, Somatomedin/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Chick Embryo , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/cytology , Humans , Insulin-Like Growth Factor II/genetics , Microvessels/cytology , Receptor, IGF Type 1 , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Receptors, Somatomedin/genetics , Zebrafish
14.
Mol Cell ; 37(5): 643-55, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20227369

ABSTRACT

The Tie family of endothelial-specific receptor tyrosine kinases is essential for cell proliferation, migration, and survival during angiogenesis. Despite considerable similarity, experiments with Tie1- or Tie2-deficient mice highlight distinct functions for these receptors in vivo. The Tie2 receptor is further unique with respect to its structurally homologous ligands. Angiopoietin-2 and -3 can function as agonists or antagonists; angiopoietin-1 and -4 are constitutive agonists. To address the role of Tie1 in angiopoietin-mediated Tie2 signaling and determine the basis for the behavior of the individual angiopoietins, we used an in vivo FRET-based proximity assay to monitor Tie1 and -2 localization and association. We provide evidence for Tie1-Tie2 complex formation on the cell surface and identify molecular surface areas essential for receptor-receptor recognition. We further demonstrate that the Tie1-Tie2 interactions are dynamic, inhibitory, and differentially modulated by angiopoietin-1 and -2. Based on the available data, we propose a unified model for angiopoietin-induced Tie2 signaling.


Subject(s)
Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Endothelial Cells/enzymology , Receptor, TIE-1/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , Cell Line , Cell Membrane/enzymology , Fluorescence Resonance Energy Transfer , Humans , Ligands , Models, Molecular , Mutation , Protein Conformation , Protein Multimerization , Protein Structure, Tertiary , RNA Interference , Receptor Cross-Talk , Receptor, TIE-1/chemistry , Receptor, TIE-1/genetics , Receptor, TIE-2/chemistry , Receptor, TIE-2/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Time Factors , Transfection
15.
Microvasc Res ; 114: 101-113, 2017 11.
Article in English | MEDLINE | ID: mdl-28732797

ABSTRACT

Diabetes mellitus type 2 predisposes patients to various microvascular complications. In the current experiment, the potent role of diabetes mellitus was investigated on the content of VEGFR-1, -2, Tie-1 and -2, and Akt in human endothelial progenitor cells. The gene expression profile of mTOR and Hedgehog signaling pathways were measured by PCR array. The possible crosstalk between RTKs, mTOR and Hedgehog signaling was also studied by bioinformatic analysis. Endothelial progenitor cells were incubated with serum from normal and diabetic for 7days. Compared to non-treated cells, diabetic serum-induced cell apoptosis (~2-fold) and prohibited cell migration toward bFGF (p<0.001). ELISA analysis showed that diabetes exposed cells had increased abundance of Tie-1, -2 and VEGFR-2 and reduced amount of VEGFR-1 (p<0.0001) in diabetic cells. Western blotting showed a marked reduction in the protein level of Akt after cells exposure to serum from diabetic subjects (p<0.0001). PCR array revealed a significant stimulation of both mTOR and Hedgehog signaling pathways in diabetic cells (p<0.05). According to data from bioinformatic datasets, we showed VEGFR-1, -2 and Tie-2, but not Tie-1, are master regulators of angiogenesis. There is a crosstalk between RTKs and mTOR signaling by involving P62, GABARAPL1, and HTT genes. It seems that physical interaction and co-expression of Akt decreased the level of VEGFR-1 in diabetic cells. Regarding data from the present experiment, diabetic serum contributed to uncontrolled induction of both mTOR and Hedgehog signaling in endothelial progenitor cells. Diabetes mellitus induces mTOR pathway by involving receptor tyrosine kinases while Hedgehog stimulation is independent of these receptors.


