Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747287

ABSTRACT

Lymphedema is a debilitating disease with no effective cure and affects an estimated 250 million individuals worldwide. Prior studies have identified mutations in piezo-type mechanosensitive ion channel component 1 (PIEZO1), angiopoietin 2 (ANGPT2), and tyrosine kinase with Ig-like and EGF-like domains 1 (TIE1) in patients with primary lymphedema. Here, we identified crosstalk between these molecules and showed that activation of the mechanosensory channel PIEZO1 in lymphatic endothelial cells (LECs) caused rapid exocytosis of the TIE ligand ANGPT2, ectodomain shedding of TIE1 by disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), and increased TIE/PI3K/AKT signaling, followed by nuclear export of the transcription factor FOXO1. These data establish a functional network between lymphedema-associated genes and provide what we believe to be the first molecular mechanism bridging channel function with vascular signaling and intracellular events culminating in transcriptional regulation of genes expressed in LECs. Our study provides insights into the regulation of lymphatic function and molecular pathways involved in human disease.


Subject(s)
Angiopoietin-2 , Forkhead Box Protein O1 , Ion Channels , Lymphangiogenesis , Lymphedema , Receptor, TIE-1 , Signal Transduction , Ion Channels/metabolism , Ion Channels/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Humans , Animals , Angiopoietin-2/metabolism , Angiopoietin-2/genetics , Lymphedema/metabolism , Lymphedema/genetics , Lymphedema/pathology , Mice , Lymphangiogenesis/genetics , Receptor, TIE-1/metabolism , Receptor, TIE-1/genetics , Endothelial Cells/metabolism , Mechanotransduction, Cellular , ADAM17 Protein/metabolism , ADAM17 Protein/genetics
2.
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
3.
J Cell Biol ; 222(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37338522

ABSTRACT

Polydom is an extracellular matrix protein involved in lymphatic vessel development. Polydom-deficient mice die immediately after birth due to defects in lymphatic vessel remodeling, but the mechanism involved is poorly understood. Here, we report that Polydom directly binds to Tie1, an orphan receptor in the Angiopoietin-Tie axis, and facilitates migration of lymphatic endothelial cells (LECs) in a Tie1-dependent manner. Polydom-induced LEC migration is diminished by PI3K inhibitors but not by an ERK inhibitor, suggesting that the PI3K/Akt signaling pathway is involved in Polydom-induced LEC migration. In line with this possibility, Akt phosphorylation in LECs is enhanced by Polydom although no significant Tie1 phosphorylation is induced by Polydom. LECs also exhibited nuclear exclusion of Foxo1, a signaling event downstream of Akt activation, which was impaired in Polydom-deficient mice. These findings indicate that Polydom is a physiological ligand for Tie1 and participates in lymphatic vessel development through activation of the PI3K/Akt pathway.


Subject(s)
Calcium-Binding Proteins , Endothelial Cells , Lymphatic Vessels , Receptor, TIE-1 , Animals , Mice , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Movement
4.
Elife ; 122023 04 25.
Article in English | MEDLINE | ID: mdl-37097004

ABSTRACT

Multiple factors are required to form functional lymphatic vessels. Here, we uncover an essential role for the secreted protein Svep1 and the transmembrane receptor Tie1 during the development of subpopulations of the zebrafish facial lymphatic network. This specific aspect of the facial network forms independently of Vascular endothelial growth factor C (Vegfc) signalling, which otherwise is the most prominent signalling axis in all other lymphatic beds. Additionally, we find that multiple specific and newly uncovered phenotypic hallmarks of svep1 mutants are also present in tie1, but not in tie2 or vegfc mutants. These phenotypes are observed in the lymphatic vasculature of both head and trunk, as well as in the development of the dorsal longitudinal anastomotic vessel under reduced flow conditions. Therefore, our study demonstrates an important function for Tie1 signalling during lymphangiogenesis as well as blood vessel development in zebrafish. Furthermore, we show genetic interaction between svep1 and tie1 in vivo, during early steps of lymphangiogenesis, and demonstrate that zebrafish as well as human Svep1/SVEP1 protein bind to the respective Tie1/TIE1 receptors in vitro. Since compound heterozygous mutations for SVEP1 and TIE2 have recently been reported in human glaucoma patients, our data have clinical relevance in demonstrating a role for SVEP1 in TIE signalling in an in vivo setting.


Subject(s)
Lymphatic Vessels , Zebrafish , Animals , Humans , Zebrafish/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Ligands , Lymphatic Vessels/metabolism , Lymphangiogenesis/genetics , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Cell Adhesion Molecules/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism
5.
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
6.
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
7.
Bioengineered ; 12(1): 6617-6628, 2021 12.
Article in English | MEDLINE | ID: mdl-34516352

ABSTRACT

Preeclampsia (PE) is a huge threat to pregnant women. Our previous study demonstrated that long non-coding RNA (lncRNA) NR_002794 was highly expressed in placentas of PE patients and could regulate the phenotypes of trophoblast cells. However, the downstream regulatory mechanisms of NR_002794 remain unknown. In this text, some potential downstream targets or signaling pathways of NR_002794 were identified through RNA sequencing (RNA-seq) and bioinformatics analysis in SWAN71 trophoblast cells. Western blot assay demonstrated that NR_002794 inactivated protein kinase B (AKT) and extracellular signal-regulated kinase 1/2 (ERK1/2) pathways and activated cell apoptotic signaling in SWAN71 cells. Both RNA-seq and reverse transcription-quantitative PCR (RT-qPCR) outcomes showed that NR_002794 up-regulation could notably inhibit the expression of C-C motif chemokine ligand 4 like 2 (CCL4L2), interleukin 15 receptor subunit alpha (IL15RA), interleukin 32 (IL32), and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), while NR_002794 knockdown induced these gene expressions in SWAN71 cells. CCK-8, BrdU, Transwell, wound healing, and flow cytometry analyses showed that NR_002794 inhibited cell proliferation and migration and induced cell apoptosis through down-regulating TIE1 in SWAN71 cells. In conclusion, lncRNA NR_002794 could exert its functions by regulating AKT and ERK1/2 pathways and TIE1 expression in human trophoblast cells.


Subject(s)
MAP Kinase Signaling System/genetics , RNA, Long Noncoding/genetics , Trophoblasts/metabolism , Cell Line , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Pre-Eclampsia , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism
8.
Cell Death Dis ; 12(7): 649, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172716

ABSTRACT

Endothelial-mesenchymal transition (EndMT) is an important source of cancer-associated fibroblasts (CAFs), which facilitates tumour progression. PDAC is characterised by abundant CAFs and tumour necrosis factor-α (TNF-α). Here, we show that TNF-α strongly induces human endothelial cells to undergo EndMT. Interestingly, TNF-α strongly downregulates the expression of the endothelial receptor TIE1, and reciprocally TIE1 overexpression partially prevents TNF-α-induced EndMT, suggesting that TNF-α acts, at least partially, through TIE1 regulation in this process. We also show that TNF-α-induced EndMT is reversible. Furthermore, TNF-α treatment of orthotopic mice resulted in an important increase in the stroma, including CAFs. Finally, secretome analysis identified TNFSF12, as a regulator that is also present in PDAC patients. With the aim of restoring normal angiogenesis and better access to drugs, our results support the development of therapies targeting CAFs or inducing the EndMT reversion process in PDAC.


Subject(s)
Cancer-Associated Fibroblasts/drug effects , Carcinoma, Pancreatic Ductal/pathology , Endothelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Pancreatic Neoplasms/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cells, Cultured , Cytokine TWEAK/genetics , Cytokine TWEAK/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Male , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
9.
Cancer Treat Res Commun ; 27: 100364, 2021.
Article in English | MEDLINE | ID: mdl-33812182

ABSTRACT

BACKGROUND: Ovarian cancer is the most lethal gynecologic malignancy due to the tumor's acquisition of chemoresistance to platinum-based chemotherapy. To solve this problem, we conducted RNAi-based large-scale screening and determined that tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE-1) is a key molecule involved in the platinum resistance of ovarian cancer cells. Recently, a variety of studies have investigated that small extracellular vesicles (sEVs) contribute to the communication between cancer cells, including the development of chemoresistance in ovarian cancer. The purpose of our study is to determine if sEVs-derived TIE-1 is involved in the chemoresistance of ovarian cancer cells. MATERIALS AND METHODS: TIE-1-overexpressed TOV112D cells, termed TOV112DTIE-1 cells, were established, and sEVs were isolated from TOV112DTIE-1 cells supernatants by ultracentrifugation. We assessed cisplatin sensitivity in recipient cells with TOV112DTIE-1-derived sEVs by cell-Titer Glo kit. We also asked whether sEV-derived TIE-1 suppressed the DNA damage response in recipient cells and evaluated the DNA damage response by counting cells positive for DNA damage foci. RESULTS: TIE-1 was contained within sEVTIE-1 derived from the cellular supernatant of TOV112DTIE-1. We showed that sEV-derived TIE-1 decreased chemosensitivity to cisplatin by suppressing the DNA damage response in recipient cells. CONCLUSION: Our findings suggest that sEV-derived TIE-1 could be a new therapeutic target for refractory ovarian cancer.


Subject(s)
DNA Damage/drug effects , Drug Resistance, Neoplasm/genetics , Extracellular Vesicles/genetics , Ovarian Neoplasms/genetics , Receptor, TIE-1/genetics , Antineoplastic Agents/pharmacology , Cell Communication , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , DNA Repair/drug effects , Extracellular Vesicles/metabolism , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptor, TIE-1/metabolism , Transfection
10.
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
11.
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
12.
EMBO Mol Med ; 12(6): e11164, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32302470

ABSTRACT

The angiopoietin (Ang)-Tie pathway has been intensely pursued as candidate second-generation anti-angiogenic target. While much of the translational work has focused on the ligand Ang2, the clinical efficacy of Ang2-targeting drugs is limited and failed to improve patient survival. In turn, the orphan receptor Tie1 remains therapeutically unexplored, although its endothelial-specific genetic deletion has previously been shown to result in a strong reduction in metastatic growth. Here, we report a novel Tie1 function-blocking antibody (AB-Tie1-39), which suppressed postnatal retinal angiogenesis. During primary tumor growth, neoadjuvant administration of AB-Tie1-39 strongly impeded systemic metastasis. Furthermore, the administration of AB-Tie1-39 in a perioperative therapeutic window led to a significant survival advantage as compared to control-IgG-treated mice. Additional in vivo experimental metastasis and in vitro transmigration assays concurrently revealed that AB-Tie1-39 treatment suppressed tumor cell extravasation at secondary sites. Taken together, the data phenocopy previous genetic work in endothelial Tie1 KO mice and thereby validate AB-Tie1-39 as a Tie1 function-blocking antibody. The study establishes Tie1 as a therapeutic target for metastasis in a perioperative or neoadjuvant setting.


Subject(s)
Neoplasms , Receptor, TIE-1 , Angiopoietin-1 , Angiopoietin-2 , Animals , Gene Deletion , Humans , Mice , Neovascularization, Pathologic , Receptor, TIE-1/genetics , Receptor, TIE-2
13.
Int J Biol Sci ; 16(7): 1274-1287, 2020.
Article in English | MEDLINE | ID: mdl-32174801

ABSTRACT

Gliomas are highly malignant nervous system tumours. Studies shown that cancer stem cells are one of the main reasons underlying recurrence, metastasis, and poor prognosis in glioma cases. Our previous studies have found that superparamagnetic iron oxide nanoparticles (SPIONs) can act as nucleic acid carriers to drive intracellular overexpression of these nucleic acids. In this study, CD44+/CD133+ glioma stem cells (HuGSCs) were first isolated from surgically resected tissues from patients. qPCR and western blot results showed that Tie1 expression in HuGSCs was significantly higher thanexpression in CD44-/CD133- glioma cells. Bioinformatic analysis and luciferase reporter assays showed that miR-485-5p binds to specific loci on the 3'-UTR of Tie1 mRNA to inhibit Tie1 expression. Subsequently, miR-485-5p/miR-mut and SPION complexes were transfected into HuGSCs. Transmission electron microscopy showed that a highly dense metallic electron cloud is present in HuGSCs. At the same time, in vivo and in vitro studies showed that miR-485-5p@SPIONs can significantly inhibit HuGSC proliferation, invasion, tumourigenicity, and angiogenesis. In-depth analysis showed that Tie1 interacts with neuronal growth factors such as FGF2, BDNF, GDNF, and GFAP. qPCR and western blot results showed that in miR-485-5p@SPIONs-HuGSCs, the expression levels of Tie1 and stem cell markers (Oct4, Sox2, Nanog, CD44, and CD133), and even FGF2, BDNF, GDNF, and GFAP were significantly lower than thelevels in the control group (miR-mut@SPIONs-HuGSCs). Therefore, this study showedthat Tie1 is an important factor that maintains glioma stem cell activity. SPIONs drive miR-485-5p overexpression in cells and inhibit endogenous Tie1 expression to downregulate the protein expression levels of Fgf2/GDNF/GFAP/BDNF and significantly weaken the in vivo and in vitro viability of gliomas.


Subject(s)
Glioma/metabolism , Magnetic Iron Oxide Nanoparticles/chemistry , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Receptor, TIE-1/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Glioma/genetics , Humans , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , Receptor, TIE-1/genetics
14.
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
15.
Vascul Pharmacol ; 114: 103-109, 2019 03.
Article in English | MEDLINE | ID: mdl-30910126

ABSTRACT

Pervasive transcription is a feature of the human genome that requires better understanding. Over the last decade or so, RNA species longer than 200 nucleotides-dubbed long non-coding RNA (lncRNAs)-had been found in sense or anti-sense orientation within or outside of genes that encode proteins. Importantly, lncRNA-mediated gene regulation and the elements that control lncRNA expression are a source of fascination among molecular biologists. In vascular biology, a dozen or so lncRNAs had been identified, and progress occurs each day. In this review, we highlighted our laboratories' contribution to the lncRNA field by discussing lessons learned from two lncRNAs in the tyrosine kinase containing immunoglobulin and epidermal growth factor homology1 (Tie1) and delta-like 4 (Dll4) loci. These genes are responsible for basic vascular patterning and pathophysiological remodeling in angiogenesis.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Neovascularization, Physiologic/genetics , RNA, Long Noncoding/genetics , Receptor, TIE-1/genetics , Vascular Remodeling/genetics , Adaptor Proteins, Signal Transducing , Animals , Body Patterning , Calcium-Binding Proteins , Gene Expression Regulation , Gene Expression Regulation, Developmental , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Animal , RNA, Long Noncoding/metabolism , Receptor, TIE-1/metabolism , Signal Transduction , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
16.
Thorac Cancer ; 10(4): 715-727, 2019 04.
Article in English | MEDLINE | ID: mdl-30806032

ABSTRACT

BACKGROUND: Although cigarette smoking is considered one of the key risk factors for lung cancer, 15% of male patients and 53% of female patients with lung cancer are non-smokers. Metabolic changes are critical features of cancer. Therapeutic target identification from a metabolic perspective in non-small cell lung cancer (NSCLC) tissue of female non-smokers has long been ignored. RESULTS: Based on microarray data retrieved from Affymetrix expression arrays E-GEOD-19804, we found that the downregulated genes in non-smoking female NSCLC patients tended to participate in protein/amino acid and lipid metabolism, while upregulated genes were more involved in protein/amino acid and carbohydrate metabolism. Combining nutrient metabolic co-expression, protein-protein interaction network construction and overall survival assessment, we identified NR4A1 and TIE1 as potential therapeutic targets for NSCLC in female non-smokers. To accelerate the drug development for non-smoking female NSCLC patients, we identified nilotinib as a potential agonist targeting NR4A1 encoded protein by molecular docking and molecular dynamic stimulation. We also show that nilotinib inhibited proliferation and induced senescence of cells in non-smoking female NSCLC patients in vitro. CONCLUSIONS: These results not only uncover nutrient metabolic characteristics in non-smoking female NSCLC patients, but also provide a new paradigm for identifying new targets and drugs for novel therapy for such patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Pyrimidines/pharmacology , Receptor, TIE-1/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Non-Smokers/statistics & numerical data , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/chemistry , Protein Interaction Maps , Pyrimidines/therapeutic use , Receptor, TIE-1/metabolism , Survival Analysis
17.
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
18.
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
19.
Cancer Med ; 6(6): 1378-1388, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28464467

ABSTRACT

The Tie receptors 1 and 2 (Tie1/2) play crucial roles in embryonic angiogenesis. Recent studies suggest enhanced expression of Tie1 in several types of cancer and negative correlations between Tie1 levels and clinical outcome. These observations suggest important functions of Tie1 not only for vascular formation but also in tumorigenesis. Ligands for Tie2, that is angiopoietins 1-4, have been identified, but not for Tie1. To determine the molecular functions of Tie1, its detailed characterization in tumors would be helpful. Herein, we report that Tie1 is up-regulated in colorectal cancer. Detailed analysis using tumor-bearing models and immunohistochemistry combined with Flow cytometric analysis and cell sorting (FACS) revealed that Tie1 protein was expressed in a small population of malignant tumor cells. Intriguingly, Tie1 expression was observed and could be maintained only in vivo. Further analysis using sphere-formation culture revealed that Tie1-positive cells are enriched within the population of tumor cells with cancer stemness properties. Indeed, Tie1-positive tumor cells derived from a murine model overexpressed Lgr5, a typical stemness marker for colorectal cancer. Our results provide a novel insight into Tie1 function in tumorigenesis and suggest clinical applications to target cancer stem cells.


Subject(s)
Colorectal Neoplasms/metabolism , Receptor, TIE-1/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Humans , Mice , Mice, Nude , Neoplastic Stem Cells , RNA, Messenger/metabolism , Receptor, TIE-1/genetics , Up-Regulation
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...