1
|
Peng H, Du Z, Li J, Wang W, Li Z, Ru S. The sprouting angiogenesis and vascular dysfunction triggered by bisphenol S and tetrabromobisphenol S through disrupting vascular endothelial-cadherin in zebrafish. ENVIRONMENTAL RESEARCH 2025; 278:121632. [PMID: 40246265 DOI: 10.1016/j.envres.2025.121632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Exogenous chemical toxicants may be important inducers of pathological angiogenesis diseases. However, few studies have investigated the associations between pathological angiogenesis diseases and chemical toxicant exposures, and the specific mechanism by which chemical toxicants induce sprouting angiogenesis is unclear. In this study, zebrafish were exposed to bisphenol S (BPS, 1-100 μg/L) and tetrabromobisphenol S (TBBPS, 0.1 and 10 μg/L) from the embryonic stage to the larval stage to investigate how pollutants interfere with angiogenesis and the function of ectopic sprouting vessels. The results showed that BPS and TBBPS promoted ectopic sprouting angiogenesis in different types of vascular plexuses, including the posterior cardinal vein (PCV) and superficial choroidal vessels (SOVs), at different developmental time points. Proteomic analyses of eGFP-positive endothelial cells (ECs) isolated from Tg(flk1: eGFP) zebrafish revealed that both BPS and TBBPS induced ectopic angiogenesis by acting on vascular endothelial-cadherin (VE-cadherin) and activating downstream proangiogenic signaling. In ectopic sprouting vessels induced by BPS and TBBPS, increased endothelial permeability resulted in white blood cell recruitment. Human oxidized lipids also tended to deposit in these ectopic vessels following BPS and TBBPS exposure. These findings suggest that chemical toxicant-induced ectopic angiogenesis is an important cause of vascular dysfunction and related diseases.
Collapse
Affiliation(s)
- Hongyuan Peng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Zehui Du
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Jiali Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| | - Ze Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
2
|
Nishimi S, Fukuse S, Miwa Y, Wakabayashi K, Isozaki T. VE-cadherin may suppress inflammation depending on the phase of inflammation of rheumatoid arthritis. Mod Rheumatol 2025; 35:449-457. [PMID: 39656893 DOI: 10.1093/mr/roae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/24/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVES A disintegrin and metalloproteinase (ADAM)-15 and vascular endothelial (VE)-cadherin are involved in angiogenesis. We investigated the relationship between ADAM-15 and VE-cadherin expressions in rheumatoid arthritis (RA). METHODS VE-cadherin concentrations in the serum of patients with RA were measured using the enzyme linked immunosorbent assay. We stimulated fibroblast-like synoviocytes (RA-FLS) with VE-cadherin and measured the vascular endothelial growth factor (VEGF) and inflammatory cytokine levels using ELISA. We also examined the correlation between serum VE-cadherin levels and DAS-28ESR, and used the Matrigel assay to examine VE-cadherin involvement in angiogenesis. RESULTS Serum VE-cadherin levels were significantly higher in patients with RA than in healthy controls. A negative correlation was observed between VE-cadherin and DAS-28ESR. VEGF, chemokine ligand 16, intercellular adhesion molecule-1, and interleukin-8 levels in the supernatant of RA-FLS or human umbilical vein endothelial cells stimulated with VE-cadherin were significantly lower than those in the controls. The number of intercellular bridges formed by endothelial cells using Matrigel significantly decreased in RA synovial fluids from which VE-cadherin had been removed compared to synovial fluids treated with control immunoglobulin G. CONCLUSION VE-cadherin may have an inhibitory effect on inflammation depending on the phase of RA inflammation.
Collapse
Affiliation(s)
- Shinichiro Nishimi
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Sayaka Fukuse
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yusuke Miwa
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kuninobu Wakabayashi
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takeo Isozaki
- Division of Rheumatology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Department of Pathogenesis and Translational Medicine, Showa University Graduate School of Pharmacy, Tokyo, Japan
| |
Collapse
|
3
|
Vergroesen TM, Vermeulen V, Merks RMH. Falsifying computational models of endothelial cell network formation through quantitative comparison with in vitro models. PLoS Comput Biol 2025; 21:e1012965. [PMID: 40305554 PMCID: PMC12074657 DOI: 10.1371/journal.pcbi.1012965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 05/13/2025] [Accepted: 03/14/2025] [Indexed: 05/02/2025] Open
Abstract
During angiogenesis, endothelial cells expand the vasculature by migrating from existing blood vessels, proliferating and collectively organizing into new capillaries. In vitro and in vivo experimentation is instrumental for identifying the molecular players and cell behaviour that regulate angiogenesis. Alongside experimental work, computational and mathematical models of endothelial cell network formation have helped to analyse if the current molecular and cellular understanding of endothelial cell behaviour is sufficient to explain the formation of endothelial cell networks. As input, the models take (a subset of) the current knowledge or hypotheses of single cell behaviour and capture it into a dynamical, mathematical description. As output, they predict the multicellular behaviour following from the actions of many individual cells, i.e., formation of a vascular-like network. Paradoxically, computational modelling based on different assumptions, i.e., completely different, sometimes non-intersecting sets of observed single cell behaviour, can reproduce the same angiogenesis-like multicellular behaviour, making it practically impossible to decide which, if any, of these models is correct. Here we present dynamical analyses of time-lapses of in vitro endothelial cell network formation experiments and compare these with dynamic analyses of three mathematical models: (1) the cell elongation model; (2) the contact-inhibited chemotaxis model; and (3) the mechanical cell-cell communication model. We extract a variety of dynamical characteristics of endothelial cell network formation using a custom time-lapse video analysis pipeline in ImageJ. We compare the dynamical network characteristics of the in vitro experiments to those of the cellular networks produced by the computational models. We test the response of the in silico dynamical cell network characteristics to changes in cell density and make related changes in the in vitro experiments. Of the three computational models that we have considered, the cell elongation model best captures the remodelling phase of in vitro endothelial cell network formation. Furthermore, in the in vitro model, the final size and number of lacunae in the network are independent of the initial cell density. This observation is also reproduced in the cell elongation model, but not in the other two models that we have considered. Altogether, we present an approach to model validation based on comparisons of time-resolved data and variations of model conditions.
Collapse
Affiliation(s)
| | - Vincent Vermeulen
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Roeland M. H. Merks
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|
4
|
Katsuura M, Homma J, Higashi Y, Sekine H, Shimizu T. Densely vascularized thick 3D tissue shows enhanced protein secretion constructed with intermittent positive pressure. Commun Biol 2025; 8:201. [PMID: 39922952 PMCID: PMC11807115 DOI: 10.1038/s42003-025-07627-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
Constructing a dense vascular endothelial network within engineered tissue is crucial for successful engraftment. The present study investigated the effects of air-compressing intermittent positive pressure (IPP) on co-cultured mesenchymal stem cells and vascular endothelial cells and evaluated the potential of IPP-cultured cell sheets for transplantation therapy. The results demonstrated that the IPP (+) group exhibited a denser vascular endothelial network and significantly increased cell sheet thickness compared to the IPP (-) group. Furthermore, in vivo experiments showed that IPP-cultured cell sheets enhanced the secretion of Gaussian luciferase by genetically modified mesenchymal stem cells. These findings highlight the IPP method as a technique that simultaneously enables the thickening of planar tissues and the construction of vascular networks. This approach demonstrates promise for fabricating functional, transplantable, and thick tissues with dense vascularization and a high capacity for protein secretion, paving the way for novel applications in regenerative medicine.
Collapse
Affiliation(s)
- Misako Katsuura
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
- Department of Pediatrics, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Jun Homma
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan.
| | | | - Hidekazu Sekine
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan.
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
5
|
Yakovlev S, Nyenhuis DA, Tjandra N, Strickland DK, Medved L. Identification of Amino Acid Residues Critical for the Interaction of Fibrin with N-Cadherin. Biochemistry 2025; 64:83-91. [PMID: 39670513 PMCID: PMC11892114 DOI: 10.1021/acs.biochem.4c00510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
We recently identified N-cadherin as a novel receptor for fibrin and localized complementary binding sites within the fibrin βN-domains and the third and fifth extracellular domains (EC3 and EC5) of N-cadherin. We also hypothesized that the His16 and Arg17 residues of the βN-domains and the (Asp/Glu)-X-(Asp/Glu) motifs present in the EC3 and EC5 domains may play roles in the interaction between fibrin and N-cadherin. The primary objectives of this study were to test these hypotheses and to further clarify the structural basis for this interaction. To test our hypotheses, we first mutated His16 and Arg17 in the recombinant (β15-66)2 fragment, which mimics the dimeric arrangement of the βN-domains in fibrin, using site-directed mutagenesis. The results revealed that the mutations of both His16 and Arg17 are critical for the interaction. Next, we mutated Asp/Glu residues in the three (Asp/Glu)-X-(Asp/Glu) motifs, M1 (Asp-Phe-Glu), M2 (Glu-Ala-Glu), and M3 (Asp-Tyr-Asp), of the fibrin-binding N-cad(3-5) fragment of N-cadherin. The results showed that Asp292 and Glu294 of M1, and Asp468 and Asp470 of M3, are critical for the interaction. Our molecular modeling of the 3D structure of the EC3-EC4-EC5 domains revealed that these residues are located at the interfaces of EC3-EC4 and EC4-EC5 and that some may also be involved in calcium binding. In conclusion, our study identified amino acid residues in the fibrin βN-domains and the EC3 and EC5 domains of N-cadherin that are critical for the interaction of fibrin with N-cadherin and localized the fibrin-binding residues in the 3D structure of N-cadherin.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - David A. Nyenhuis
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nico Tjandra
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases and Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| |
Collapse
|
6
|
Magnusen AF, Pandey MK. Complement System and Adhesion Molecule Skirmishes in Fabry Disease: Insights into Pathogenesis and Disease Mechanisms. Int J Mol Sci 2024; 25:12252. [PMID: 39596318 PMCID: PMC11594573 DOI: 10.3390/ijms252212252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Fabry disease is a rare X-linked lysosomal storage disorder caused by mutations in the galactosidase alpha (GLA) gene, resulting in the accumulation of globotriaosylceramide (Gb3) and its deacetylated form, globotriaosylsphingosine (Lyso-Gb3) in various tissues and fluids throughout the body. This pathological accumulation triggers a cascade of processes involving immune dysregulation and complement system activation. Elevated levels of complement 3a (C3a), C5a, and their precursor C3 are observed in the plasma, serum, and tissues of patients with Fabry disease, correlating with significant endothelial cell abnormalities and vascular dysfunction. This review elucidates how the complement system, particularly through the activation of C3a and C5a, exacerbates disease pathology. The activation of these pathways leads to the upregulation of adhesion molecules, including vascular cell adhesion molecule 1 (VCAM1), intercellular adhesion molecule 1 (ICAM1), platelet and endothelial cell adhesion molecule 1 (PECAM1), and complement receptor 3 (CR3) on leukocytes and endothelial cells. This upregulation promotes the excessive recruitment of leukocytes, which in turn exacerbates disease pathology. Targeting complement components C3a, C5a, or their respective receptors, C3aR (C3a receptor) and C5aR1 (C5a receptor 1), could potentially reduce inflammation, mitigate tissue damage, and improve clinical outcomes for individuals with Fabry disease.
Collapse
Affiliation(s)
- Albert Frank Magnusen
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
7
|
Zhang D, Zhu Z, Wen K, Zhang S, Liu J. Netrin‑4 promotes VE‑cadherin expression in endothelial cells through the NF‑κB signaling pathway. Exp Ther Med 2024; 28:351. [PMID: 39071904 PMCID: PMC11273250 DOI: 10.3892/etm.2024.12640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024] Open
Abstract
Netrin-4 (NTN4), a secreted protein from the Netrin family, has been recognized for its role in vascular development, endothelial homeostasis and angiogenesis. Vascular endothelial (VE)-cadherin is a specialized adhesion protein located at the intercellular junctions of endothelial cells (ECs), and regulates migration, proliferation and permeability. To date, the relationship between NTN4 and VE-cadherin in ECs remains unclear. In the present study, human umbilical vein ECs (HUVECs) were transfected with NTN4 overexpression plasmid, resulting in NTN4 overexpression. Reverse transcription-quantitative PCR and western blotting were used to determine gene and protein expression. CCK8, wound healing, and Transwell assays were performed to evaluate cell proliferation, migration and permeability. NTN4 overexpression decreased HUVEC viability and migration. In addition, NTN4 overexpression increased the expression of VE-cadherin and decreased the permeability of HUVECs. Subsequent studies showed that NTN4 overexpression increased the NF-κB protein level and decreased IκB-α protein expression in HUVECs. In HUVECs treated with NF-κB inhibitor pyrrolidine dithiocarbamate, the expression of VE-cadherin failed to increase with NTN4 overexpression. Taken together, the results indicated that NTN4 overexpression increased VE-cadherin expression through the activation of the NF-κB signaling pathway in HUVECs. The present findings revealed a novel regulatory mechanism for VE-cadherin expression and suggested a novel avenue for future research on the role of NTN4 in endothelial barrier-related diseases.
Collapse
Affiliation(s)
- Datong Zhang
- Department of Orthodontics, School of Dentistry, Shandong University, Jinan, Shandong 250100, P.R. China
| | - Zhiying Zhu
- Institute of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Keting Wen
- Institute of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Shijie Zhang
- Department of Orthodontics, School of Dentistry, Shandong University, Jinan, Shandong 250100, P.R. China
- Department of Stomatology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ju Liu
- Institute of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
8
|
Rämö JT, Gorman B, Weng LC, Jurgens SJ, Singhanetr P, Tieger MG, van Dijk EH, Halladay CW, Wang X, Brinks J, Choi SH, Luo Y, Pyarajan S, Nealon CL, Gorin MB, Wu WC, Sobrin L, Kaarniranta K, Yzer S, Palotie A, Peachey NS, Turunen JA, Boon CJ, Ellinor PT, Iyengar SK, Daly MJ, Rossin EJ. Rare genetic variation in VE-PTP is associated with central serous chorioretinopathy, venous dysfunction and glaucoma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.08.24307013. [PMID: 38766240 PMCID: PMC11100937 DOI: 10.1101/2024.05.08.24307013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Central serous chorioretinopathy (CSC) is a fluid maculopathy whose etiology is not well understood. Abnormal choroidal veins in CSC patients have been shown to have similarities with varicose veins. To identify potential mechanisms, we analyzed genotype data from 1,477 CSC patients and 455,449 controls in FinnGen. We identified an association for a low-frequency (AF=0.5%) missense variant (rs113791087) in the gene encoding vascular endothelial protein tyrosine phosphatase (VE-PTP) (OR=2.85, P=4.5×10-9). This was confirmed in a meta-analysis of 2,452 CSC patients and 865,767 controls from 4 studies (OR=3.06, P=7.4×10-15). Rs113791087 was associated with a 56% higher prevalence of retinal abnormalities (35.3% vs 22.6%, P=8.0×10-4) in 708 UK Biobank participants and, surprisingly, with varicose veins (OR=1.31, P=2.3×10-11) and glaucoma (OR=0.82, P=6.9×10-9). Predicted loss-of-function variants in VEPTP, though rare in number, were associated with CSC in All of Us (OR=17.10, P=0.018). These findings highlight the significance of VE-PTP in diverse ocular and systemic vascular diseases.
Collapse
Affiliation(s)
- Joel T Rämö
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Massachusetts Eye and Ear, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Bryan Gorman
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Lu-Chen Weng
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean J Jurgens
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Panisa Singhanetr
- Massachusetts Eye and Ear, Boston, MA, USA
- Mettapracharak Eye Institute, Mettapracharak (Wat Rai Khing) Hospital, Nakhon Pathom, Thailand
| | - Marisa G Tieger
- New England Eye Center, Tufts Medical Center, Boston, MA, USA
| | - Elon Hc van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christopher W Halladay
- Center of Innovation in Long Term Services and Supports, Providence VA Medical Center, Providence, RI, USA
| | - Xin Wang
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joost Brinks
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Seung Hoan Choi
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Yuyang Luo
- Massachusetts Eye and Ear, Boston, MA, USA
| | - Saiju Pyarajan
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard School of Medicine, Boston, MA, USA
| | - Cari L Nealon
- Eye Clinic, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
| | - Michael B Gorin
- Department of Ophthalmology, David Geffen School of Medicine, Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wen-Chih Wu
- Section of Cardiology, Medical Service, VA Providence Healthcare System, Providence, RI, USA
| | - Lucia Sobrin
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Suzanne Yzer
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Neal S Peachey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Joni A Turunen
- Folkhälsan Research Center, Biomedicum, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Camiel Jf Boon
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sudha K Iyengar
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Mark J Daly
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elizabeth J Rossin
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
9
|
Cruz-Holguín VJ, González-García LD, Velázquez-Cervantes MA, Arévalo-Romero H, De Jesús-González LA, Helguera-Repetto AC, León-Reyes G, Salazar MI, Cedillo-Barrón L, León-Juárez M. Collateral Damage in the Placenta during Viral Infection in Pregnancy: A Possible Mechanism for Vertical Transmission and an Adverse Pregnancy Outcome. Diseases 2024; 12:59. [PMID: 38534983 PMCID: PMC10969698 DOI: 10.3390/diseases12030059] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 11/11/2024] Open
Abstract
In mammals, the placenta is a connection between a mother and a new developing organism. This tissue has a protective function against some microorganisms, transports nutrients, and exchanges gases and excretory substances between the mother and the fetus. Placental tissue is mainly composed of chorionic villi functional units called trophoblasts (cytotrophoblasts, the syncytiotrophoblast, and extravillous trophoblasts). However, some viruses have developed mechanisms that help them invade the placenta, causing various conditions such as necrosis, poor perfusion, and membrane rupture which, in turn, can impact the development of the fetus and put the mother's health at risk. In this study, we collected the most relevant information about viral infection during pregnancy which can affect both the mother and the fetus, leading to an increase in the probability of vertical transmission. Knowing these mechanisms could be relevant for new research in the maternal-fetal context and may provide options for new therapeutic targets and biomarkers in fetal prognosis.
Collapse
Affiliation(s)
- Victor Javier Cruz-Holguín
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
| | - Luis Didier González-García
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
- Posgrado de Inmunología, Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico;
| | - Manuel Adrián Velázquez-Cervantes
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
| | - Haruki Arévalo-Romero
- Laboratorio de Inmunologia y Microbiologia Molecular, Division Academica Multidisciplinaria de Jalpa de Méndez, Jalpa de Mendez 86205, Mexico;
| | | | | | - Guadalupe León-Reyes
- Laboratorio de Nutrigenética y Nutrigenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Ma. Isabel Salazar
- Posgrado de Inmunología, Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico;
- Laboratorio Nacional de Vacunología y Virus Tropicales (LNVyVT), Escuela Nacional de Ciencias Biologócas (ENCB), Instituto Politecnico Naciona, Mexico City 11350, Mexico
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del IPN (CINVESTAV), Mexico City 07360, Mexico;
| | - Moisés León-Juárez
- Laboratorio de Virologia Perinatal y Diseño Molecular de Antigenos y Biomarcadores, Departamento de Inmunobioquimica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico; (V.J.C.-H.); (L.D.G.-G.); (M.A.V.-C.)
| |
Collapse
|
10
|
Keshavanarayana P, Spill F. A mechanical modeling framework to study endothelial permeability. Biophys J 2024; 123:334-348. [PMID: 38169215 PMCID: PMC10870174 DOI: 10.1016/j.bpj.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/06/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
The inner lining of blood vessels, the endothelium, is made up of endothelial cells. Vascular endothelial (VE)-cadherin protein forms a bond with VE-cadherin from neighboring cells to determine the size of gaps between the cells and thereby regulate the size of particles that can cross the endothelium. Chemical cues such as thrombin, along with mechanical properties of the cell and extracellular matrix are known to affect the permeability of endothelial cells. Abnormal permeability is found in patients suffering from diseases including cardiovascular diseases, cancer, and COVID-19. Even though some of the regulatory mechanisms affecting endothelial permeability are well studied, details of how several mechanical and chemical stimuli acting simultaneously affect endothelial permeability are not yet understood. In this article, we present a continuum-level mechanical modeling framework to study the highly dynamic nature of the VE-cadherin bonds. Taking inspiration from the catch-slip behavior that VE-cadherin complexes are known to exhibit, we model the VE-cadherin homophilic bond as cohesive contact with damage following a traction-separation law. We explicitly model the actin cytoskeleton and substrate to study their role in permeability. Our studies show that mechanochemical coupling is necessary to simulate the influence of the mechanical properties of the substrate on permeability. Simulations show that shear between cells is responsible for the variation in permeability between bicellular and tricellular junctions, explaining the phenotypic differences observed in experiments. An increase in the magnitude of traction force due to disturbed flow that endothelial cells experience results in increased permeability, and it is found that the effect is higher on stiffer extracellular matrix. Finally, we show that the cylindrical monolayer exhibits higher permeability than the planar monolayer under unconstrained cases. Thus, we present a contact mechanics-based mechanochemical model to investigate the variation in the permeability of endothelial monolayer due to multiple loads acting simultaneously.
Collapse
Affiliation(s)
| | - Fabian Spill
- School of Mathematics, University of Birmingham, Birmingham, United Kingdom.
| |
Collapse
|
11
|
Yakovlev S, Tjandra N, Strickland DK, Medved L. Identification of Neural (N)-Cadherin as a Novel Endothelial Cell Receptor for Fibrin and Localization of the Complementary Binding Sites. Biochemistry 2024; 63:202-211. [PMID: 38156948 PMCID: PMC10848343 DOI: 10.1021/acs.biochem.3c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Based on the high structural homology between vascular endothelial (VE)-cadherin and neural (N)-cadherin, we hypothesized that fibrin, which is known to interact with VE-cadherin and promote angiogenesis through this interaction, may also interact with N-cadherin. To test this hypothesis, we prepared fibrin and its plasmin-produced and recombinant fragments covering practically all parts of the fibrin molecule. We also prepared the soluble extracellular portion of N-cadherin (sN-cadherin), which includes all five extracellular N-cadherin domains, and studied its interaction with fibrinogen, fibrin, and the aforementioned fibrin fragments using two independent methods, ELISA and SPR. The experiments confirmed our hypothesis, revealing that fibrin interacts with sN-cadherin with high affinity. Furthermore, the experiments localized the N-cadherin binding site within the fibrin βN-domains. Notably, the recombinant dimeric (β15-66)2 fragment, corresponding to these domains and mimicking their dimeric arrangement in fibrin, preserved the N-cadherin-binding properties of fibrin. To localize the fibrin binding site within N-cadherin, we performed ELISA and SPR experiments with (β15-66)2 and recombinant N-cadherin fragments representing its individual extracellular domains and combinations thereof. The results obtained indicate that the interaction of fibrin with N-cadherin occurs through the third and fifth extracellular domains of the latter. This is in contrast to our previous study, which revealed that fibrin interacts only with the third extracellular domain of VE-cadherin. In conclusion, our study identified N-cadherin as a novel receptor for fibrin and localized complementary binding sites within both fibrin and N-cadherin. The pathophysiological role of this interaction remains to be established.
Collapse
Affiliation(s)
- Sergiy Yakovlev
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Nico Tjandra
- Laboratory of Structural Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases and Departments of Surgery and Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Leonid Medved
- Center for Vascular and Inflammatory Diseases and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| |
Collapse
|
12
|
Nguyen M, Sullivan J, Shen W. Retinal vascular remodeling in photoreceptor degenerative disease. Exp Eye Res 2023; 234:109566. [PMID: 37423458 DOI: 10.1016/j.exer.2023.109566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/05/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
Abnormal vasculature in the retina, specifically tortuous vessels and capillary degeneration, is common in many of the most prevalent retinal degenerative diseases, currently affecting millions of people across the world. However, the formation and development of abnormal vasculature in the context of retinal degenerative diseases are still poorly understood. The FVB/N (rd1) and rd10 mice are well-studied animal models of retinal degenerative diseases, but how photoreceptor degeneration leads to vascular abnormality in the diseases remains to be elucidated. Here, we used advancements in confocal microscopy, immunohistochemistry, and image analysis software to systematically characterize the pathological vasculature in the FVB/N (rd1) and rd10 mice, known as a chronic, rapid and slower retinal degenerative model, respectively. We demonstrated that there was plexus-specific vascular degeneration in the retinal trilaminar vascular network paralleled to photoreceptor degeneration in the diseased retinas. We also quantitatively analyzed the vascular structural architecture in the wild-type and diseased retinas to provide valuable information on vascular remodeling in retinal degenerative disease.
Collapse
Affiliation(s)
- Matthew Nguyen
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - James Sullivan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Wen Shen
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA.
| |
Collapse
|
13
|
Bouillet L, Deroux A, Benmarce M, Guérin C, Bouvet L, Garnier O, Martin DK, Vilgrain I. Molecular Mechanisms of Endothelialitis in SARS-CoV-2 Infection: Evidence for VE-Cadherin Cleavage by ACE2. Int J Mol Sci 2023; 24:12525. [PMID: 37569899 PMCID: PMC10419376 DOI: 10.3390/ijms241512525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Long COVID-19 syndrome appears after Severe Acute Respiratory Syndrome-Corona Virus (SARS-CoV-2) infection with acute damage to microcapillaries, microthrombi, and endothelialitis. However, the mechanisms involved in these processes remain to be elucidated. All blood vessels are lined with a monolayer of endothelial cells called vascular endothelium, which provides a the major function is to prevent coagulation. A component of endothelial cell junctions is VE-cadherin, which is responsible for maintaining the integrity of the vessels through homophilic interactions of its Ca++-dependent adhesive extracellular domain. Here we provide the first evidence that VE-cadherin is a target in vitro for ACE2 cleavage because its extracellular domain (hrVE-ED) contains two amino acid sequences for ACE2 substrate recognition at the positions 256P-F257 and 321PMKP-325L. Indeed, incubation of hrVE-ED with the active ectopeptidase hrACE2 for 16 hrs in the presence of 10 μM ZnCl2 showed a dose-dependent (from 0.2 ng/μL to 2 ng/μL) decrease of the VE-cadherin immunoreactive band. In vivo, in the blood from patients having severe COVID-19 we detected a circulating form of ACE2 with an apparent molecular mass of 70 kDa, which was barely detectable in patients with mild COVID-19. Of importance, in the patients with severe COVID-19 disease, the presence of three soluble fragments of VE-cadherin (70, 62, 54 kDa) were detected using the antiEC1 antibody while only the 54 kDa fragment was present in patients with mild disease. Altogether, these data clearly support a role for ACE2 to cleave VE-cadherin, which leads to potential biomarkers of SARS-CoV-2 infection related with the vascular disease in "Long COVID-19".
Collapse
Affiliation(s)
- Laurence Bouillet
- University Grenoble Alpes, CNRS, TIMC-IMAG/T-RAIG (UMR 5525), 38000 Grenoble, France; (L.B.)
- Grenoble Hospital Grenoble Alpes (CHUGA), University Grenoble Alpes, 38000 Grenoble, France;
- Internal Medicine, University Hospital Centre Grenoble Alpes, CEDEX 9, 38043 Grenoble, France;
| | - Alban Deroux
- Grenoble Hospital Grenoble Alpes (CHUGA), University Grenoble Alpes, 38000 Grenoble, France;
| | - Meryem Benmarce
- University Grenoble Alpes, CNRS, TIMC-IMAG/SyNaBi (UMR 5525), 38000 Grenoble, France (D.K.M.)
| | - Chloé Guérin
- Internal Medicine, University Hospital Centre Grenoble Alpes, CEDEX 9, 38043 Grenoble, France;
- University Grenoble Alpes, INSERM U13, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Laboratory of Biosciences et Bioingénierie Pour la Santé (BGE)-Biomics, 38000 Grenoble, France
| | - Laura Bouvet
- University Grenoble Alpes, CNRS, TIMC-IMAG/T-RAIG (UMR 5525), 38000 Grenoble, France; (L.B.)
| | - Olivia Garnier
- University Grenoble Alpes, INSERM U13, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Laboratory of Biosciences et Bioingénierie Pour la Santé (BGE)-Biomics, 38000 Grenoble, France
| | - Donald K. Martin
- University Grenoble Alpes, CNRS, TIMC-IMAG/SyNaBi (UMR 5525), 38000 Grenoble, France (D.K.M.)
| | - Isabelle Vilgrain
- University Grenoble Alpes, INSERM U13, CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Laboratory of Biosciences et Bioingénierie Pour la Santé (BGE)-Biomics, 38000 Grenoble, France
| |
Collapse
|
14
|
Tonami K, Hayashi T, Uchijima Y, Kanai M, Yura F, Mada J, Sugahara K, Kurihara Y, Kominami Y, Ushijima T, Takubo N, Liu X, Tozawa H, Kanai Y, Tokihiro T, Kurihara H. Coordinated linear and rotational movements of endothelial cells compartmentalized by VE-cadherin drive angiogenic sprouting. iScience 2023; 26:107051. [PMID: 37426350 PMCID: PMC10329149 DOI: 10.1016/j.isci.2023.107051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/22/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Angiogenesis is a sequential process to extend new blood vessels from preexisting ones by sprouting and branching. During angiogenesis, endothelial cells (ECs) exhibit inhomogeneous multicellular behaviors referred to as "cell mixing," in which ECs repetitively exchange their relative positions, but the underlying mechanism remains elusive. Here we identified the coordinated linear and rotational movements potentiated by cell-cell contact as drivers of sprouting angiogenesis using in vitro and in silico approaches. VE-cadherin confers the coordinated linear motility that facilitated forward sprout elongation, although it is dispensable for rotational movement, which was synchronous without VE-cadherin. Mathematical modeling recapitulated the EC motility in the two-cell state and angiogenic morphogenesis with the effects of VE-cadherin-knockout. Finally, we found that VE-cadherin-dependent EC compartmentalization potentiated branch elongations, and confirmed this by mathematical simulation. Collectively, we propose a way to understand angiogenesis, based on unique EC behavioral properties that are partially dependent on VE-cadherin function.
Collapse
Affiliation(s)
- Kazuo Tonami
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
| | - Tatsuya Hayashi
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Graduate School of Mathematical Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8914, Japan
- Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Yasunobu Uchijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masahiro Kanai
- Department of Education and Creation Engineering, Kurume Institute of Technology, 2228-66 Kamitsu-machi, Kurume, Fukuoka 830-0052, Japan
| | - Fumitaka Yura
- Department of Complex and Intelligent Systems, School of Systems Information Science, Future University Hakodate, 116-2 Kamedanakano-cho, Hakodate, Hokkaido 041-8655, Japan
| | - Jun Mada
- College of Industrial Technology, Nihon University, 2-11-1 Shin-ei, Narashino, Chiba 275-8576, Japan
| | - Kei Sugahara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuri Kominami
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-865, Japan
| | - Toshiyuki Ushijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naoko Takubo
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Isotope Science Center, The University of Tokyo, 2-11-16, Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Xiaoxiao Liu
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideto Tozawa
- Department of Chemistry, Graduate School of Science, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshimitsu Kanai
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Tetsuji Tokihiro
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Graduate School of Mathematical Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8914, Japan
- Department of Mathematical Engineering, Faculty of Engineering, Musashino University, 3-3-3 Ariake, Koto-ku, Tokyo 135-8181, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
15
|
Davies EM, Gurung R, Le KQ, Roan KT, Harvey RP, Mitchell GM, Schwarz Q, Mitchell CA. PI(4,5)P 2-dependent regulation of endothelial tip cell specification contributes to angiogenesis. SCIENCE ADVANCES 2023; 9:eadd6911. [PMID: 37000875 PMCID: PMC10065449 DOI: 10.1126/sciadv.add6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Dynamic positioning of endothelial tip and stalk cells, via the interplay between VEGFR2 and NOTCH signaling, is essential for angiogenesis. VEGFR2 activates PI3K, which phosphorylates PI(4,5)P2 to PI(3,4,5)P3, activating AKT; however, PI3K/AKT does not direct tip cell specification. We report that PI(4,5)P2 hydrolysis by the phosphoinositide-5-phosphatase, INPP5K, contributes to angiogenesis. INPP5K ablation disrupted tip cell specification and impaired embryonic angiogenesis associated with enhanced DLL4/NOTCH signaling. INPP5K degraded a pool of PI(4,5)P2 generated by PIP5K1C phosphorylation of PI(4)P in endothelial cells. INPP5K ablation increased PI(4,5)P2, thereby releasing β-catenin from the plasma membrane, and concurrently increased PI(3,4,5)P3-dependent AKT activation, conditions that licensed DLL4/NOTCH transcription. Suppression of PI(4,5)P2 in INPP5K-siRNA cells by PIP5K1C-siRNA, restored β-catenin membrane localization and normalized AKT signaling. Pharmacological NOTCH or AKT inhibition in vivo or genetic β-catenin attenuation rescued angiogenesis defects in INPP5K-null mice. Therefore, PI(4,5)P2 is critical for β-catenin/DLL4/NOTCH signaling, which governs tip cell specification during angiogenesis.
Collapse
Affiliation(s)
- Elizabeth M. Davies
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Rajendra Gurung
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Kai Qin Le
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Katherine T. T. Roan
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
- School of Clinical Medicine and School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Geraldine M. Mitchell
- O’Brien Institute Department of St Vincent’s Institute and University of Melbourne, Department of Surgery, St. Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
- Health Sciences Faculty, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia 5001, Australia
| | - Christina A. Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| |
Collapse
|
16
|
Garnier O, Vilgrain I. Dialogue between VE-Cadherin and Sphingosine 1 Phosphate Receptor1 (S1PR1) for Protecting Endothelial Functions. Int J Mol Sci 2023; 24:ijms24044018. [PMID: 36835432 PMCID: PMC9959973 DOI: 10.3390/ijms24044018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
The endothelial cells (EC) of established blood vessels in adults remain extraordinarily quiescent in the sense that they are not actively proliferating, but they fulfill the necessary role to control the permeability of their monolayer that lines the interior of blood vessels. The cell-cell junctions between ECs in the endothelium comprise tight junctions and adherens homotypic junctions, which are ubiquitous along the vascular tree. Adherens junctions are adhesive intercellular contacts that are crucial for the organization of the EC monolayer and its maintenance and regulation of normal microvascular function. The molecular components and underlying signaling pathways that control the association of adherens junctions have been described in the last few years. In contrast, the role that dysfunction of these adherens junctions has in contributing to human vascular disease remains an important open issue. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid mediator found at high concentrations in blood which has important roles in the control of the vascular permeability, cell recruitment, and clotting that follow inflammatory processes. This role of S1P is achieved through a signaling pathway mediated through a family of G protein-coupled receptors designated as S1PR1. This review highlights novel evidence for a direct linkage between S1PR1 signaling and the mediation of EC cohesive properties that are controlled by VE-cadherin.
Collapse
|
17
|
Li H, Chang HM, Li S, Klausen C, Shi Z, Leung PC. Characterization of the roles of amphiregulin and transforming growth factor β1 in microvasculature-like formation in human granulosa-lutein cells. Front Cell Dev Biol 2022; 10:968166. [PMID: 36092732 PMCID: PMC9448859 DOI: 10.3389/fcell.2022.968166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Vascular endothelial-cadherin (VE-cadherin) is an essential component that regulates angiogenesis during corpus luteum formation. Amphiregulin (AREG) and transforming growth factor β1 (TGF-β1) are two intrafollicular factors that possess opposite functions in directing corpus luteum development and progesterone synthesis in human granulosa-lutein (hGL) cells. However, whether AREG or TGF-β1 regulates the VE-cadherin expression and subsequent angiogenesis in the human corpus luteum remains to be elucidated. Results showed that hGL cells cultured on Matrigel spontaneously formed capillary-like and sprout-like microvascular networks. Results of specific inhibitor treatment and small interfering RNA-mediated knockdown revealed that AREG promoteed microvascular-like formation in hGL cells by upregulating the VE-cadherin expression mediated by the epidermal growth factor receptor (EGFR)-extracellular signal-regulated kinase1/2 (ERK1/2) signaling pathway. However, TGF-β1 suppressed microvascular-like formation in hGL cells by downregulating VE-cadherin expression mediated by the activin receptor-like kinase (ALK)5-Sma- and Mad-related protein (SMAD)2/3/4 signaling pathway. Collectively, this study provides important insights into the underlying molecular mechanisms by which TGF-β1 and AREG differentially regulate corpus luteum formation in human ovaries.
Collapse
Affiliation(s)
- Hui Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Hsun-Ming Chang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
- *Correspondence: Hsun-Ming Chang, ; Peter C.K. Leung,
| | - Saijiao Li
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Zhendan Shi
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Peter C.K. Leung
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Hsun-Ming Chang, ; Peter C.K. Leung,
| |
Collapse
|
18
|
Ontoria-Oviedo I, Földes G, Tejedor S, Panadero J, Kitani T, Vázquez A, Wu JC, Harding SE, Sepúlveda P. Modeling Transposition of the Great Arteries with Patient-Specific Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms222413270. [PMID: 34948064 PMCID: PMC8705900 DOI: 10.3390/ijms222413270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
The dextro-transposition of the great arteries (d-TGA) is one of the most common congenital heart diseases. To identify biological processes that could be related to the development of d-TGA, we established induced pluripotent stem cell (iPSC) lines from two patients with d-TGA and from two healthy subjects (as controls) and differentiated them into endothelial cells (iPSC-ECs). iPSC-EC transcriptome profiling and bioinformatics analysis revealed differences in the expression level of genes involved in circulatory system and animal organ development. iPSC-ECs from patients with d-TGA showed impaired ability to develop tubular structures in an in vitro capillary-like tube formation assay, and interactome studies revealed downregulation of biological processes related to Notch signaling, circulatory system development and angiogenesis, pointing to alterations in vascular structure development. Our study provides an iPSC-based cellular model to investigate the etiology of d-TGA.
Collapse
Affiliation(s)
- Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
- Correspondence: (I.O.-O.); (P.S.); Tel.: +34-96-1246632 (I.O.-O.); +34-96-1246635 (P.S.)
| | - Gabor Földes
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (G.F.); (S.E.H.)
- Heart and Vascular Center, Semmelweis University, H1122 Budapest, Hungary
| | - Sandra Tejedor
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
| | - Joaquín Panadero
- IGENOMIX S.L., Edificios Europark, Parque Tecnológico, 46980 Paterna, Spain;
| | - Tomoya Kitani
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.K.); (J.C.W.)
| | - Alejandro Vázquez
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; (T.K.); (J.C.W.)
| | - Sian E. Harding
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (G.F.); (S.E.H.)
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (S.T.); (A.V.)
- Correspondence: (I.O.-O.); (P.S.); Tel.: +34-96-1246632 (I.O.-O.); +34-96-1246635 (P.S.)
| |
Collapse
|
19
|
Liu Y, Tian Y, Guo Y, Yan Z, Xue C, Wang J. DHA-enriched phosphatidylcholine suppressed angiogenesis by activating PPARγ and modulating the VEGFR2/Ras/ERK pathway in human umbilical vein endothelial cells. Food Sci Biotechnol 2021; 30:1543-1553. [PMID: 34868703 DOI: 10.1007/s10068-021-00990-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/05/2021] [Accepted: 09/28/2021] [Indexed: 10/20/2022] Open
Abstract
Docosahexaenoic acid-enriched phosphatidylcholine (DHA-PC) is a new generation of omega-3 lipids, which contains an ester bond linking DHA at the sn-2 position of phospholipid. DHA-PC has become the interest recently as its better bioavailability and anti-oxidation capacity. In this study, the anti-angiogenic effect of DHA-PC was evaluated. The capacities of proliferation, migration, tube formation of human umbilical vein endothelial cells were significantly declined after DHA-PC treatment. Furthermore, DHA-PC inhibited the neovascularization of the chick chorioallantoic membrane in vivo. Mechanism results indicated that DHA-PC enhances the expression of peroxisome proliferator-activated receptor γ (PPARγ) at transcriptional and translational level, subsequently down-regulates the VEGFR2 expression and VEGFR2-mediated downstream Ras/ERK pathway, resulting in significant reduction in proliferation and differentiation. Additionally, PPARγ-specific antagonist GW9662 partly reversed the inhibition effects of DHA-PC on tube formation and neovascularization, suggesting that DHA-PC exerts anti-angiogenesis effect through activating PPARγ. These findings indicated that DHA-PC has a great prospect of anti-tumor angiogenesis therapy.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yingying Tian
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yao Guo
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Ziyi Yan
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
20
|
Kline J, Vardarajan B, Abhyankar A, Kytömaa S, Levin B, Sobreira N, Tang A, Thomas-Wilson A, Zhang R, Jobanputra V. Embryonic lethal genetic variants and chromosomally normal pregnancy loss. Fertil Steril 2021; 116:1351-1358. [PMID: 34756330 DOI: 10.1016/j.fertnstert.2021.06.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To examine whether rare damaging genetic variants are associated with chromosomally normal pregnancy loss and estimate the magnitude of the association. DESIGN Case-control. SETTING Cases were derived from a consecutive series of karyotyped losses at one New Jersey hospital. Controls were derived from the National Database for Autism Research. PATIENT(S) Cases comprised 19 chromosomally normal loss conceptus-parent trios. Controls comprised 547 unaffected siblings of autism case-parent trios. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The rate of damaging variants in the exome (loss of function and missense-damaging) and the proportions of probands with at least one such variant among cases vs. controls. RESULTS The proportions of probands with at least one rare damaging variant were 36.8% among cases and 22.9% among controls (odds ratio, 2.0; 99% confidence interval, 0.5-7.3). No case had a variant in a known fetal anomaly gene. The proportion with variants in possibly embryonic lethal genes increased in case probands (odds ratio, 14.5; 99% confidence interval, 1.5-89.7); variants occurred in BAZ1A, FBN2, and TIMP2. CONCLUSION(S) Rare genetic variants in the conceptus may be a cause of chromosomally normal pregnancy loss. A larger sample is needed to estimate the magnitude of the association with precision and identify relevant biologic pathways.
Collapse
Affiliation(s)
- Jennie Kline
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York; Gertrude H. Sergievsky Center, Columbia University, New York, New York.
| | - Badri Vardarajan
- Gertrude H. Sergievsky Center, Columbia University, New York, New York
| | | | - Sonja Kytömaa
- Boston University School of Medicine, Boston, Massachusetts
| | - Bruce Levin
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Andrew Tang
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | | | - Ruiwei Zhang
- Life Sciences Practice, Charles River Associates, New York, New York
| | - Vaidehi Jobanputra
- Department of Pathology and Cell Biology, Columbia University, New York, New York; New York Genome Center, New York, New York
| |
Collapse
|
21
|
Horvat D, Afroze SH, Cromer WE, Pantho AF, Ashraf AHMZ, Kuehl TJ, Zawieja DC, Uddin MN. Cartiotonic steroids affect monolayer permeability in lymphatic endothelial cells. Mol Cell Biochem 2021; 476:3207-3213. [PMID: 33866492 DOI: 10.1007/s11010-021-04147-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022]
Abstract
Edema is common in preeclampsia (preE), a hypertensive disorder of pregnancy. Cardiotonic steroids (CTSs) such as marinobufagenin (MBG) are involved in the pathogenesis of preE. To assess whether CTSs are involved in the leakage of lymphatic endothelial cell (LEC), we evaluated their effect on monolayer permeability of LECs (MPLEC) in culture. A rat mesenteric LECs were treated with DMSO (vehicle), and CTSs (MBG, CINO, OUB) at concentrations of 1, 10, and 100 nM. Some LECs were pretreated with 1 μM L-NAME (N-Nitro-L-Arginine Methyl Ester) before adding 100 nM MBG or cinobufotalin (CINO). Expression of β-catenin and vascular endothelial (VE)-cadherin in CTS-treated LECs was measured by immunofluorescence and MPLEC was quantified using a fluorescence plate reader. Western blot was performed to measure β-catenin and VE-cadherin protein levels and myosin light chain 20 (MLC20) phosphorylation. MBG (≥ 1 nM) and CINO (≥ 10 nM) caused an increase (p < 0.05) in the MPLEC compared to DMSO while ouabain (OUB) had no effect. Pretreatment of LECs with 1 μM L-NAME attenuated (p < 0.05) the MPLEC. The β-catenin expression in LECs was downregulated (p < 0.05) by MBG and CINO. However, there was no effect on the LECs tight junctions for the CINO group. VE-cadherin expression was downregulated (p < 0.05) by CINO, and MLC20 phosphorylation was upregulated (p < 0.05) by MBG. We demonstrated that MBG and CINO caused an increase in the MPLEC, which were attenuated by L-NAME pretreatment. The data suggest that CTSs exert their effect via nitric-oxide-dependent signaling pathway and may be involved in vascular leak syndrome of LEC lining in preE.
Collapse
Affiliation(s)
- Darijana Horvat
- Orion Institute for Translational Medicine, Temple, TX, USA.,Emergent Biotechnologies LLC, Temple, TX, USA
| | - Syeda H Afroze
- Orion Institute for Translational Medicine, Temple, TX, USA.,Emergent Biotechnologies LLC, Temple, TX, USA
| | - Walter E Cromer
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA
| | - Ahmed F Pantho
- Orion Institute for Translational Medicine, Temple, TX, USA.,Emergent Biotechnologies LLC, Temple, TX, USA
| | | | - Thomas J Kuehl
- Orion Institute for Translational Medicine, Temple, TX, USA.,Emergent Biotechnologies LLC, Temple, TX, USA
| | - David C Zawieja
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA
| | - Mohammad Nasir Uddin
- Orion Institute for Translational Medicine, Temple, TX, USA. .,Emergent Biotechnologies LLC, Temple, TX, USA. .,Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA.
| |
Collapse
|
22
|
Wang S, Lee MP, Jones S, Liu J, Waldhaus J. Mapping the regulatory landscape of auditory hair cells from single-cell multi-omics data. Genome Res 2021; 31:1885-1899. [PMID: 33837132 DOI: 10.1101/gr.271080.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/23/2021] [Indexed: 11/25/2022]
Abstract
Auditory hair cells transduce sound to the brain and in mammals these cells reside together with supporting cells in the sensory epithelium of the cochlea, called the organ of Corti. To establish the organ's delicate function during development and differentiation, spatiotemporal gene expression is strictly controlled by chromatin accessibility and cell type-specific transcription factors, jointly representing the regulatory landscape. Bulk-sequencing technology and cellular heterogeneity obscured investigations on the interplay between transcription factors and chromatin accessibility in inner ear development. To study the formation of the regulatory landscape in hair cells, we collected single-cell chromatin accessibility profiles accompanied by single-cell RNA data from genetically labeled murine hair cells and supporting cells after birth. Using an integrative approach, we predicted cell type-specific activating and repressing functions of developmental transcription factors. Furthermore, by integrating gene expression and chromatin accessibility datasets, we reconstructed gene regulatory networks. Then, using a comparative approach, 20 hair cell-specific activators and repressors, including putative downstream target genes, were identified. Clustering of target genes resolved groups of related transcription factors and was utilized to infer their developmental functions. Finally, the heterogeneity in the single-cell data allowed us to spatially reconstruct transcriptional as well as chromatin accessibility trajectories, indicating that gradual changes in the chromatin accessibility landscape were lagging behind the transcriptional identity of hair cells along the organ's longitudinal axis. Overall, this study provides a strategy to spatially reconstruct the formation of a lineage specific regulatory landscape using a single-cell multi-omics approach.
Collapse
Affiliation(s)
- Shuze Wang
- University of Michigan, Kresge Hearing Research Institute
| | - Mary P Lee
- University of Michigan, Kresge Hearing Research Institute
| | - Scott Jones
- University of Michigan, Kresge Hearing Research Institute
| | | | - Joerg Waldhaus
- University of Michigan, Kresge Hearing Research Institute;
| |
Collapse
|
23
|
Guo Z, Mo Z. Regulation of endothelial cell differentiation in embryonic vascular development and its therapeutic potential in cardiovascular diseases. Life Sci 2021; 276:119406. [PMID: 33785330 DOI: 10.1016/j.lfs.2021.119406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
During vertebrate development, the cardiovascular system begins operating earlier than any other organ in the embryo. Endothelial cell (EC) forms the inner lining of blood vessels, and its extensive proliferation and migration are requisite for vasculogenesis and angiogenesis. Many aspects of cellular biology are involved in vasculogenesis and angiogenesis, including the tip versus stalk cell specification. Recently, epigenetics has attracted growing attention in regulating embryonic vascular development and controlling EC differentiation. Some proteins that regulate chromatin structure have been shown to be directly implicated in human cardiovascular diseases. Additionally, the roles of important EC signaling such as vascular endothelial growth factor and its receptors, angiopoietin-1 and tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-2, and transforming growth factor-β in EC differentiation during embryonic vasculature development are briefly discussed in this review. Recently, the transplantation of human induced pluripotent stem cell (iPSC)-ECs are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction. Patient-specific iPSC-derived EC is a potential new target to study differences in gene expression or response to drugs. However, clinical application of the iPSC-ECs in regenerative medicine is often limited by the challenges of maintaining cell viability and function. Therefore, novel insights into the molecular mechanisms underlying EC differentiation might provide a better understanding of embryonic vascular development and bring out more effective EC-based therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Duong CN, Nottebaum AF, Butz S, Volkery S, Zeuschner D, Stehling M, Vestweber D. Interference With ESAM (Endothelial Cell-Selective Adhesion Molecule) Plus Vascular Endothelial-Cadherin Causes Immediate Lethality and Lung-Specific Blood Coagulation. Arterioscler Thromb Vasc Biol 2020; 40:378-393. [DOI: 10.1161/atvbaha.119.313545] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective:
Vascular endothelial (VE)-cadherin is of dominant importance for the formation and stability of endothelial junctions, yet induced gene inactivation enhances vascular permeability in the lung but does not cause junction rupture. This study aims at identifying the junctional adhesion molecule, which is responsible for preventing endothelial junction rupture in the pulmonary vasculature in the absence of VE-cadherin.
Approach and Results:
We have compared the relevance of ESAM (endothelial cell-selective adhesion molecule), JAM (junctional adhesion molecule)-A, PECAM (platelet endothelial cell adhesion molecule)-1, and VE-cadherin for vascular barrier integrity in various mouse tissues. Gene inactivation of ESAM enhanced vascular permeability in the lung but not in the heart, skin, and brain. In contrast, deletion of JAM-A or PECAM-1 did not affect barrier integrity in any of these organs. Blocking VE-cadherin with antibodies caused lethality in ESAM
−/−
mice within 30 minutes but had no such effect in JAM-A
−/−
, PECAM-1
−/−
or wild-type mice. Likewise, induced gene inactivation of VE-cadherin caused rapid lethality only in the absence of ESAM. Ultrastructural analysis revealed that only combined interference with VE-cadherin and ESAM disrupted endothelial junctions and caused massive blood coagulation in the lung. Mechanistically, we could exclude a role of platelet ESAM in coagulation, changes in the expression of other junctional proteins or a contribution of cytoplasmic signaling domains of ESAM.
Conclusions:
Despite well-documented roles of JAM-A and PECAM-1 for the regulation of endothelial junctions, only for ESAM, we detected an essential role for endothelial barrier integrity in a tissue-specific way. In addition, we found that it is ESAM which prevents endothelial junction rupture in the lung when VE-cadherin is absent.
Collapse
Affiliation(s)
- Cao Nguyen Duong
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Astrid F. Nottebaum
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Stefan Butz
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Stefan Volkery
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy and Flow Cytometry Unit (D.Z., M.S.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Martin Stehling
- Electron Microscopy and Flow Cytometry Unit (D.Z., M.S.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dietmar Vestweber
- From the Department of Vascular Cell Biology (C.N.D., A.F.N., S.B., S.V., D.V.), Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
25
|
Takase Y, Takahashi Y. Blood flow-mediated gene transfer and siRNA-knockdown in the developing vasculature in a spatio-temporally controlled manner in chicken embryos. Dev Biol 2019; 456:8-16. [PMID: 31400307 DOI: 10.1016/j.ydbio.2019.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 07/23/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
We describe a method by which early developing vasculature can be gene-manipulated independently of the heart in a spatio-temporally controlled manner. Lipofectamine 2000 or 3000, an easy-to-use lipid reagent, has been found to yield a high efficiency of transfection when co-injected with GFP DNA within a critical range of lipid concentration. By exploiting developmentally changing patterns of vasculature and blood flow, we have succeed in controlling the site of transfection: injection with a lipid-DNA cocktail into the heart before or after the blood circulation starts results in a limited and widely spread patterns of transfection, respectively. Furthermore, a cocktail injection into the right dorsal aorta leads to transgenesis of the right half of embryonic vasculature. In addition, this method combined with the siRNA technique has allowed, for the first time, to knockdown the endogenous expression of VE-cadherin (also called Cdh5), which has been implicated in assembly of nasant blood vessels: when Cah5 siRNA is injected into the right dorsal aorta, pronounced defects in the right half of vasculature are observed without heart defects. Whereas infusion-mediated gene transfection method has previously been reported using lipid reagents that were elaborately prepared on their own, Lipofectamine is an easy-use reagent with no requirement of special expertise. The methods reported here would overcome shortcomings of conventional vascular-transgenic animals, such as mice and zebrafish, in which pan-endothelial enhancer-driven transgenesis often leads to the heart malformation, which, in turn, indirectly affects peripheral vasculature due to flow defects. Since a variety of subtypes in vasculature have increasingly been appreciated, the spatio-temporally controllable gene manipulation described in this study offers a powerful tool to understand how the vasculature is established at the molecular level.
Collapse
Affiliation(s)
- Yuta Takase
- Mathematics-based Creation of Science Program (MACS), Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto, 606-8502, Japan; Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto, 606-8502, Japan; AMED Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda-ku, Tokyo, 100-0004, Japan.
| |
Collapse
|
26
|
Yu W, Yang L, Li T, Zhang Y. Cadherin Signaling in Cancer: Its Functions and Role as a Therapeutic Target. Front Oncol 2019; 9:989. [PMID: 31637214 PMCID: PMC6788064 DOI: 10.3389/fonc.2019.00989] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022] Open
Abstract
Cadherin family includes lists of transmembrane glycoproteins which mediate calcium-dependent cell-cell adhesion. Cadherin-mediated adhesion regulates cell growth and differentiation throughout life. Through the establishment of the cadherin-catenin complex, cadherins provide normal cell-cell adhesion and maintain homeostatic tissue architecture. In the process of cell recognition and adhesion, cadherins act as vital participators. As results, the disruption of cadherin signaling has significant implications on tumor formation and progression. Altered cadherin expression plays a vital role in tumorigenesis, tumor progression, angiogenesis, and tumor immune response. Based on ongoing research into the role of cadherin signaling in malignant tumors, cadherins are now being considered as potential targets for cancer therapies. This review will demonstrate the mechanisms of cadherin involvement in tumor progression, and consider the clinical significance of cadherins as therapeutic targets.
Collapse
Affiliation(s)
- Weina Yu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Ting Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Yang Y, Cha B, Motawe ZY, Srinivasan RS, Scallan JP. VE-Cadherin Is Required for Lymphatic Valve Formation and Maintenance. Cell Rep 2019; 28:2397-2412.e4. [PMID: 31461654 PMCID: PMC6743082 DOI: 10.1016/j.celrep.2019.07.072] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/18/2019] [Accepted: 07/19/2019] [Indexed: 11/16/2022] Open
Abstract
The lymphatic vasculature requires intraluminal valves to maintain forward lymph flow. Lymphatic valves form and are constantly maintained by oscillatory fluid flow throughout life, yet the earliest steps of how lymphatic endothelial cells are able to respond to fluid shear stress remain unknown. Here, we show that the adherens junction protein VE-cadherin is required for the upregulation of valve-specific transcription factors. Conditional deletion of VE-cadherin in vivo prevented valve formation in the embryo and caused postnatal regression of nearly all lymphatic valves in multiple tissues. Since VE-cadherin is known to signal through β-catenin and the VEGFR/AKT pathway, each pathway was probed. Expression of a constitutively active β-catenin mutant or direct pharmacologic activation of AKT in vivo significantly rescued valve regression in the VE-cadherin-deficient lymphatic vessels. In conclusion, VE-cadherin-dependent signaling is required for lymphatic valve formation and maintenance and therapies to augment downstream pathways hold potential to treat lymphedema in patients.
Collapse
Affiliation(s)
- Ying Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Zeinab Y Motawe
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
28
|
Braun LJ, Zinnhardt M, Vockel M, Drexler HC, Peters K, Vestweber D. VE-PTP inhibition stabilizes endothelial junctions by activating FGD5. EMBO Rep 2019; 20:e47046. [PMID: 31267715 PMCID: PMC6607018 DOI: 10.15252/embr.201847046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 04/01/2019] [Accepted: 04/18/2019] [Indexed: 01/06/2023] Open
Abstract
Inhibition of VE-PTP, an endothelial receptor-type tyrosine phosphatase, triggers phosphorylation of the tyrosine kinase receptor Tie-2, which leads to the suppression of inflammation-induced vascular permeability. Analyzing the underlying mechanism, we show here that inhibition of VE-PTP and activation of Tie-2 induce tyrosine phosphorylation of FGD5, a GTPase exchange factor (GEF) for Cdc42, and stimulate its translocation to cell contacts. Interfering with the expression of FGD5 blocks the junction-stabilizing effect of VE-PTP inhibition in vitro and in vivo. Likewise, FGD5 is required for strengthening cortical actin bundles and inhibiting radial stress fiber formation, which are each stimulated by VE-PTP inhibition. We identify Y820 of FGD5 as the direct substrate for VE-PTP. The phosphorylation of FGD5-Y820 is required for the stabilization of endothelial junctions and for the activation of Cdc42 by VE-PTP inhibition but is dispensable for the recruitment of FGD5 to endothelial cell contacts. Thus, activation of FGD5 is a two-step process that comprises membrane recruitment and phosphorylation of Y820. These steps are necessary for the junction-stabilizing effect stimulated by VE-PTP inhibition and Tie-2 activation.
Collapse
Affiliation(s)
- Laura J Braun
- Max Planck Institute of Molecular BiomedicineMünsterGermany
| | | | - Matthias Vockel
- Max Planck Institute of Molecular BiomedicineMünsterGermany
- Present address:
Institute for Human GeneticsUniversity of MünsterMünsterGermany
| | | | | | | |
Collapse
|
29
|
Rodriguez D, Nourizadeh S, De Tomaso AW. The biology of the extracorporeal vasculature of Botryllus schlosseri. Dev Biol 2019; 448:309-319. [PMID: 30760410 DOI: 10.1016/j.ydbio.2018.10.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/29/2018] [Accepted: 10/17/2018] [Indexed: 02/08/2023]
Abstract
The extracorporeal vasculature of the colonial ascidian Botryllus schlosseri plays a key role in several biological processes: transporting blood, angiogenesis, regeneration, self-nonself recognition, and parabiosis. The vasculature also interconnects all individuals in a colony and is composed of a single layer of ectodermally-derived cells. These cells form a tube with the basal lamina facing the lumen, and the apical side facing an extracellular matrix that consists of cellulose and other proteins, known as the tunic. Vascular tissue is transparent and can cover several square centimeters, which is much larger than any single individual within the colony. It forms a network that ramifies and expands to the perimeter of each colony and terminates into oval-shaped protrusions known as ampullae. Botryllus individuals replace themselves through a weekly budding cycle, and vasculature is added to ensure the interconnection of each new individual, thus there is continuous angiogenesis occurring naturally. The vascular tissue itself is highly regenerative; surgical removal of the ampullae and peripheral vasculature triggers regrowth within 24-48 h, which includes forming new ampullae. When two individuals, whether in the wild or in the lab, come into close contact and their ampullae touch, they can either undergo parabiosis through anastomosing vessels, or reject vascular fusion. The vasculature is easily manipulated by direct means such as microinjections, microsurgeries, and pharmacological reagents. Its transparent nature allows for in vivo analysis by bright field and fluorescence microscopy. Here we review the techniques and approaches developed to study the different biological processes that involve the extracorporeal vasculature.
Collapse
Affiliation(s)
- Delany Rodriguez
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA.
| | - Shane Nourizadeh
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Anthony W De Tomaso
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| |
Collapse
|
30
|
Blatchley MR, Hall F, Wang S, Pruitt HC, Gerecht S. Hypoxia and matrix viscoelasticity sequentially regulate endothelial progenitor cluster-based vasculogenesis. SCIENCE ADVANCES 2019; 5:eaau7518. [PMID: 30906859 PMCID: PMC6426463 DOI: 10.1126/sciadv.aau7518] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/30/2019] [Indexed: 05/14/2023]
Abstract
Vascular morphogenesis is the formation of endothelial lumenized networks. Cluster-based vasculogenesis of endothelial progenitor cells (EPCs) has been observed in animal models, but the underlying mechanism is unknown. Here, using O2-controllabe hydrogels, we unveil the mechanism by which hypoxia, co-jointly with matrix viscoelasticity, induces EPC vasculogenesis. When EPCs are subjected to a 3D hypoxic gradient ranging from <2 to 5%, they rapidly produce reactive oxygen species that up-regulate proteases, most notably MMP-1, which degrade the surrounding extracellular matrix. EPC clusters form and expand as the matrix degrades. Cell-cell interactions, including those mediated by VE-cadherin, integrin-β2, and ICAM-1, stabilize the clusters. Subsequently, EPC sprouting into the stiffer, intact matrix leads to vascular network formation. In vivo examination further corroborated hypoxia-driven clustering of EPCs. Overall, this is the first description of how hypoxia mediates cluster-based vasculogenesis, advancing our understanding toward regulating vascular development as well as postnatal vasculogenesis in regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Michael R Blatchley
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology and Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Franklyn Hall
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology and Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Songnan Wang
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology and Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hawley C Pruitt
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology and Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology and Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
31
|
Nair SK, Basu S, Sen B, Lin MH, Kumar AN, Yuan Y, Cullen PJ, Sarkar D. Colloidal Gels with Tunable Mechanomorphology Regulate Endothelial Morphogenesis. Sci Rep 2019; 9:1072. [PMID: 30705322 PMCID: PMC6355882 DOI: 10.1038/s41598-018-37788-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/13/2018] [Indexed: 12/04/2022] Open
Abstract
Endothelial morphogenesis into capillary networks is dependent on the matrix morphology and mechanical properties. In current 3D gels, these two matrix features are interdependent and their distinct roles in endothelial organization are not known. Thus, it is important to decouple these parameters in the matrix design. Colloidal gels can be engineered to regulate the microstructural morphology and mechanics in an independent manner because colloidal gels are formed by the aggregation of particles into a self-similar 3D network. In this work, gelatin based colloidal gels with distinct mechanomorphology were developed by engineering the electrostatic interaction mediated aggregation of particles. By altering the mode of aggregation, colloidal gels showed either compact dense microstructure or tenuous strand-like networks, and the matrix stiffness was controlled independently by varying the particle fraction. Endothelial Cell (EC) networks were favored in tenuous strand-like microstructure through increased cell-matrix and cell-cell interactions, while compact dense microstructure inhibited the networks. For a given microstructure, as the gel stiffness was increased, the extent of EC network was reduced. This result demonstrates that 3D matrix morphology and mechanics provide distinct signals in a bidirectional manner during EC network formation. Colloidal gels can be used to interrogate the angiogenic responses of ECs and can be developed as a biomaterial for vascularization.
Collapse
Affiliation(s)
- Smruti K Nair
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Sukanya Basu
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Ballari Sen
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Meng-Hsuan Lin
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Arati N Kumar
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Yuan Yuan
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Paul J Cullen
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Debanjan Sarkar
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA.
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA.
| |
Collapse
|
32
|
The precise molecular signals that control endothelial cell-cell adhesion within the vessel wall. Biochem Soc Trans 2018; 46:1673-1680. [PMID: 30514769 PMCID: PMC6299237 DOI: 10.1042/bst20180377] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 12/23/2022]
Abstract
Endothelial cell–cell adhesion within the wall of the vasculature controls a range of physiological processes, such as growth, integrity and barrier function. The adhesive properties of endothelial cells are tightly controlled by a complex cascade of signals transmitted from the surrounding environment or from within the cells themselves, with the dynamic nature of cellular adhesion and the regulating signalling networks now beginning to be appreciated. Here, we summarise the current knowledge of the mechanisms controlling endothelial cell–cell adhesion in the developing and mature blood vasculature.
Collapse
|
33
|
Hägerling R, Hoppe E, Dierkes C, Stehling M, Makinen T, Butz S, Vestweber D, Kiefer F. Distinct roles of VE-cadherin for development and maintenance of specific lymph vessel beds. EMBO J 2018; 37:embj.201798271. [PMID: 30297530 DOI: 10.15252/embj.201798271] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
Endothelial cells line blood and lymphatic vessels and form intercellular junctions, which preserve vessel structure and integrity. The vascular endothelial cadherin, VE-cadherin, mediates endothelial adhesion and is indispensible for blood vessel development and permeability regulation. However, its requirement for lymphatic vessels has not been addressed. During development, VE-cadherin deletion in lymphatic endothelial cells resulted in abortive lymphangiogenesis, edema, and prenatal death. Unexpectedly, inducible postnatal or adult deletion elicited vessel bed-specific responses. Mature dermal lymph vessels resisted VE-cadherin loss and maintained button junctions, which was associated with an upregulation of junctional molecules. Very different, mesenteric lymphatic collectors deteriorated and formed a strongly hyperplastic layer of lymphatic endothelial cells on the mesothelium. This massive hyperproliferation may have been favored by high mesenteric VEGF-C expression and was associated with VEGFR-3 phosphorylation and upregulation of the transcriptional activator TAZ Finally, intestinal lacteals fragmented into cysts or became highly distended possibly as a consequence of the mesenteric defects. Taken together, we demonstrate here the importance of VE-cadherin for lymphatic vessel development and maintenance, which is however remarkably vessel bed-specific.
Collapse
Affiliation(s)
- René Hägerling
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Esther Hoppe
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany
| | - Cathrin Dierkes
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Taija Makinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Butz
- Department Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dietmar Vestweber
- Department Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,DFG Cluster of Excellence 1003 "CiM - Cells in Motion", Münster, Germany
| | - Friedemann Kiefer
- Mammalian Cell Signaling Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany .,European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.,DFG Cluster of Excellence 1003 "CiM - Cells in Motion", Münster, Germany
| |
Collapse
|
34
|
Plein A, Fantin A, Denti L, Pollard JW, Ruhrberg C. Erythro-myeloid progenitors contribute endothelial cells to blood vessels. Nature 2018; 562:223-228. [PMID: 30258231 PMCID: PMC6289247 DOI: 10.1038/s41586-018-0552-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 08/17/2018] [Indexed: 12/16/2022]
Abstract
The earliest blood vessels in mammalian embryos are formed when endothelial cells differentiate from angioblasts and coalesce into tubular networks. Thereafter, the endothelium is thought to expand solely by proliferation of pre-existing endothelial cells. Here we show that a complementary source of endothelial cells is recruited into pre-existing vasculature after differentiation from the earliest precursors of erythrocytes, megakaryocytes and macrophages, the erythro-myeloid progenitors (EMPs) that are born in the yolk sac. A first wave of EMPs contributes endothelial cells to the yolk sac endothelium, and a second wave of EMPs colonizes the embryo and contributes endothelial cells to intraembryonic endothelium in multiple organs, where they persist into adulthood. By demonstrating that EMPs constitute a hitherto unrecognized source of endothelial cells, we reveal that embryonic blood vascular endothelium expands in a dual mechanism that involves both the proliferation of pre-existing endothelial cells and the incorporation of endothelial cells derived from haematopoietic precursors.
Collapse
Affiliation(s)
- Alice Plein
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
35
|
CMTM4 regulates angiogenesis by promoting cell surface recycling of VE-cadherin to endothelial adherens junctions. Angiogenesis 2018; 22:75-93. [PMID: 30097810 PMCID: PMC6510885 DOI: 10.1007/s10456-018-9638-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/21/2018] [Indexed: 02/06/2023]
Abstract
Vascular endothelial (VE) cadherin is a key component of endothelial adherens junctions (AJs) and plays an important role in maintaining vascular integrity. Endocytosis of VE-cadherin regulates junctional strength and a decrease of surface VE-cadherin reduces vascular stability. However, disruption of AJs is also a requirement for vascular sprouting. Identifying novel regulators of endothelial endocytosis could enhance our understanding of angiogenesis. Here, we evaluated the angiogenic potential of (CKLF-like MARVEL transmembrane domain 4) CMTM4 and assessed in which molecular pathway CMTM4 is involved during angiogenesis. Using a 3D vascular assay composed of GFP-labeled HUVECs and dsRED-labeled pericytes, we demonstrated in vitro that siRNA-mediated CMTM4 silencing impairs vascular sprouting. In vivo, CMTM4 silencing by morpholino injection in zebrafish larvae inhibits intersomitic vessel growth. Intracellular staining revealed that CMTM4 colocalizes with Rab4+ and Rab7+ vesicles, both markers of the endocytic trafficking pathway. CMTM4 colocalizes with both membrane-bound and internalized VE-cadherin. Adenovirus-mediated CMTM4 overexpression enhances the endothelial endocytic pathway, in particular the rapid recycling pathway, shown by an increase in early endosomal antigen-1 positive (EEA1+), Rab4+, Rab11+ , and Rab7+ vesicles. CMTM4 overexpression enhances membrane-bound VE-cadherin internalization, whereas CMTM4 knockdown decreases internalization of VE-cadherin. CMTM4 overexpression promotes endothelial barrier function, shown by an increase in recovery of transendothelial electrical resistance (TEER) after thrombin stimulation. We have identified in this study a novel regulatory function for CMTM4 in angiogenesis. CMTM4 plays an important role in the turnover of membrane-bound VE-cadherin at AJs, mediating endothelial barrier function and controlling vascular sprouting.
Collapse
|
36
|
Szymborska A, Gerhardt H. Hold Me, but Not Too Tight-Endothelial Cell-Cell Junctions in Angiogenesis. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029223. [PMID: 28851748 DOI: 10.1101/cshperspect.a029223] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endothelial cell-cell junctions must perform seemingly incompatible tasks during vascular development-providing stable connections that prevent leakage, while allowing dynamic cellular rearrangements during sprouting, anastomosis, lumen formation, and functional remodeling of the vascular network. This review aims to highlight recent insights into the molecular mechanisms governing endothelial cell-cell adhesion in the context of vascular development.
Collapse
Affiliation(s)
- Anna Szymborska
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,Vascular Patterning Laboratory, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, 3000 Leuven, Belgium.,DZHK (German Centre for Cardiovascular Research), partner site Berlin.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| |
Collapse
|
37
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
38
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
39
|
Mu LM, Ju RJ, Liu R, Bu YZ, Zhang JY, Li XQ, Zeng F, Lu WL. Dual-functional drug liposomes in treatment of resistant cancers. Adv Drug Deliv Rev 2017; 115:46-56. [PMID: 28433739 DOI: 10.1016/j.addr.2017.04.006] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/26/2022]
Abstract
Efficacy of regular chemotherapy is significantly hampered by multidrug resistance (MDR) and severe systemic toxicity. The reduced toxicity has been evidenced after administration of drug liposomes, consisting of the first generation of regular drug liposomes, the second generation of long-circulation drug liposomes, and the third generation of targeting drug liposomes. However, MDR of cancers remains as an unsolved issue. The objective of this article is to review the dual-functional drug liposomes, which demonstrate the potential in overcoming MDR. Herein, dual-functional drug liposomes are referring to the drug-containing phospholipid bilayer vesicles that possess a dual-function of providing the basic efficacy of drug and the extended effect of the drug carrier. They exhibit unique roles in treatment of resistant cancer via circumventing drug efflux caused by adenosine triphosphate binding cassette (ABC) transporters, eliminating cancer stem cells, destroying mitochondria, initiating apoptosis, regulating autophagy, destroying supply channels, utilizing microenvironment, and silencing genes of the resistant cancer. As the prospect of an estimation, dual-functional drug liposomes would exhibit more strength in their extended function, hence deserving further investigation for clinical validation.
Collapse
|
40
|
Chrifi I, Louzao-Martinez L, Brandt M, van Dijk CGM, Burgisser P, Zhu C, Kros JM, Duncker DJ, Cheng C. CMTM3 (CKLF-Like Marvel Transmembrane Domain 3) Mediates Angiogenesis by Regulating Cell Surface Availability of VE-Cadherin in Endothelial Adherens Junctions. Arterioscler Thromb Vasc Biol 2017; 37:1098-1114. [PMID: 28428220 DOI: 10.1161/atvbaha.116.308792] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/10/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Decrease in VE-cadherin adherens junctions reduces vascular stability, whereas disruption of adherens junctions is a requirement for neovessel sprouting during angiogenesis. Endocytosis plays a key role in regulating junctional strength by altering bioavailability of cell surface proteins, including VE-cadherin. Identification of new mediators of endothelial endocytosis could enhance our understanding of angiogenesis. Here, we assessed the function of CMTM3 (CKLF-like MARVEL transmembrane domain 3), which we have previously identified as highly expressed in Flk1+ endothelial progenitor cells during embryonic development. APPROACH AND RESULTS Using a 3-dimensional coculture of human umbilical vein endothelial cells-GFP (green fluorescent protein) and pericytes-RFP (red fluorescent protein), we demonstrated that siRNA-mediated CMTM3 silencing in human umbilical vein endothelial cells impairs angiogenesis. In vivo CMTM3 inhibition by morpholino injection in developing zebrafish larvae confirmed that CMTM3 expression is required for vascular sprouting. CMTM3 knockdown in human umbilical vein endothelial cells does not affect proliferation or migration. Intracellular staining demonstrated that CMTM3 colocalizes with early endosome markers EEA1 (early endosome marker 1) and Clathrin+ vesicles and with cytosolic VE-cadherin in human umbilical vein endothelial cells. Adenovirus-mediated CMTM3 overexpression enhances endothelial endocytosis, shown by an increase in Clathrin+, EEA1+, Rab11+, Rab5+, and Rab7+ vesicles. CMTM3 overexpression enhances, whereas CMTM3 knockdown decreases internalization of cell surface VE-cadherin in vitro. CMTM3 promotes loss of endothelial barrier function in thrombin-induced responses, shown by transendothelial electric resistance measurements in vitro. CONCLUSIONS In this study, we have identified a new regulatory function for CMTM3 in angiogenesis. CMTM3 is involved in VE-cadherin turnover and is a regulator of the cell surface pool of VE-cadherin. Therefore, CMTM3 mediates cell-cell adhesion at adherens junctions and contributes to the control of vascular sprouting.
Collapse
Affiliation(s)
- Ihsan Chrifi
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Laura Louzao-Martinez
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Maarten Brandt
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Christian G M van Dijk
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Petra Burgisser
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Changbin Zhu
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Johan M Kros
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Dirk J Duncker
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Caroline Cheng
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.).
| |
Collapse
|
41
|
Sauteur L, Affolter M, Belting HG. Distinct and redundant functions of Esama and VE-cadherin during vascular morphogenesis. Development 2017; 144:1554-1565. [PMID: 28264837 DOI: 10.1242/dev.140038] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 02/28/2017] [Indexed: 01/08/2023]
Abstract
The cardiovascular system forms during early embryogenesis and adapts to embryonic growth by sprouting angiogenesis and vascular remodeling. These processes require fine-tuning of cell-cell adhesion to maintain and re-establish endothelial contacts, while allowing cell motility. We have compared the contribution of two endothelial cell-specific adhesion proteins, VE-cadherin (VE-cad/Cdh5) and Esama (endothelial cell-selective adhesion molecule a), during angiogenic sprouting and blood vessel fusion (anastomosis) in the zebrafish embryo by genetic analyses. Different combinations of mutant alleles can be placed into a phenotypic series with increasing defects in filopodial contact formation. Contact formation in esama mutants appears similar to wild type, whereas esama-/-; ve-cad+/- and ve-cad single mutants exhibit intermediate phenotypes. The lack of both proteins interrupts filopodial interaction completely. Furthermore, double mutants do not form a stable endothelial monolayer, and display intrajunctional gaps, dislocalization of Zo-1 and defects in apical-basal polarization. In summary, VE-cadherin and Esama have distinct and redundant functions during blood vessel morphogenesis, and both adhesion proteins are central to endothelial cell recognition during anastomosis.
Collapse
Affiliation(s)
- Loïc Sauteur
- Biozentrum der Universität Basel, Klingelbergstrasse 70, Basel CH-4056, Switzerland
| | - Markus Affolter
- Biozentrum der Universität Basel, Klingelbergstrasse 70, Basel CH-4056, Switzerland
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Klingelbergstrasse 70, Basel CH-4056, Switzerland
| |
Collapse
|
42
|
Gu W, Zhan H, Zhou XY, Yao L, Yan M, Chen A, Liu J, Ren X, Zhang X, Liu JX, Liu G. MicroRNA-22 regulates inflammation and angiogenesisviatargeting VE-cadherin. FEBS Lett 2017; 591:513-526. [DOI: 10.1002/1873-3468.12565] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 11/10/2022]
Affiliation(s)
- Wei Gu
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Huihui Zhan
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Xin-Ying Zhou
- Key Laboratory of Fresh Water Animal Breeding; College of Fisheries; Huazhong Agricultural University; Wuhan Hubei China
| | - Lun Yao
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Meiping Yan
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Ao Chen
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Jie Liu
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Xiaojiao Ren
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Xinhua Zhang
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| | - Jing-Xia Liu
- Key Laboratory of Fresh Water Animal Breeding; College of Fisheries; Huazhong Agricultural University; Wuhan Hubei China
| | - Guoquan Liu
- Department of Basic Veterinary Medicine; College of Animal Science and Veterinary Medicine; Huazhong Agricultural University; Wuhan Hubei China
| |
Collapse
|
43
|
Palm MM, Dallinga MG, van Dijk E, Klaassen I, Schlingemann RO, Merks RMH. Computational Screening of Tip and Stalk Cell Behavior Proposes a Role for Apelin Signaling in Sprout Progression. PLoS One 2016; 11:e0159478. [PMID: 27828952 PMCID: PMC5102492 DOI: 10.1371/journal.pone.0159478] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/24/2016] [Indexed: 12/30/2022] Open
Abstract
Angiogenesis involves the formation of new blood vessels by sprouting or splitting of existing blood vessels. During sprouting, a highly motile type of endothelial cell, called the tip cell, migrates from the blood vessels followed by stalk cells, an endothelial cell type that forms the body of the sprout. To get more insight into how tip cells contribute to angiogenesis, we extended an existing computational model of vascular network formation based on the cellular Potts model with tip and stalk differentiation, without making a priori assumptions about the differences between tip cells and stalk cells. To predict potential differences, we looked for parameter values that make tip cells (a) move to the sprout tip, and (b) change the morphology of the angiogenic networks. The screening predicted that if tip cells respond less effectively to an endothelial chemoattractant than stalk cells, they move to the tips of the sprouts, which impacts the morphology of the networks. A comparison of this model prediction with genes expressed differentially in tip and stalk cells revealed that the endothelial chemoattractant Apelin and its receptor APJ may match the model prediction. To test the model prediction we inhibited Apelin signaling in our model and in an in vitro model of angiogenic sprouting, and found that in both cases inhibition of Apelin or of its receptor APJ reduces sprouting. Based on the prediction of the computational model, we propose that the differential expression of Apelin and APJ yields a "self-generated" gradient mechanisms that accelerates the extension of the sprout.
Collapse
Affiliation(s)
- Margriet M. Palm
- Life Sciences Group, Centrum Wiskunde & Informatica, Amsterdam, the Netherlands
| | | | - Erik van Dijk
- Life Sciences Group, Centrum Wiskunde & Informatica, Amsterdam, the Netherlands
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Roeland M. H. Merks
- Life Sciences Group, Centrum Wiskunde & Informatica, Amsterdam, the Netherlands
- Mathematical Institute, Leiden University, Leiden, the Netherlands
| |
Collapse
|
44
|
Souvannakitti D, Peerapen P, Thongboonkerd V. Hypobaric hypoxia down-regulated junctional protein complex: Implications to vascular leakage. Cell Adh Migr 2016; 11:360-366. [PMID: 27627890 DOI: 10.1080/19336918.2016.1225633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Acute mountain sickness (AMS) can cause capillary hyper-permeability and vasogenic edema. However, its underlying mechanisms remained unclear and there is no previous in vitro study on AMS. We therefore conducted an in vitro study and examined whether continuous hypobaric hypoxia (CHH) could alter expression of junctional protein complex of vascular endothelial cells, causing hyper-permeabilization. EA.hy926 human endothelial cells were exposed to either CHH or normoxia for up to 24 h. Flow cytometry using annexin V/propidium iodide co-staining demonstrated that cell death had no significant difference at 12-h, but was increased by CHH at 24-h. Transendothelial resistance (TER) of endothelial cell monolayer was progressively decreased by CHH from 1-h to 24-h. Western blot analysis and immunofluorescence study demonstrated decreased expression levels of VE-cadherin, PECAM-1 and ZO-1 junctional proteins at both 12-h and 24-h exposure time-points. Interestingly, while the main form of ZO-1 (220 kDa) was decreased, its degraded form (100 kDa) was increased by 24-h CHH that might be linked to the increased cell death. Our data have demonstrated that CHH caused vascular endothelial hyper-permeability and defective junctional protein complex by reducing expression levels of VE-cadherin, PECAM-1, and ZO-1. Taken together, these data may explain pathophysiology underlying vascular hyper-permeability in AMS.
Collapse
Affiliation(s)
- Dangjai Souvannakitti
- a Department of Physiology , Phramongkutklao College of Medicine , Bangkok , Thailand
| | - Paleerath Peerapen
- b Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University , Bangkok , Thailand.,c Center for Research in Complex Systems Science (CRCSS), Mahidol University , Bangkok , Thailand
| | - Visith Thongboonkerd
- b Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University , Bangkok , Thailand.,c Center for Research in Complex Systems Science (CRCSS), Mahidol University , Bangkok , Thailand
| |
Collapse
|
45
|
Jenny Zhou H, Qin L, Zhang H, Tang W, Ji W, He Y, Liang X, Wang Z, Yuan Q, Vortmeyer A, Toomre D, Fuh G, Yan M, Kluger MS, Wu D, Min W. Endothelial exocytosis of angiopoietin-2 resulting from CCM3 deficiency contributes to cerebral cavernous malformation. Nat Med 2016; 22:1033-1042. [PMID: 27548575 PMCID: PMC5014607 DOI: 10.1038/nm.4169] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 07/21/2016] [Indexed: 12/14/2022]
Abstract
Cerebral cavernous malformations (CCMs) are vascular malformations that affect the central nervous system and result in cerebral hemorrhage, seizure and stroke. CCMs arise from loss-of-function mutations in one of three genes: KRIT1 (also known as CCM1), CCM2 or PDCD10 (also known as CCM3). PDCD10 mutations in humans often result in a more severe form of the disease relative to mutations in the other two CCM genes, and PDCD10-knockout mice show severe defects, the mechanistic basis for which is unclear. We have recently reported that CCM3 regulates exocytosis mediated by the UNC13 family of exocytic regulatory proteins. Here, in investigating the role of endothelial cell exocytosis in CCM disease progression, we found that CCM3 suppresses UNC13B- and vesicle-associated membrane protein 3 (VAMP3)-dependent exocytosis of angiopoietin 2 (ANGPT2) in brain endothelial cells. CCM3 deficiency in endothelial cells augments the exocytosis and secretion of ANGPT2, which is associated with destabilized endothelial cell junctions, enlarged lumen formation and endothelial cell-pericyte dissociation. UNC13B deficiency, which blunts ANGPT2 secretion from endothelial cells, or treatment with an ANGPT2-neutralizing antibody normalizes the defects in the brain and retina caused by endothelial-cell-specific CCM3 deficiency, including the disruption of endothelial cell junctions, vessel dilation and pericyte dissociation. Thus, enhanced secretion of ANGPT2 in endothelial cells contributes to the progression of CCM disease, providing a new therapeutic approach for treating this devastating pathology.
Collapse
Affiliation(s)
- Huanjiao Jenny Zhou
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lingfeng Qin
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Haifeng Zhang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Wenwen Tang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Weidong Ji
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangzhou Darron Medscience, Co. Ltd, Guangzhou, China
| | - Yun He
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Department of Toxicology, School of Public Health, Sun Yat-sen University of Medical Sciences, Guangzhou, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zongren Wang
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianying Yuan
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Alexander Vortmeyer
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Derek Toomre
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Germaine Fuh
- Department of Antibody Engineering, Genentech Inc, South San Francisco, CA
| | - Minghong Yan
- Department of Molecular Oncology, Genentech Inc, South San Francisco, CA
| | - Martin S. Kluger
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Dianqing Wu
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Wang Min
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangzhou Darron Medscience, Co. Ltd, Guangzhou, China
| |
Collapse
|
46
|
Chatterjee I, Baruah J, Lurie EE, Wary KK. Endothelial lipid phosphate phosphatase-3 deficiency that disrupts the endothelial barrier function is a modifier of cardiovascular development. Cardiovasc Res 2016; 111:105-18. [PMID: 27125875 PMCID: PMC4909162 DOI: 10.1093/cvr/cvw090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/21/2016] [Indexed: 01/08/2023] Open
Abstract
Aims Lipid phosphate phosphatase-3 (LPP3) is expressed at high levels in endothelial cells (ECs). Although LPP3 is known to hydrolyse the phosphate group from lysolipids such as spingosine-1-phosphate and its structural homologues, the function of Lpp3 in ECs is not completely understood. In this study, we investigated how tyrosine-protein kinase receptor (TEK or Tie2) promoter–dependent deletion of Lpp3 alters EC activities. Methods and results Lpp3fl/fl mice were crossed with the tg.Tie2Cre transgenic line. Vasculogenesis occurred normally in embryos with Tie2Cre-mediated deletion of Lpp3 (called Lpp3ECKO), but embryonic lethality occurred in two waves, the first wave between E8.5 and E10.5, while the second between E11.5 and E13.5. Lethality in Lpp3ECKO embryos after E11.5 was accompanied by vascular leakage and haemorrhage, which likely resulted in insufficient cardiovascular development. Analyses of haematoxylin- and eosin-stained heart sections from E11.5 Lpp3ECKO embryos showed insufficient heart growth associated with decreased trabeculation, reduced growth of the compact wall, and absence of cardiac cushions. Staining followed by microscopic analyses of Lpp3ECKO embryos revealed the presence of apoptotic ECs. Furthermore, Lpp3-deficient ECs showed decreased gene expression and protein levels of Cyclin-D1, VE-cadherin, Fibronectin, Klf2, and Klf4. To determine the underlying mechanisms of vascular leakage and barrier disruption, we performed knockdown and rescue experiments in cultured ECs. LPP3 knockdown decreased transendothelial electrical resistance and increased permeability. Re-expression of β-catenin cDNA in LPP3-knockdown ECs partially restored the effect of the LPP3 loss, whereas re-expression of p120ctn cDNA did not. Conclusion These findings demonstrate the essential roles of LPP3 in the maturation of EC barrier integrity and normal cardiovascular development.
Collapse
Affiliation(s)
- Ishita Chatterjee
- Department of Pharmacology, University of Illinois, 835 S. Wolcott Avenue, Room E403, Chicago, IL 60612, USA
| | - Jugajyoti Baruah
- Department of Pharmacology, University of Illinois, 835 S. Wolcott Avenue, Room E403, Chicago, IL 60612, USA
| | - Erin E Lurie
- Department of Pharmacology, University of Illinois, 835 S. Wolcott Avenue, Room E403, Chicago, IL 60612, USA
| | - Kishore K Wary
- Department of Pharmacology, University of Illinois, 835 S. Wolcott Avenue, Room E403, Chicago, IL 60612, USA
| |
Collapse
|
47
|
Goonoo N, Bhaw-Luximon A, Passanha P, Esteves SR, Jhurry D. Third generation poly(hydroxyacid) composite scaffolds for tissue engineering. J Biomed Mater Res B Appl Biomater 2016; 105:1667-1684. [PMID: 27080439 DOI: 10.1002/jbm.b.33674] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/10/2016] [Accepted: 03/20/2016] [Indexed: 12/13/2022]
Abstract
Bone tissue engineering based on scaffolds is quite a complex process as a whole gamut of criteria needs to be satisfied to promote cellular attachment, proliferation and differentiation: biocompatibility, right surface properties, adequate mechanical performance, controlled bioresorbability, osteoconductivity, angiogenic cues, and vascularization. Third generation scaffolds are more of composite types to maximize biological-mechanical-chemical properties. In the present review, our focus is on the performance of micro-organism-derived polyhydroxyalkanoates (PHAs)-polyhydroxybutyrate (PHB) and polyhydroxybutyrate-co-valerate (PHBV)-composite scaffolds with ceramics and natural polymers for tissue engineering applications with emphasis on bone tissue. We particularly emphasize on how material properties of the composites affect scaffold performance. PHA-based composites have demonstrated their biocompatibility with a range of tissues and their capacity to induce osteogenesis due to their piezoelectric properties. Electrospun PHB/PHBV fiber mesh in combination with human adipose tissue-derived stem cells (hASCs) were shown to improve vascularization in engineered bone tissues. For nerve and skin tissue engineering applications, natural polymers such as collagen and chitosan remain the gold standard but there is scope for development of scaffolds combining PHAs with other natural polymers which can address some of the limitations such as brittleness, lack of bioactivity and slow degradation rate presented by the latter. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1667-1684, 2017.
Collapse
Affiliation(s)
- Nowsheen Goonoo
- Centre for Biomedical and Biomaterials Research, University of Mauritius, MSIRI Building, Réduit, Mauritius
| | - Archana Bhaw-Luximon
- Centre for Biomedical and Biomaterials Research, University of Mauritius, MSIRI Building, Réduit, Mauritius
| | - Pearl Passanha
- Sustainable Environment Research Centre, Faculty of Computing, Engineering and Science, University of South Wales, Pontypridd, Wales, CF37 1DL, UK
| | - Sandra R Esteves
- Sustainable Environment Research Centre, Faculty of Computing, Engineering and Science, University of South Wales, Pontypridd, Wales, CF37 1DL, UK
| | - Dhanjay Jhurry
- Centre for Biomedical and Biomaterials Research, University of Mauritius, MSIRI Building, Réduit, Mauritius
| |
Collapse
|
48
|
Chen W, Thein-Han W, Weir MD, Chen Q, Xu HHK. Prevascularization of biofunctional calcium phosphate cement for dental and craniofacial repairs. Dent Mater 2016; 30:535-44. [PMID: 24731858 DOI: 10.1016/j.dental.2014.02.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 02/12/2014] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Calcium phosphate cement (CPC) is promising for dental and craniofacial repairs. Vascularization in bone tissue engineering constructs is currently a major challenge. The objectives of this study were to investigate the prevascularization of macroporous CPC via coculturing human umbilical vein endothelial cells (HUVEC) and human osteoblasts (HOB), and determine the effect of RGD in CPC on microcapillary formation for the first time. METHODS Macroporous CPC scaffold was prepared using CPC powder, chitosan liquid and gas-foaming porogen. Chitosan was grafted with Arg-Gly-Asp (RGD) to biofunctionalize the CPC. HUVEC and HOB were cocultured on macroporous CPC-RGD and CPC control without RGD for up to 42d. The osteogenic and angiogenic differentiation, bone matrix mineral synthesis, and formation of microcapillary-like structures were measured. RESULTS RGD-grafting in CPC increased the gene expressions of osteogenic and angiogenic differentiation markers than those of CPC control without RGD. Cell-synthesized bone mineral content also increased on CPC-RGD, compared to CPC control (p<0.05). Immunostaining with endothelial marker showed that the amount of microcapillary-like structures on CPC scaffolds increased with time. At 42d, the cumulative vessel length for CPC-RGD scaffold was 1.69-fold that of CPC control. SEM examination confirmed the morphology of self-assembled microcapillary-like structures on CPC scaffolds. SIGNIFICANCE HUVEC+HOB coculture on macroporous CPC scaffold successfully achieved prevascularization. RGD incorporation in CPC enhanced osteogenic differentiation, bone mineral synthesis, and microcapillary-like structure formation. The novel prevascularized CPC-RGD constructs are promising for dental, craniofacial and orthopedic applications.
Collapse
Affiliation(s)
- Wenchuan Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China; Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - WahWah Thein-Han
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Mechanical Engineering, University of Maryland, Baltimore County, MD 21250, USA.
| |
Collapse
|
49
|
Chidiac R, Zhang Y, Tessier S, Faubert D, Delisle C, Gratton JP. Comparative Phosphoproteomics Analysis of VEGF and Angiopoietin-1 Signaling Reveals ZO-1 as a Critical Regulator of Endothelial Cell Proliferation. Mol Cell Proteomics 2016; 15:1511-25. [PMID: 26846344 DOI: 10.1074/mcp.m115.053298] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Indexed: 12/18/2022] Open
Abstract
VEGF and angiopoietin-1 (Ang-1) are essential factors to promote angiogenesis through regulation of a plethora of signaling events in endothelial cells (ECs). Although pathways activated by VEGF and Ang-1 are being established, the unique signaling nodes conferring specific responses to each factor remain poorly defined. Thus, we conducted a large-scale comparative phosphoproteomic analysis of signaling pathways activated by VEGF and Ang-1 in ECs using mass spectrometry. Analysis of VEGF and Ang-1 networks of regulated phosphoproteins revealed that the junctional proteins ZO-1, ZO-2, JUP and p120-catenin are part of a cluster of proteins phosphorylated following VEGF stimulation that are linked to MAPK1 activation. Down-regulation of these junctional proteins led to MAPK1 activation and accordingly, increased proliferation of ECs stimulated specifically by VEGF, but not by Ang-1. We identified ZO-1 as the central regulator of this effect and showed that modulation of cellular ZO-1 levels is necessary for EC proliferation during vascular development of the mouse postnatal retina. In conclusion, we uncovered ZO-1 as part of a signaling node activated by VEGF, but not Ang-1, that specifically modulates EC proliferation during angiogenesis.
Collapse
Affiliation(s)
- Rony Chidiac
- From the ‡Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; §Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Ying Zhang
- From the ‡Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; §Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Sylvain Tessier
- ¶Proteomics discovery platform, Institut de recherches cliniques de Montréal (IRCM), Montreal, Quebec, Canada
| | - Denis Faubert
- ¶Proteomics discovery platform, Institut de recherches cliniques de Montréal (IRCM), Montreal, Quebec, Canada
| | - Chantal Delisle
- From the ‡Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Philippe Gratton
- From the ‡Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada;
| |
Collapse
|
50
|
Timmerman I, Daniel AE, Kroon J, van Buul JD. Leukocytes Crossing the Endothelium: A Matter of Communication. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:281-329. [PMID: 26940521 DOI: 10.1016/bs.ircmb.2015.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leukocytes cross the endothelial vessel wall in a process called transendothelial migration (TEM). The purpose of leukocyte TEM is to clear the causing agents of inflammation in underlying tissues, for example, bacteria and viruses. During TEM, endothelial cells initiate signals that attract and guide leukocytes to sites of tissue damage. Leukocytes react by attaching to these sites and signal their readiness to move back to endothelial cells. Endothelial cells in turn respond by facilitating the passage of leukocytes while retaining overall integrity. In this review, we present recent findings in the field and we have endeavored to synthesize a coherent picture of the intricate interplay between endothelial cells and leukocytes during TEM.
Collapse
Affiliation(s)
- Ilse Timmerman
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Anna E Daniel
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jaap D van Buul
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands.
| |
Collapse
|