Subject(s)
Diabetes Mellitus, Type 2/blood , Endothelial Progenitor Cells/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Adult , Apoptosis , Case-Control Studies , Cell Movement , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Endothelial Progenitor Cells/pathology , Gene Expression Regulation , Hedgehog Proteins/metabolism , Humans , Male , Neovascularization, Physiologic , Proto-Oncogene Proteins c-akt/genetics , Receptor Cross-Talk , Receptor, TIE-1/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Time Factors , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Clin Sci (Lond) ; 131(1): 87-103, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27941161

ABSTRACT

Endothelial cells that form the inner layer of blood and lymphatic vessels are important regulators of vascular functions and centrally involved in the pathogenesis of vascular diseases. In addition to the vascular endothelial growth factor (VEGF) receptor pathway, the angiopoietin (Ang)-Tie system is a second endothelial cell specific ligand-receptor signalling system necessary for embryonic cardiovascular and lymphatic development. The Ang-Tie system also regulates postnatal angiogenesis, vessel remodelling, vascular permeability and inflammation to maintain vascular homoeostasis in adult physiology. This system is implicated in numerous diseases where the vasculature has an important contribution, such as cancer, sepsis, diabetes, atherosclerosis and ocular diseases. Furthermore, mutations in the TIE2 signalling pathway cause defects in vascular morphogenesis, resulting in venous malformations and primary congenital glaucoma. Here, we review recent advances in the understanding of the Ang-Tie signalling system, including cross-talk with the vascular endothelial protein tyrosine phosphatase (VE-PTP) and the integrin cell adhesion receptors, focusing on the Ang-Tie system in vascular development and pathogenesis of vascular diseases.


Subject(s)
Angiopoietins/metabolism , Cardiovascular System/metabolism , Lymphatic System/metabolism , Receptor, TIE-1/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , Angiopoietins/genetics , Animals , Cardiovascular System/enzymology , Cardiovascular System/growth & development , Humans , Lymphatic System/enzymology , Lymphatic System/growth & development , Receptor, TIE-1/genetics , Receptor, TIE-2/genetics
17.
Dev Biol ; 399(1): 117-128, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25576926

ABSTRACT

Tie1 is a receptor tyrosine kinase with broad expression in embryonic endothelium. Reduction of Tie1 levels in mouse embryos with a hypomorphic Tie1 allele resulted in abnormal lymphatic patterning and architecture, decreased lymphatic draining efficiency, and ultimately, embryonic demise. Here we report that Tie1 is present uniformly throughout the lymphatics and from late embryonic/early postnatal stages, becomes more restricted to lymphatic valve regions. To investigate later events of lymphatic development, we employed Cre-loxP recombination utilizing a floxed Tie1 allele and an Nfatc1Cre line, to provide loxP excision predominantly in lymphatic endothelium and developing valves. Interestingly, unlike the early prenatal defects previously described by ubiquitous endothelial deletion, excision of Tie1 with Nfatc1Cre resulted in abnormal lymphatic defects in postnatal mice and was characterized by agenesis of lymphatic valves and a deficiency of collecting lymphatic vessels. Attenuation of Tie1 signaling in lymphatic endothelium prevented initiation of lymphatic valve specification by Prox1 high expression lymphatic endothelial cells that is associated with the onset of turbulent flow in the lymphatic circulation. Our findings reveal a fundamental role for Tie1 signaling during lymphatic vessel remodeling and valve morphogenesis and implicate it as a candidate gene involved in primary lymphedema.


Subject(s)
Embryo, Mammalian/metabolism , Lymphatic System/metabolism , Lymphatic Vessels/metabolism , Receptor, TIE-1/metabolism , Animals , Animals, Newborn , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Immunohistochemistry , Lymphangiogenesis/genetics , Lymphatic System/embryology , Lymphatic Vessels/embryology , Mice, Inbred C57BL , Mice, Knockout , Receptor, TIE-1/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Proc Natl Acad Sci U S A ; 110(18): 7205-10, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23592718

ABSTRACT

Angiogenesis is a complex cellular process involving multiple regulatory growth factors and growth factor receptors. Among them, the ligands for the endothelial-specific tunica intima endothelial receptor tyrosine kinase 2 (Tie2) receptor kinase, angiopoietin-1 (Ang1) and Ang2, play essential roles in balancing vessel stability and regression during both developmental and tumor-induced angiogenesis. Despite possessing a high degree of sequence identity, Ang1 and Ang2 have distinct functional roles and cell-signaling characteristics. Here, we present the crystal structures of Ang1 both unbound and in complex with the Tie2 ectodomain. Comparison of the Ang1-containing structures with their Ang2-containing counterparts provide insight into the mechanism of receptor activation and reveal molecular surfaces important for interactions with Tie2 coreceptors and associated signaling proteins. Using structure-based mutagenesis, we identify a loop within the angiopoietin P domain, adjacent to the receptor-binding interface, which confers the specific agonist/antagonist properties of the molecule. We demonstrate using cell-based assays that an Ang2 chimera containing the Ang1 loop sequence behaves functionally similarly to Ang1 as a constitutive Tie2 agonist, able to efficiently dissociate the inhibitory Tie1/Tie2 complex and elicit Tie2 clustering and downstream signaling.


Subject(s)
Angiopoietin-1/chemistry , Angiopoietin-1/metabolism , Signal Transduction , Angiopoietin-2/chemistry , Angiopoietin-2/metabolism , Conserved Sequence , Crystallography, X-Ray , HEK293 Cells , Humans , Models, Molecular , Protein Structure, Tertiary , Receptor, TIE-1/chemistry , Receptor, TIE-1/metabolism , Receptor, TIE-2/chemistry , Receptor, TIE-2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Static Electricity , Structure-Activity Relationship
19.
Lancet Oncol ; 16(8): 937-48, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26184520

ABSTRACT

BACKGROUND: Tumour mutational status is an important determinant of the response of metastatic colorectal cancer to targeted treatments. However, the genotype of the tissue obtained at the time of diagnosis might not accurately represent tumour genotype after multiple lines of treatment. This retrospective exploratory analysis investigated the clinical activity of regorafenib in biomarker subgroups of the CORRECT study population defined by tumour mutational status or plasma protein levels. METHODS: We used BEAMing technology to identify KRAS, PIK3CA, and BRAF mutations in DNA obtained from the plasma of 503 patients with metastatic colorectal cancer who enrolled in the CORRECT trial. We quantified total human genomic DNA isolated from plasma samples for 503 patients using a modified version of human long interspersed nuclear element-1 (LINE-1) quantitive real-time PCR. We also measured the concentration of 15 proteins of interest-angiopoietin 2, interleukin 6, interleukin 8, placental growth factor, soluble TIE-1, soluble VEGFR1, VEGF-A, VEGF-C, VEGF-D, VEGF-A isoform 121, bone morphogenetic protein 7, macrophage colony-stimulating factor, stromal cell-derived factor-1, tissue inhibitor of metalloproteinase 2, and von Willebrand factor-in plasma samples from 611 patients. We did correlative analyses of overall survival and progression-free survival in patient subgroups based on mutational status, circulating DNA concentration, and protein concentrations. The CORRECT trial was registered with ClinicalTrials.gov, number NCT01103323. FINDINGS: Tumour-associated mutations were readily detected with BEAMing of plasma DNA, with KRAS mutations identified in 349 (69%) of 503 patients, PIK3CA mutations in 84 (17%) of 503 patients, and BRAF mutations in 17 (3%) of 502 patients. We did not do correlative analysis based on BRAF genotype because of the low mutational frequency detected for this gene. Some of the most prevalent individual hot-spot mutations we identified included: KRAS (KRAS G12D, 116 [28%] of 413 mutations; G12V, 72 [17%]; and G13D, 67 [16%]) and PIK3CA (PIK3CA E542K, 27 [30%] of 89 mutations; E545K, 37 [42%]; and H1047R, 12 [14%]). 41 (48%) of 86 patients who had received anti-EGFR therapy and whose archival tumour tissue DNA was KRAS wild-type in BEAMing analysis were identified as having KRAS mutations in BEAMing analysis of fresh plasma DNA. Correlative analyses suggest a clinical benefit favouring regorafenib across patient subgroups defined by KRAS and PIK3CA mutational status (progression-free survival with regorafenib vs placebo: hazard ratio [HR] 0·52, 95% CI 0·35-0·76 for KRAS wild-type; HR 0·51, 95% CI 0·40-0·65 for KRAS mutant [KRAS wild type vs mutant, pinteraction=0·74]; HR 0·50, 95% CI 0·40-0·63 for PIK3CA wild-type; HR 0·54, 95% CI 0·32-0·89 for PIK3CA mutant [PIK3CA wild-type vs mutant, pinteraction=0·85]) or circulating DNA concentration (progression-free survival with regorafenib vs placebo: HR 0·53, 95% CI 0·40-0·71, for low circulating DNA concentrations; HR 0·52, 95% CI 0·40-0·70, for high circulating DNA concentrations; low vs high circulating DNA, pinteraction=0·601). With the exception of von Willebrand factor, assessed with the median cutoff method, plasma protein concentrations were also not associated with regorafenib activity in terms of progression-free survival. In univariable analyses, the only plasma protein that was associated with overall survival was TIE-1, high concentrations of which were associated with longer overall survival compared with low TIE-1 concentrations. This association was not significant in multivariable analyses. INTERPRETATION: BEAMing of circulating DNA could be a viable approach for non-invasive analysis of tumour genotype in real time and for the identification of potentially clinically relevant mutations that are not detected in archival tissue. Additionally, the results show that regorafenib seems to be consistently associated with a clinical benefit in a range of patient subgroups based on mutational status and protein biomarker concentrations. FUNDING: Bayer HealthCare Pharmaceuticals.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Phenylurea Compounds/therapeutic use , Pyridines/therapeutic use , Receptor, TIE-1/blood , Adenocarcinoma/blood , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Aged , Class I Phosphatidylinositol 3-Kinases , Clinical Trials, Phase III as Topic , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Disease Progression , Disease-Free Survival , Female , Genetic Predisposition to Disease , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Patient Selection , Phenotype , Phosphatidylinositol 3-Kinases/blood , Phosphatidylinositol 3-Kinases/genetics , Precision Medicine , Predictive Value of Tests , Proportional Hazards Models , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/blood , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Randomized Controlled Trials as Topic , Real-Time Polymerase Chain Reaction , Retrospective Studies , Time Factors , Treatment Outcome , ras Proteins/blood , ras Proteins/genetics
20.
Microvasc Res ; 101: 38-47, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26092680

ABSTRACT

Pericytes are perivascular support cells, the origin of which in tumor tissue is not clear. Recently, we identified a Tie1(+) precursor cell that differentiates into vascular smooth muscle, in a Notch-dependent manner. To understand the involvement of Notch in the ontogeny of tumor pericytes we used a novel flow immunophenotyping strategy to define CD146(+)/CD45(-)/CD31(-/lo) pericytes in the tumor stroma. This strategy combined with ex vivo co-culture experiments identified a novel pericyte progenitor cell population defined as Sca1(hi)/CD146(-)/CD45(-)/CD31(-). The differentiation of these progenitor cells was stimulated by co-culture with endothelial cells. Overexpression of the Notch ligand Jagged1 in endothelial cells further stimulated the differentiation of Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells into pericytes, while inhibition of Notch signaling with a γ-secretase inhibitor reduced this differentiation. However, Notch inhibition specifically in Tie1-expressing cells did not change the abundance of pericytes in tumors, suggesting that the pericyte precursor is distinct from the vascular smooth muscle cell precursor. Transplant experiments showed that the bone marrow contributes minimally to tumor pericytes. Immunophenotyping revealed that Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells have greater potential to differentiate into pericytes and have increased expression of classic mesenchymal stem cell markers (CD13, CD44, Nt5e and Thy-1) compared to Sca1(-/lo)/CD146(-)/CD45(-)/CD31(-) cells. Our results suggest that a local Sca1(hi)/CD146(-)/CD45(-)/CD31(-) pericyte progenitor resides in the tumor microenvironment and requires Notch signaling for differentiation into mature pericytes.


Subject(s)
Neoplasms/metabolism , Pericytes/cytology , Receptors, Notch/metabolism , Animals , Ataxin-1/metabolism , Bone Marrow Transplantation , CD146 Antigen/metabolism , Carcinoma, Lewis Lung , Cell Differentiation , Coculture Techniques , Flow Cytometry , Human Umbilical Vein Endothelial Cells , Humans , Leukocyte Common Antigens/metabolism , Melanoma, Experimental , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptor, TIE-1/metabolism , Signal Transduction , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL