101
|
Chang F, Flavahan S, Flavahan NA. Impaired activity of adherens junctions contributes to endothelial dilator dysfunction in ageing rat arteries. J Physiol 2017; 595:5143-5158. [PMID: 28561330 DOI: 10.1113/jp274189] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Ageing-induced endothelial dysfunction contributes to organ dysfunction and progression of cardiovascular disease. VE-cadherin clustering at adherens junctions promotes protective endothelial functions, including endothelium-dependent dilatation. Ageing increased internalization and degradation of VE-cadherin, resulting in impaired activity of adherens junctions. Inhibition of VE-cadherin clustering at adherens junctions (function-blocking antibody; FBA) reduced endothelial dilatation in young arteries but did not affect the already impaired dilatation in old arteries. After junctional disruption with the FBA, dilatation was similar in young and old arteries. Src tyrosine kinase activity and tyrosine phosphorylation of VE-cadherin were increased in old arteries. Src inhibition increased VE-cadherin at adherens junctions and increased endothelial dilatation in old, but not young, arteries. Src inhibition did not increase dilatation in old arteries treated with the VE-cadherin FBA. Ageing impairs the activity of adherens junctions, which contributes to endothelial dilator dysfunction. Restoring the activity of adherens junctions could be of therapeutic benefit in vascular ageing. ABSTRACT Endothelial dilator dysfunction contributes to pathological vascular ageing. Experiments assessed whether altered activity of endothelial adherens junctions (AJs) might contribute to this dysfunction. Aortas and tail arteries were isolated from young (3-4 months) and old (22-24 months) F344 rats. VE-cadherin immunofluorescent staining at endothelial AJs and AJ width were reduced in old compared to young arteries. A 140 kDa VE-cadherin species was present on the cell surface and in TTX-insoluble fractions, consistent with junctional localization. Levels of the 140 kDa VE-cadherin were decreased, whereas levels of a TTX-soluble 115 kDa VE-cadherin species were increased in old compared to young arteries. Acetylcholine caused endothelium-dependent dilatation that was decreased in old compared to young arteries. Disruption of VE-cadherin clustering at AJs (function-blocking antibody, FBA) inhibited dilatation to acetylcholine in young, but not old, arteries. After the FBA, there was no longer any difference in dilatation between old and young arteries. Src activity and tyrosine phosphorylation of VE-cadherin were increased in old compared to young arteries. In old arteries, Src inhibition (saracatinib) increased: (i) 140 kDa VE-cadherin in the TTX-insoluble fraction, (ii) VE-cadherin intensity at AJs, (iii) AJ width, and (iv) acetylcholine dilatation. In old arteries treated with the FBA, saracatinib no longer increased acetylcholine dilatation. Saracatinib did not affect dilatation in young arteries. Therefore, ageing impairs AJ activity, which appears to reflect Src-induced phosphorylation, internalization and degradation of VE-cadherin. Moreover, impaired AJ activity can account for the endothelial dilator dysfunction in old arteries. Restoring endothelial AJ activity may be a novel therapeutic approach to vascular ageing.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sheila Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas A Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
102
|
Yu P, Wilhelm K, Dubrac A, Tung JK, Alves TC, Fang JS, Xie Y, Zhu J, Chen Z, De Smet F, Zhang J, Jin SW, Sun L, Sun H, Kibbey RG, Hirschi KK, Hay N, Carmeliet P, Chittenden TW, Eichmann A, Potente M, Simons M. FGF-dependent metabolic control of vascular development. Nature 2017; 545:224-228. [PMID: 28467822 PMCID: PMC5427179 DOI: 10.1038/nature22322] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/29/2017] [Indexed: 12/22/2022]
Abstract
Blood and lymphatic vasculatures are intimately involved in tissue oxygenation and fluid homeostasis maintenance. Assembly of these vascular networks involves sprouting, migration and proliferation of endothelial cells. Recent studies have suggested that changes in cellular metabolism are important to these processes. Although much is known about vascular endothelial growth factor (VEGF)-dependent regulation of vascular development and metabolism, little is understood about the role of fibroblast growth factors (FGFs) in this context. Here we identify FGF receptor (FGFR) signalling as a critical regulator of vascular development. This is achieved by FGF-dependent control of c-MYC (MYC) expression that, in turn, regulates expression of the glycolytic enzyme hexokinase 2 (HK2). A decrease in HK2 levels in the absence of FGF signalling inputs results in decreased glycolysis, leading to impaired endothelial cell proliferation and migration. Pan-endothelial- and lymphatic-specific Hk2 knockouts phenocopy blood and/or lymphatic vascular defects seen in Fgfr1/Fgfr3 double mutant mice, while HK2 overexpression partly rescues the defects caused by suppression of FGF signalling. Thus, FGF-dependent regulation of endothelial glycolysis is a pivotal process in developmental and adult vascular growth and development.
Collapse
MESH Headings
- Animals
- Cell Movement
- Cell Proliferation
- Endothelial Cells/cytology
- Endothelial Cells/metabolism
- Female
- Fibroblast Growth Factors/metabolism
- Glycolysis
- Hexokinase/metabolism
- Lymphangiogenesis
- Lymphatic Vessels/cytology
- Lymphatic Vessels/metabolism
- Mice
- Mice, Inbred C57BL
- Neovascularization, Physiologic
- Proto-Oncogene Proteins c-myc/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/deficiency
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/deficiency
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Pengchun Yu
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
| | - Kerstin Wilhelm
- Angiogenesis & Metabolism Laboratory, Max Plank Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Alexandre Dubrac
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
| | - Joe K. Tung
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
| | - Tiago C. Alves
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine
| | - Jennifer S. Fang
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
| | - Yi Xie
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
| | - Jie Zhu
- Department of Cellular and Molecular Physiology, Yale University School of Medicine
| | - Zehua Chen
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, MA, USA
| | - Frederik De Smet
- Switch Laboratory, VIB-KU Leuven, Leuven, B-3000, Belgium
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jiasheng Zhang
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
| | - Suk-Won Jin
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Lele Sun
- Genomics Laboratory, WuXi NextCODE, Shanghai, China
| | - Hongye Sun
- Genomics Laboratory, WuXi NextCODE, Shanghai, China
| | - Richard G. Kibbey
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine
| | - Karen K. Hirschi
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, University of Leuven, Leuven, B-3000, Belgium
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, B-3000, Belgium
| | - Thomas W. Chittenden
- Computational Statistics and Bioinformatics Group, Advanced Artificial Intelligence Research Laboratory, WuXi NextCODE, Cambridge, MA, USA
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
- U970, Paris Cardiovascular Research Center, 56 Rue Leblanc, 75015 Paris, France
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Max Plank Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Michael Simons
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
103
|
Helbing T, Wiltgen G, Hornstein A, Brauers EZ, Arnold L, Bauer A, Esser JS, Diehl P, Grundmann S, Fink K, Patterson C, Bode C, Moser M. Bone Morphogenetic Protein-Modulator BMPER Regulates Endothelial Barrier Function. Inflammation 2017; 40:442-453. [PMID: 27995357 DOI: 10.1007/s10753-016-0490-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The endothelium serves as a selective barrier and controls the exchange of nutrients, hormones, and leukocytes between blood and tissues. Molecular mechanisms contributing to the pathogenesis of endothelial barrier dysfunction remain incompletely understood. Accumulating evidence implicates bone morphogenetic protein (BMP)-modulator BMPER as a key regulator in endothelial biology. Herein, we analyze the impact of BMPER in the control of endothelial barrier function. To assess the role of BMPER in vascular barrier function in mice, we measured the leakage of Evans blue dye from blood into interstitial lung tissue. BMPER+/- mice exhibited a significantly higher degree of vascular leak compared with wild-type siblings. In accordance with our in vivo observation, siRNA-based BMPER knockdown in human umbilical endothelial cells increased endothelial permeability measured by FITC-dextran passage in transwell assays. Mechanistically, BMPER knockdown reduced the expression of VE-cadherin, a pivotal component of endothelial adherens junctions. Conversely, recombinant human BMPER protein upregulated VE-cadherin protein levels and improved endothelial barrier function in transwell assays. The effects of BMPER knockdown on VE-cadherin expression and endothelial permeability were induced by enhanced BMP activity. Supporting this notion, activation of BMP4-Smad-Id1 signaling reduced VE-cadherin levels and impaired endothelial barrier function in vitro. In vivo, Evans blue dye accumulation was higher in the lungs of BMP4-treated C57BL/6 mice compared to controls indicating that BMP4 increased vascular permeability. High levels of BMPER antagonized BMP4-Smad5-Id1 signaling and prevented BMP4-induced downregulation of VE-cadherin and endothelial leakage, suggesting that BMPER exerts anti-BMP effects and restores endothelial barrier function. Taken together, this data demonstrates that BMPER-modulated BMP pathway activity regulates VE-cadherin expression and vascular barrier function.
Collapse
Affiliation(s)
- Thomas Helbing
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.
| | - Gwendoline Wiltgen
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Alexandra Hornstein
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Elena Z Brauers
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Linus Arnold
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Adrian Bauer
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Jennifer S Esser
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Philipp Diehl
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Sebastian Grundmann
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Katrin Fink
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Department of Emergency Medicine, University Hospital of Freiburg, Freiburg im Breisgau, Germany
| | - Cam Patterson
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- New York-Presbyterian Hospital, New York City, NY, 10065, USA
| | - Christoph Bode
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Martin Moser
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
104
|
Khuu LA, Tayyari F, Sivak JM, Flanagan JG, Singer S, Brent MH, Huang D, Tan O, Hudson C. Aqueous humour concentrations of TGF-β, PLGF and FGF-1 and total retinal blood flow in patients with early non-proliferative diabetic retinopathy. Acta Ophthalmol 2017; 95:e206-e211. [PMID: 27678201 DOI: 10.1111/aos.13230] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/11/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE To correlate angiogenic cytokines in the aqueous humour with total retinal blood flow in subjects with type 2 diabetes with non-proliferative diabetic retinopathy (NPDR). METHODS A total of 17 controls and 16 NPDR patients were recruited into the study. Aqueous humour was collected at the start of cataract surgery to assess the concentration of 14 angiogenic cytokines. Aqueous humour was analysed using the suspension array method. Six images were acquired to assess total retinal blood flow (TRBF) using the prototype RTVue™ Doppler Fourier domain optical coherence tomography (Doppler FD-OCT) (Optovue, Inc., Fremont, CA) using a double circular scan protocol, 1 month postsurgery. At the same visit, forearm blood was collected to determine glycosylated haemoglobin (A1c). RESULTS Transforming growth factor beta (TGF-β1, TGF-β2) and PLGF were increased while FGF-1 was reduced in NPDR compared to controls (Bonferroni corrected, p < 0.003 for all). Total retinal blood flow (TRBF) was significantly reduced in the NPDR group compared to controls (33.1 ± 9.9 versus 43.3 ± 5.3 μl/min, p = 0.002). Aqueous FGF-1 significantly correlated with TRBF in the NPDR group (r = 0.71, p = 0.01; r2 = 0.51). In a multiple regression analysis, A1c was found to be a significant predictor of aqueous TGF-β1 and FGF-1 (p = 0.018 and p = 0.020, respectively). CONCLUSION Aqueous angiogenic cytokines (TGF-β1, TGF-β2 and PLGF) were elevated in conjunction with a reduction in TRBF in patients with NPDR compared to controls. Non-invasive measurement of TRBF may be useful for predicting aqueous FGF-1 levels and severity of vasculopathy in DR.
Collapse
Affiliation(s)
- Lee-Anne Khuu
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Vision Science and Ophthalmology; Toronto Western Hospital; Toronto ON Canada
| | - Faryan Tayyari
- School of Optometry and Vision Science; University of Waterloo; Waterloo ON Canada
| | - Jeremy M. Sivak
- Vision Science and Ophthalmology; Toronto Western Hospital; Toronto ON Canada
| | - John G. Flanagan
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Vision Science and Ophthalmology; Toronto Western Hospital; Toronto ON Canada
| | - Shaun Singer
- Vision Science and Ophthalmology; Toronto Western Hospital; Toronto ON Canada
| | - Michael H. Brent
- Vision Science and Ophthalmology; Toronto Western Hospital; Toronto ON Canada
| | - David Huang
- Casey Eye Institute; Oregon Health and Science University; Portland OR USA
| | - Ou Tan
- Casey Eye Institute; Oregon Health and Science University; Portland OR USA
| | - Christopher Hudson
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Vision Science and Ophthalmology; Toronto Western Hospital; Toronto ON Canada
- School of Optometry and Vision Science; University of Waterloo; Waterloo ON Canada
| |
Collapse
|
105
|
Aragon-Sanabria V, Pohler SE, Eswar VJ, Bierowski M, Gomez EW, Dong C. VE-Cadherin Disassembly and Cell Contractility in the Endothelium are Necessary for Barrier Disruption Induced by Tumor Cells. Sci Rep 2017; 7:45835. [PMID: 28393886 PMCID: PMC5385522 DOI: 10.1038/srep45835] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/03/2017] [Indexed: 12/20/2022] Open
Abstract
During metastasis, breakdown of the endothelial barrier is critical for tumor cell extravasation through blood vessel walls and is mediated by a combination of tumor secreted soluble factors and receptor-ligand interactions. However, a complete mechanism governing tumor cell transendothelial migration remains unclear. Here, we investigate the roles of tumor-associated signals in regulating endothelial cell contractility and adherens junction disassembly leading to endothelial barrier breakdown. We show that Src mediates VE-cadherin disassembly in response to metastatic melanoma cells. Through the use of pharmacological inhibitors of cytoskeletal contractility we find that endothelial cell contractility is responsive to interactions with metastatic cancer cells and that reducing endothelial cell contractility abrogates migration of melanoma cells across endothelial monolayers. Furthermore, we find that a combination of tumor secreted soluble factors and receptor-ligand interactions mediate activation of Src within endothelial cells that is necessary for phosphorylation of VE-cadherin and for breakdown of the endothelial barrier. Together, these results provide insight into how tumor cell signals act in concert to modulate cytoskeletal contractility and adherens junctions disassembly during extravasation and may aid in identification of therapeutic targets to block metastasis.
Collapse
Affiliation(s)
- Virginia Aragon-Sanabria
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| | - Steven E. Pohler
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Vikram J. Eswar
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| | - Matthew Bierowski
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Esther W. Gomez
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Pennsylvania State university, University Park, PA, 16802, USA
| |
Collapse
|
106
|
Maeda K, Alarcon EI, Suuronen EJ, Ruel M. Optimizing the host substrate environment for cardiac angiogenesis, arteriogenesis, and myogenesis. Expert Opin Biol Ther 2017; 17:435-447. [PMID: 28274146 DOI: 10.1080/14712598.2017.1293038] [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] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The diseased host milieu, such as endothelial dysfunction (ED), decreased NO bioavailability, and ischemic/inflammatory post-MI environment, hamper the clinical success of existing cardiac regenerative therapies. Area covered: In this article, current strategies including pharmacological and nonpharmacological approaches for improving the diseased host milieu are reviewed. Specifically, the authors provide focus on: i) the mechanism of ED in patients with cardiovascular diseases, ii) the current results of ED improving strategies in pre-clinical and clinical studies, and iii) the use of biomaterials as a novel modulator in damaged post-MI environment. Expert opinion: Adjunct therapies which improve host endothelial function have demonstrated promising outcomes, potentially overcoming disappointing results of cell therapy in human studies. In the future, elucidation of the interactions between the host tissue and therapeutic agents, as well as downstream signaling pathways, will be the next challenges in enhancing regenerative therapy. More careful investigations are also required to establish these agents' safety and efficacy for wide usage in humans.
Collapse
Affiliation(s)
- Kay Maeda
- a Divisions of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , ON , Canada
| | - Emilio I Alarcon
- a Divisions of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , ON , Canada
| | - Erik J Suuronen
- a Divisions of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , ON , Canada
| | - Marc Ruel
- a Divisions of Cardiac Surgery , University of Ottawa Heart Institute , Ottawa , ON , Canada
| |
Collapse
|
107
|
Dejana E, Hirschi KK, Simons M. The molecular basis of endothelial cell plasticity. Nat Commun 2017; 8:14361. [PMID: 28181491 PMCID: PMC5309780 DOI: 10.1038/ncomms14361] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 12/14/2016] [Indexed: 02/06/2023] Open
Abstract
The endothelium is capable of remarkable plasticity. In the embryo, primitive endothelial cells differentiate to acquire arterial, venous or lymphatic fates. Certain endothelial cells also undergo hematopoietic transition giving rise to multi-lineage hematopoietic stem and progenitors while others acquire mesenchymal properties necessary for heart development. In the adult, maintenance of differentiated endothelial state is an active process requiring constant signalling input. The failure to do so leads to the development of endothelial-to-mesenchymal transition that plays an important role in pathogenesis of a number of diseases. A better understanding of these phenotypic changes may lead to development of new therapeutic interventions. Vascular endothelium possesses remarkable plasticity in response to cues from its surroundings, leading to great heterogeneity of endothelial cells in different vascular beds. Here the authors explain the molecular basis of endothelial plasticity during embryogenesis and in various diseases.
Collapse
Affiliation(s)
- Elisabetta Dejana
- Vascular Biology Unit, FIRC Institute of Molecular Oncology, Milan 20129, Italy
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 751 85, Sweden
| | - Karen K. Hirschi
- Yale Cardiovasc. Res. Center, Departments of Internal Medicine, Genetics and Biomedical Engineering New Haven, Connecticut CT06511, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine and Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut CT06511, USA
| |
Collapse
|
108
|
Hänggi K, Vasilikos L, Valls AF, Yerbes R, Knop J, Spilgies LM, Rieck K, Misra T, Bertin J, Gough PJ, Schmidt T, de Almodòvar CR, Wong WWL. RIPK1/RIPK3 promotes vascular permeability to allow tumor cell extravasation independent of its necroptotic function. Cell Death Dis 2017; 8:e2588. [PMID: 28151480 PMCID: PMC5386469 DOI: 10.1038/cddis.2017.20] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/22/2016] [Accepted: 01/03/2017] [Indexed: 12/13/2022]
Abstract
Necroptosis is an inflammatory form of programmed cell death requiring receptor-interacting protein kinase 1, 3 (RIPK1, RIPK3) and mixed lineage kinase domain-like protein (MLKL). The kinase of RIPK3 phosphorylates MLKL causing MLKL to form a pore-like structure, allowing intracellular contents to release and cell death to occur. Alternatively, RIPK1 and RIPK3 have been shown to regulate cytokine production directly influencing inflammatory immune infiltrates. Recent data suggest that necroptosis may contribute to the malignant transformation of tumor cells in vivo and we asked whether necroptosis may have a role in the tumor microenvironment altering the ability of the tumor to grow or metastasize. To determine if necroptosis in the tumor microenvironment could promote inflammation alone or by initiating necroptosis and thereby influencing growth or metastasis of tumors, we utilized a syngeneic tumor model of metastasis. Loss of RIPK3 in the tumor microenvironment reduced the number of tumor nodules in the lung by 46%. Loss of the kinase activity in RIPK1, a member of the necrosome also reduced tumor nodules in the lung by 38%. However, the loss of kinase activity in RIPK3 or the loss of MLKL only marginally altered the ability of tumor cells to form in the lung. Using bone marrow chimeras, the decrease in tumor nodules in the Ripk3-/- appeared to be due to the stromal compartment rather than the hematopoietic compartment. Transmigration assays showed decreased ability of tumor cells to transmigrate through the vascular endothelial layer, which correlated with decreased permeability in the Ripk3-/- mice after tumor injection. In response to permeability factors, such as vascular endothelial growth factor, RIPK3 null endothelial cells showed decreased p38/HSP27 activation. Taken together, our results suggest an alternative function for RIPK1/RIPK3 in vascular permeability leading to decreased number of metastasis.
Collapse
Affiliation(s)
- Kay Hänggi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Lazaros Vasilikos
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Aida Freire Valls
- Biochemistry Center, Heidelberg University, Heidelberg, Germany.,Department of General, Visceral and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Rosario Yerbes
- Biochemistry Center, Heidelberg University, Heidelberg, Germany
| | - Janin Knop
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Lisanne M Spilgies
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Kristy Rieck
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Tvisha Misra
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA, USA
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA, USA
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | | | - W Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
109
|
|
110
|
Dephosphorylation of Y685-VE-Cadherin Involved in Pulmonary Microvascular Endothelial Barrier Injury Induced by Angiotensin II. Mediators Inflamm 2016; 2016:8696481. [PMID: 28119542 PMCID: PMC5227173 DOI: 10.1155/2016/8696481] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/04/2016] [Accepted: 11/22/2016] [Indexed: 11/23/2022] Open
Abstract
Angiotensin II (AngII) caused pulmonary microvascular endothelial barrier injury, which induced acute aortic dissection (AAD) combined with acute lung injury (ALI). However, the exact mechanism is unclear. We investigated the role of dephosphorylation of Y685-VE-cadherin in the AngII induced pulmonary microvascular endothelial barrier injury. Mice or pulmonary microvascular endothelial cells (PMVECs) were divided into control group, AngII group, AngII+PP2 (Src kinase inhibitor) group, and PP2 group. PP2 was used to inhibit the phosphorylation of Y685-VE-cadherin. Pathological changes, infiltration of macrophages and neutrophils, and pulmonary microvascular permeability were used to determine the pulmonary microvascular endothelial barrier function. Flow cytometry was used to determine the apoptosis of PMVECs, and immunofluorescence was used to determine the skeletal arrangement. Transendothelial resistance was used to detect the permeability of endothelial barrier. Phosphorylation of Y685-VE-cadherin was significantly reduced after AngII stimulation (P < 0.05), together with skeletal rearrangement, and elevation of endothelial permeability which finally induced endothelial barrier injury. After PP2 interference, the phosphorylation of Y685-VE-cadherin was further reduced and the endothelial permeability was further elevated. These data indicated that AngII could induce pulmonary injury by triggering endothelial barrier injury, and such process may be related to the dephosphorylation of Y685-VE-cadherin and the endothelial skeletal rearrangement.
Collapse
|
111
|
Ghebre YT, Yakubov E, Wong WT, Krishnamurthy P, Sayed N, Sikora AG, Bonnen MD. Vascular Aging: Implications for Cardiovascular Disease and Therapy. TRANSLATIONAL MEDICINE (SUNNYVALE, CALIF.) 2016; 6:183. [PMID: 28932625 PMCID: PMC5602592 DOI: 10.4172/2161-1025.1000183] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The incidence and prevalence of cardiovascular disease is highest among the elderly, in part, due to deleterious effects of advancing age on the heart and blood vessels. Aging, a known cardiovascular risk factor, is progressively associated with structural and functional changes to the vasculature including hemodynamic disturbance due to increased oxidative stress, premature cellular senescence and impairments in synthesis and/or secretion of endothelium-derived vasoactive molecules. These molecular and physiological changes lead to vessel wall stiffening and thickening, as well as other vascular complications that culminate to loss of vascular tone regulation and endothelial function. Intriguingly, the vessel wall, a biochemically active structure composed of collagen, connective tissue, smooth muscle and endothelial cells, is adversely affected by processes involved in premature or normal aging. Notably, the inner most layer of the vessel wall, the endothelium, becomes senescent and dysfunctional with advancing age. As a result, its ability to release vasoactive molecules such as acetylcholine (ACh), prostacyclin (PGI2), endothelium-derived hyperpolarizing factor (EDHF), and nitric oxide (NO) is reduced and the cellular response to these molecules is also impaired. By contrast, the vascular endothelium increases its generation and release of reactive oxygen (ROS) and nitrogen (RNS) species, vasoconstrictors such as endothelin (ET) and angiotensin (AT), and endogenous inhibitors of NO synthases (NOSs) to block NO. This skews the balance of the endothelium in favor of the release of highly tissue reactive and harmful molecules that promote DNA damage, telomere erosion, senescence, as well as stiffened and hardened vessel wall that is prone to the development of hypertension, diabetes, atherosclerosis and other cardiovascular risk factors. This Review discusses the impact of advancing age on cardiovascular health, and highlights the cellular and molecular mechanisms that underlie age-associated vascular changes. In addition, the role of pharmacological interventions in preventing or delaying age-related cardiovascular disease is discussed.
Collapse
Affiliation(s)
- Yohannes T Ghebre
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Eduard Yakubov
- phaRNA Comprehensive RNA Technologies, Houston, Texas, USA
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nazish Sayed
- Department of Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Andrew G Sikora
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Mark D Bonnen
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
112
|
Cadwell CM, Su W, Kowalczyk AP. Cadherin tales: Regulation of cadherin function by endocytic membrane trafficking. Traffic 2016; 17:1262-1271. [PMID: 27624909 DOI: 10.1111/tra.12448] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 12/17/2022]
Abstract
Cadherins are the primary adhesion molecules in adherens junctions and desmosomes and play essential roles in embryonic development. Although significant progress has been made in understanding cadherin structure and function, we lack a clear vision of how cells confer plasticity upon adhesive junctions to allow for cellular rearrangements during development, wound healing and metastasis. Endocytic membrane trafficking has emerged as a fundamental mechanism by which cells confer a dynamic state to adhesive junctions. Recent studies indicate that the juxtamembrane domain of classical cadherins contains multiple endocytic motifs, or "switches," that can be used by cellular membrane trafficking machinery to regulate adhesion. The cadherin-binding protein p120-catenin (p120) appears to be the master regulator of access to these switches, thereby controlling cadherin endocytosis and turnover. This review focuses on p120 and other cadherin-binding proteins, ubiquitin ligases, and growth factors as key modulators of cadherin membrane trafficking.
Collapse
Affiliation(s)
- Chantel M Cadwell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Wenji Su
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Biochemistry, Cell, and Developmental Biology Graduate Training Program, Emory University, Atlanta, Georgia
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
113
|
Wu Z, Liu H, Ren W, Dai F, Chang J, Li B. VE-cadherin involved in the pulmonary microvascular endothelial cell barrier injury induced by angiotensin II through modulating the cellular apoptosis and skeletal rearrangement. Am J Transl Res 2016; 8:4310-4319. [PMID: 27830014 PMCID: PMC5095323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/25/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE Angiotensin II (AngII) involved in the pathogenesis of pulmonary injury through impairing the integrity of pulmonary microvascular endothelial barrier, but the mechanism is still not clear. We aim to determine the roles of VE-cadherin, playing crucial roles in the adhesion of the vascular endothelial barrier and the barrier function, in the pulmonary microvascular endothelial cell (PMVEC) barrier injury mediated by AngII. METHODS Mice acute lung injury (ALI) model was induced through pumping of AngII. The infiltration of macrophages and neutrophils as well as the PMVEC permeability were determined in order to determine the barrier injury in vivo and in vitro. Knockdown of VE-cadherin was established using siRNA technique, and its roles in the apoptosis and skeletal rearrangement in the PMVECs were evaluated. RESULTS After AngII interference, the expression of VE-cadherin in the PMVECs and pulmonary tissues in mice was down-regulated. Upon VE-cadherin knockdown through siRNA technique, AngII induced susceptibility of PMVECs to apoptosis. Knockdown of VE-cadherin contributed to the skeletal rearrangement in the endothelial cells, together with increase of permeability. CONCLUSIONS VE-cadherin expression is closely related to the apoptosis and skeletal rearrangement of PMVECs induced by AngII.
Collapse
Affiliation(s)
- Zhiyong Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430060, China
| | - Huagang Liu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430060, China
| | - Wei Ren
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430060, China
| | - Feifeng Dai
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430060, China
| | - Jinxing Chang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430060, China
| | - Bowen Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University Wuhan 430060, China
| |
Collapse
|
114
|
Gao F, Artham S, Sabbineni H, Al-Azayzih A, Peng XD, Hay N, Adams RH, Byzova TV, Somanath PR. Akt1 promotes stimuli-induced endothelial-barrier protection through FoxO-mediated tight-junction protein turnover. Cell Mol Life Sci 2016; 73:3917-33. [PMID: 27113546 PMCID: PMC5023469 DOI: 10.1007/s00018-016-2232-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 11/28/2022]
Abstract
Vascular permeability regulated by the vascular endothelial growth factor (VEGF) through endothelial-barrier junctions is essential for inflammation. Mechanisms regulating vascular permeability remain elusive. Although 'Akt' and 'Src' have been implicated in the endothelial-barrier regulation, it is puzzling how both agents that protect and disrupt the endothelial-barrier activate these kinases to reciprocally regulate vascular permeability. To delineate the role of Akt1 in endothelial-barrier regulation, we created endothelial-specific, tamoxifen-inducible Akt1 knockout mice and stable ShRNA-mediated Akt1 knockdown in human microvascular endothelial cells. Akt1 loss leads to decreased basal and angiopoietin1-induced endothelial-barrier resistance, and enhanced VEGF-induced endothelial-barrier breakdown. Endothelial Akt1 deficiency resulted in enhanced VEGF-induced vascular leakage in mice ears, which was rescued upon re-expression with Adeno-myrAkt1. Furthermore, co-treatment with angiopoietin1 reversed VEGF-induced vascular leakage in an Akt1-dependent manner. Mechanistically, our study revealed that while VEGF-induced short-term vascular permeability is independent of Akt1, its recovery is reliant on Akt1 and FoxO-mediated claudin expression. Pharmacological inhibition of FoxO transcription factors rescued the defective endothelial barrier due to Akt1 deficiency. Here we provide novel insights on the endothelial-barrier protective role of VEGF in the long term and the importance of Akt1-FoxO signaling on tight-junction stabilization and prevention of vascular leakage through claudin expression.
Collapse
Affiliation(s)
- Fei Gao
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sandeep Artham
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Harika Sabbineni
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Ahmad Al-Azayzih
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
- College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Xiao-Ding Peng
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ralf H Adams
- Max Plank Institute of Molecular Biomedicine, Röntgenstraße 20, Münster, Germany
| | - Tatiana V Byzova
- Department of Molecular Cardiology, Joseph J. Jacob's Center for Thrombosis and Vascular Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA.
- Department of Medicine and Vascular Biology Center, Augusta University, HM1200, Augusta, GA, 30912, USA.
| |
Collapse
|
115
|
|
116
|
Ramasamy SK, Kusumbe AP, Itkin T, Gur-Cohen S, Lapidot T, Adams RH. Regulation of Hematopoiesis and Osteogenesis by Blood Vessel-Derived Signals. Annu Rev Cell Dev Biol 2016; 32:649-675. [PMID: 27576121 DOI: 10.1146/annurev-cellbio-111315-124936] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In addition to their conventional role as a versatile transport system, blood vessels provide signals controlling organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular osteoprogenitor cells and thereby control bone formation. Blood vessels are also a critical component of niche microenvironments for hematopoietic stem cells. Here we discuss key pathways and factors controlling endothelial cell behavior in bone, the role of vessels in osteogenesis, and the nature of vascular stem cell niches in bone marrow.
Collapse
Affiliation(s)
- Saravana K Ramasamy
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Anjali P Kusumbe
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Tomer Itkin
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Shiri Gur-Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Tsvee Lapidot
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| |
Collapse
|
117
|
Sawicka M, Janowska J, Chudek J. Potential beneficial effect of some adipokines positively correlated with the adipose tissue content on the cardiovascular system. Int J Cardiol 2016; 222:581-589. [PMID: 27513655 DOI: 10.1016/j.ijcard.2016.07.054] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/12/2016] [Accepted: 07/04/2016] [Indexed: 01/30/2023]
Abstract
Obesity is a risk factor of cardiovascular diseases. However, in the case of heart failure, obese and overweight patients have a more favourable prognosis compared to patients who have a normal body weight. This phenomenon is referred to as the "obesity paradox," and it is explained by, among others, a positive effect of adipokines produced by adipose tissue, particularly by the tissue located in the direct vicinity of the heart and blood vessels. The favourable effect on the cardiovascular system is mostly associated with adiponectin and omentin, but the levels of these substances are reduced in obese patients. Among the adipokines which levels are positively correlated with the adipose tissue content, favourable activity is demonstrated by apelin, progranulin, chemerin, TNF-α (tumour necrosis factor-)α, CTRP-3 (C1q/tumour necrosis factor (TNF) related protein), leptin, visfatin and vaspin. This activity is associated with the promotion of regeneration processes in the damaged myocardium, formation of new blood vessels, reduction of the afterload, improvement of metabolic processes in cardiomyocytes and myocardial contractile function, inhibition of apoptosis and fibrosis of the myocardium, as well as anti-inflammatory and anti-atheromatous effects. The potential use of these properties in the treatment of heart failure and ischaemic heart disease, as well as in pulmonary hypertension, arterial hypertension and the limitation of the loss of cardiomyocytes during cardioplegia-requiring cardiosurgical procedures, is studied. The most advanced studies focus on analogues of apelin and progranulin.
Collapse
Affiliation(s)
- Magdalena Sawicka
- Department of Cardiology, Congenital Heart Diseases and Electrotherapy, Silesian Center for Heart Diseases, 9 Maria Skłodowska- Curie Street, 41-800 Zabrze, Poland; Department of Pathophysiology, Faculty of Medicine, Medical University of Silesia, 18 Medyków Street, 40-027 Katowice, Poland.
| | - Joanna Janowska
- Department of Pathophysiology, Faculty of Medicine, Medical University of Silesia, 18 Medyków Street, 40-027 Katowice, Poland
| | - Jerzy Chudek
- Department of Pathophysiology, Faculty of Medicine, Medical University of Silesia, 18 Medyków Street, 40-027 Katowice, Poland
| |
Collapse
|
118
|
Intrinsic FGF2 and FGF5 promotes angiogenesis of human aortic endothelial cells in 3D microfluidic angiogenesis system. Sci Rep 2016; 6:28832. [PMID: 27357248 PMCID: PMC4928073 DOI: 10.1038/srep28832] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/06/2016] [Indexed: 12/29/2022] Open
Abstract
The human body contains different endothelial cell types and differences in their angiogenic potential are poorly understood. We compared the functional angiogenic ability of human aortic endothelial cells (HAECs) and human umbilical vein endothelial cells (HUVECs) using a three-dimensional (3D) microfluidic cell culture system. HAECs and HUVECs exhibited similar cellular characteristics in a 2D culture system; however, in the 3D microfluidic angiogenesis system, HAECs exhibited stronger angiogenic potential than HUVECs. Interestingly, the expression level of fibroblast growth factor (FGF)2 and FGF5 under vascular endothelial growth factor (VEGF)-A stimulation was significantly higher in HAECs than in HUVECs. Moreover, small interfering RNA-mediated knockdown of FGF2 and FGF5 more significantly attenuated vascular sprouting induced from HAECs than HUVECs. Our results suggest that HAECs have greater angiogenic potential through FGF2 and FGF5 upregulation and could be a compatible endothelial cell type to achieve robust angiogenesis.
Collapse
|
119
|
Chang F, Flavahan S, Flavahan NA. Immature endothelial cells initiate endothelin-mediated constriction of newborn arteries. J Physiol 2016; 594:4933-44. [PMID: 27062279 DOI: 10.1113/jp272176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/29/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Endothelial expression and the release of endothelin-1 (ET-1) in levels sufficient to initiate vasoconstriction is considered to be a hallmark feature of pathological endothelial dysfunction. During the immediate postnatal period, arterial endothelial cells undergo remarkable structural and functional changes as they transition to a mature protective cell layer, which includes a marked increase in NO dilator activity. The present study demonstrates that endothelial cells lining newborn central arteries express high levels of ET-1 peptides and, in response to endothelial stimulation, rapidly release ET-1 and initiate powerful ET-1-mediated constriction. This activity is lost as the endothelium matures in the postnatal period. Heightened activity of ET-1 in the neonatal endothelium might contribute to inappropriate responses of immature arteries to stress or injury. Indeed, the immature endothelium resembles dysfunctional endothelial cells, and retention or re-emergence of this phenotype may contribute to the development of vascular disease. ABSTRACT Endothelial cells lining fetal and newborn arteries have an unusual phenotype, including reduced NO activity, prominent actin stress fibres and poorly developed cellular junctions. Experiments were performed to determine whether the immature endothelium of newborn arteries also expresses and releases endothelin-1 (ET-1) and initiates endothelium-dependent constriction. Carotid arteries were isolated from newborn (postnatal day 1; P1), postnatal day 7 (P7) and postnatal day 21 (P21) mice and assessed in a pressure myograph system. Endothelial stimulation with A23187 or thrombin caused constriction in P1 arteries, no significant change in diameter of P7 arteries, and dilatation in P21 arteries. In P1 arteries, constriction to thrombin or A23187 was inhibited by endothelial-denudation, by ET-1 receptor antagonists (BQ123 plus BQ788) or by inhibition of endothelin-converting enzyme (phosphoramidon or SM19712). ET-1 receptor antagonism did not affect responses to thrombin or A23187 in more mature arteries. Exogenous ET-1 caused similar concentration-dependent constrictions of P1, P7 and P21 arteries. Endothelial stimulation with thrombin rapidly increased the endothelial release of ET-1 from P1 but not P21 aortas. Endothelial expression of ET-1 peptides, as assessed by immunofluorescence analysis, was increased in P1 compared to P21 arteries. Therefore, newborn endothelial cells express high levels of ET-1 peptides, rapidly release ET-1 in response to endothelial stimulation, and initiate ET-1-mediated endothelium-dependent constriction. This activity is diminished as the endothelium matures in the immediate postnatal period. Heightened activity of ET-1 in neonatal endothelium probably reflects an early developmental role of the peptide, although this might contribute to inappropriate responses of immature arteries to stress or injury.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sheila Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas A Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
120
|
Nguyen T, Mège RM. N-Cadherin and Fibroblast Growth Factor Receptors crosstalk in the control of developmental and cancer cell migrations. Eur J Cell Biol 2016; 95:415-426. [PMID: 27320194 DOI: 10.1016/j.ejcb.2016.05.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/13/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022] Open
Abstract
Cell migrations are diverse. They constitutemajor morphogenetic driving forces during embryogenesis, but they contribute also to the loss of tissue homeostasis and cancer growth. Capabilities of cells to migrate as single cells or as collectives are controlled by internal and external signalling, leading to the reorganisation of their cytoskeleton as well as by the rebalancing of cell-matrix and cell-cell adhesions. Among the genes altered in numerous cancers, cadherins and growth factor receptors are of particular interest for cell migration regulation. In particular, cadherins such as N-cadherin and a class of growth factor receptors, namely FGFRs cooperate to regulate embryonic and cancer cell behaviours. In this review, we discuss on reciprocal crosstalk between N-cadherin and FGFRs during cell migration. Finally, we aim at clarifying the synergy between N-cadherin and FGFR signalling that ensure cellular reorganization during cell movements, mainly during cancer cell migration and metastasis but also during developmental processes.
Collapse
Affiliation(s)
- Thao Nguyen
- Institut Jacques Monod, CNRS, Université Paris Diderot, Paris, France
| | - René Marc Mège
- Institut Jacques Monod, CNRS, Université Paris Diderot, Paris, France.
| |
Collapse
|
121
|
Distinct bone marrow blood vessels differentially regulate haematopoiesis. Nature 2016; 532:323-8. [PMID: 27074509 PMCID: PMC6450701 DOI: 10.1038/nature17624] [Citation(s) in RCA: 499] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/25/2016] [Indexed: 12/15/2022]
Abstract
Bone marrow (BM) endothelial cells (BMECs) form a network of blood vessels (BVs) which regulate both leukocyte trafficking and hematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance these dual roles and if these events occur at the same vascular site. We found that BM stem cell maintenance and leukocyte trafficking are regulated by distinct BV types with different permeability properties. Less permeable arterial BVs maintain HSCs in a low reactive oxygen species (ROS) state, whereas the more permeable sinusoids promote HSPC activation and are the exclusive site for immature and mature leukocyte trafficking to and from the BM. A functional consequence of high BVs permeability is that exposure to blood plasma increases BM HSPC ROS levels, augmenting their migration capacity while compromising their long term repopulation and survival potential. These findings may have relevance for clinical hematopoietic stem cell transplantation and mobilization protocols.
Collapse
|
122
|
Dimasi DP, Pitson SM, Bonder CS. Examining the Role of Sphingosine Kinase-2 in the Regulation of Endothelial Cell Barrier Integrity. Microcirculation 2016; 23:248-65. [DOI: 10.1111/micc.12271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/25/2016] [Indexed: 12/30/2022]
Affiliation(s)
- David P. Dimasi
- Centre for Cancer Biology; University of South Australia and SA Pathology; Adelaide South Australia Australia
| | - Stuart M. Pitson
- Centre for Cancer Biology; University of South Australia and SA Pathology; Adelaide South Australia Australia
- School of Medicine; University of Adelaide; Adelaide South Australia Australia
- School of Biological Sciences; University of Adelaide; Adelaide South Australia Australia
| | - Claudine S. Bonder
- Centre for Cancer Biology; University of South Australia and SA Pathology; Adelaide South Australia Australia
- School of Medicine; University of Adelaide; Adelaide South Australia Australia
- School of Biological Sciences; University of Adelaide; Adelaide South Australia Australia
| |
Collapse
|
123
|
Human vascular tissue models formed from human induced pluripotent stem cell derived endothelial cells. Stem Cell Rev Rep 2016; 11:511-25. [PMID: 25190668 DOI: 10.1007/s12015-014-9549-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Here we describe a strategy to model blood vessel development using a well-defined induced pluripotent stem cell-derived endothelial cell type (iPSC-EC) cultured within engineered platforms that mimic the 3D microenvironment. The iPSC-ECs used here were first characterized by expression of endothelial markers and functional properties that included VEGF responsiveness, TNF-α-induced upregulation of cell adhesion molecules (MCAM/CD146; ICAM1/CD54), thrombin-dependent barrier function, shear stress-induced alignment, and 2D and 3D capillary-like network formation in Matrigel. The iPSC-ECs also formed 3D vascular networks in a variety of engineering contexts, yielded perfusable, interconnected lumen when co-cultured with primary human fibroblasts, and aligned with flow in microfluidics devices. iPSC-EC function during tubule network formation, barrier formation, and sprouting was consistent with that of primary ECs, and the results suggest a VEGF-independent mechanism for sprouting, which is relevant to therapeutic anti-angiogenesis strategies. Our combined results demonstrate the feasibility of using a well-defined, stable source of iPSC-ECs to model blood vessel formation within a variety of contexts using standard in vitro formats.
Collapse
|
124
|
Wang ZG, Cheng Y, Yu XC, Ye LB, Xia QH, Johnson NR, Wei X, Chen DQ, Cao G, Fu XB, Li XK, Zhang HY, Xiao J. bFGF Protects Against Blood-Brain Barrier Damage Through Junction Protein Regulation via PI3K-Akt-Rac1 Pathway Following Traumatic Brain Injury. Mol Neurobiol 2015; 53:7298-7311. [PMID: 26687235 DOI: 10.1007/s12035-015-9583-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/29/2015] [Indexed: 01/07/2023]
Abstract
Many traumatic brain injury (TBI) survivors sustain neurological disability and cognitive impairments due to the lack of defined therapies to reduce TBI-induced blood-brain barrier (BBB) breakdown. Exogenous basic fibroblast growth factor (bFGF) has been shown to have neuroprotective function in brain injury. The present study therefore investigates the beneficial effects of bFGF on the BBB after TBI and the underlying mechanisms. In this study, we demonstrate that bFGF reduces neurofunctional deficits and preserves BBB integrity in a mouse model of TBI. bFGF suppresses RhoA and upregulates tight junction proteins, thereby mitigating BBB breakdown. In vitro, bFGF exerts a protective effect on BBB by upregulating tight junction proteins claudin-5, occludin, zonula occludens-1, p120-catenin, and β-catenin under oxygen glucose deprivation/reoxygenation (OGD) in human brain microvascular endothelial cells (HBMECs). Both the in vivo and in vitro effects are related to the activation of the downstream signaling pathway, PI3K/Akt/Rac-1. Inhibition of the PI3K/Akt or Rac-1 by specific inhibitors LY294002 or si-Rac-1, respectively, partially reduces the protective effect of bFGF on BBB integrity. Overall, our results indicate that the protective role of bFGF on BBB involves the regulation of tight junction proteins and RhoA in the TBI model and OGD-induced HBMECs injury, and that activation of the PI3K/Akt /Rac-1 signaling pathway underlies these effects.
Collapse
Affiliation(s)
- Zhou-Guang Wang
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yi Cheng
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Pharmacy, Longyou People's Hospital, Quzhou, 324400, China
| | - Xi-Chong Yu
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
| | - Li-Bing Ye
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qing-Hai Xia
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
| | - Noah R Johnson
- Department of Bioengineering and the McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Xiaojie Wei
- Department of Neurosurgery, Cixi People's Hospital, Wenzhou Medical University, Ningbo, 315300, China
| | - Da-Qing Chen
- Department of Emergency, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiao-Bing Fu
- Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiao-Kun Li
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hong-Yu Zhang
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Jian Xiao
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
125
|
Yang X, Liaw L, Prudovsky I, Brooks PC, Vary C, Oxburgh L, Friesel R. Fibroblast growth factor signaling in the vasculature. Curr Atheroscler Rep 2015; 17:509. [PMID: 25813213 DOI: 10.1007/s11883-015-0509-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite their discovery as angiogenic factors and mitogens for endothelial cells more than 30 years ago, much remains to be determined about the role of fibroblast growth factors (FGFs) and their receptors in vascular development, homeostasis, and disease. In vitro studies show that members of the FGF family stimulate growth, migration, and sprouting of endothelial cells, and growth, migration, and phenotypic plasticity of vascular smooth muscle cells. Recent studies have revealed important roles for FGFs and their receptors in the regulation of endothelial cell sprouting and vascular homeostasis in vivo. Furthermore, recent work has revealed roles for FGFs in atherosclerosis, vascular calcification, and vascular dysfunction. The large number of FGFs and their receptors expressed in endothelial and vascular smooth muscle cells complicates these studies. In this review, we summarize recent studies in which new and unanticipated roles for FGFs and their receptors in the vasculature have been revealed.
Collapse
Affiliation(s)
- Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | | | | | | | | | | | | |
Collapse
|
126
|
Bouleti C, Mewton N, Germain S. The no-reflow phenomenon: State of the art. Arch Cardiovasc Dis 2015; 108:661-74. [PMID: 26616729 DOI: 10.1016/j.acvd.2015.09.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/28/2015] [Accepted: 09/07/2015] [Indexed: 02/06/2023]
Abstract
Primary percutaneous coronary intervention (PCI) is the best available reperfusion strategy for acute ST-segment elevation myocardial infarction (STEMI), with nearly 95% of occluded coronary vessels being reopened in this setting. Despite re-establishing epicardial coronary vessel patency, primary PCI may fail to restore optimal myocardial reperfusion within the myocardial tissue, a failure at the microvascular level known as no-reflow (NR). NR has been reported to occur in up to 60% of STEMI patients with optimal coronary vessel reperfusion. When it does occur, it significantly attenuates the beneficial effect of reperfusion therapy, leading to poor outcomes. The pathophysiology of NR is complex and incompletely understood. Many phenomena are known to contribute to NR, including leukocyte infiltration, vasoconstriction, activation of inflammatory pathways and cellular oedema. Vascular damage and haemorrhage may also play important roles in the establishment of NR. In this review, we describe the pathophysiological mechanisms of NR and the tools available for diagnosing it. We also describe the microvasculature and the endothelial mechanisms involved in NR, which may provide relevant therapeutic targets for reducing NR and improving the prognosis for patients.
Collapse
Affiliation(s)
- Claire Bouleti
- Service de cardiologie, hôpital Bichat, AP-HP, Paris, France; DHU FIRE, université Paris Diderot, Paris, France; Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, France; CNRS/UMR 7241, Paris, France; Inserm U 1050, Paris, France
| | - Nathan Mewton
- Hôpital cardiovasculaire Louis-Pradel, centre d'investigation clinique unité, hospices civils de Lyon, Bron, France; Inserm U 1407, Lyon, France
| | - Stéphane Germain
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, France; CNRS/UMR 7241, Paris, France; Inserm U 1050, Paris, France.
| |
Collapse
|
127
|
Zhang H, Chang H, Wang LM, Ren KF, Martins MCL, Barbosa MA, Ji J. Effect of Polyelectrolyte Film Stiffness on Endothelial Cells During Endothelial-to-Mesenchymal Transition. Biomacromolecules 2015; 16:3584-93. [PMID: 26477358 DOI: 10.1021/acs.biomac.5b01057] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT), during which endothelial cells (ECs) transdifferentiate into mesenchymal phenotype, plays a key role in the development of vascular implant complications such as endothelium dysfunction and in-stent restenosis. Substrate stiffness has been confirmed as a key factor to influence EC behaviors; however, so far, the relationship between substrate stiffness and EndMT has been rarely studied. Here, ECs were cultured on the (poly(L-lysine)/hyaluronate acid) (PLL/HA) multilayer films with controlled stiffness for 2 weeks, and their EndMT behaviors were studied. We demonstrated that ECs lost their markers (vWf and CD31) in a stiffness-dependent manner even without supplement of growth factors, and the softer film favored the maintaining of EC phenotype. Further, induced by transforming growth factor β1 (TGF-β1), ECs underwent EndMT, as characterized by losing their typical cobblestone morphology and markers and gaining smooth muscle cell markers (α-smooth muscle actin and calponin). Interestingly, stronger EndMT was observed when ECs were cultured on the stiffer film. Collectively, our findings suggest that substrate stiffness has significant effects on EndMT, and a softer substrate is beneficial to ECs by keeping their phenotype and inhibiting EndMT, which presents a new strategy for surface design of vascular implant materials.
Collapse
Affiliation(s)
- He Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, China
| | - Hao Chang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, China
| | - Li-mei Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, China
| | - Ke-feng Ren
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University , Shanghai, China
| | - M Cristina L Martins
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto , Rua do Campo Alegre, 823, 4150-180, Porto, Portugal
| | - Mário A Barbosa
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto , Rua do Campo Alegre, 823, 4150-180, Porto, Portugal
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, China
| |
Collapse
|
128
|
Regulation of Endothelial Adherens Junctions by Tyrosine Phosphorylation. Mediators Inflamm 2015; 2015:272858. [PMID: 26556953 PMCID: PMC4628659 DOI: 10.1155/2015/272858] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/16/2015] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells form a semipermeable, regulated barrier that limits the passage of fluid, small molecules, and leukocytes between the bloodstream and the surrounding tissues. The adherens junction, a major mechanism of intercellular adhesion, is comprised of transmembrane cadherins forming homotypic interactions between adjacent cells and associated cytoplasmic catenins linking the cadherins to the cytoskeleton. Inflammatory conditions promote the disassembly of the adherens junction and a loss of intercellular adhesion, creating openings or gaps in the endothelium through which small molecules diffuse and leukocytes transmigrate. Tyrosine kinase signaling has emerged as a central regulator of the inflammatory response, partly through direct phosphorylation and dephosphorylation of the adherens junction components. This review discusses the findings that support and those that argue against a direct effect of cadherin and catenin phosphorylation in the disassembly of the adherens junction. Recent findings indicate a complex interaction between kinases, phosphatases, and the adherens junction components that allow a fine regulation of the endothelial permeability to small molecules, leukocyte migration, and barrier resealing.
Collapse
|
129
|
Glial influences on BBB functions and molecular players in immune cell trafficking. Biochim Biophys Acta Mol Basis Dis 2015; 1862:472-82. [PMID: 26454208 DOI: 10.1016/j.bbadis.2015.10.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) constitutes an elaborate structure formed by specialized capillary endothelial cells, which together with pericytes and perivascular glial cells regulates the exchanges between the central nervous system (CNS) and the periphery. Intricate interactions between the different cellular constituents of the BBB are crucial in establishing a functional BBB and maintaining the delicate homeostasis of the CNS microenvironment. In this review, we discuss the role of astrocytes and microglia in inducing and maintaining barrier properties under physiological conditions as well as their involvement during neuroinflammatory pathologies. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
130
|
Broux B, Gowing E, Prat A. Glial regulation of the blood-brain barrier in health and disease. Semin Immunopathol 2015; 37:577-90. [PMID: 26245144 DOI: 10.1007/s00281-015-0516-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/15/2015] [Indexed: 12/14/2022]
Abstract
The brain is the organ with the highest metabolic demand in the body. Therefore, it needs specialized vasculature to provide it with the necessary oxygen and nutrients, while protecting it against pathogens and toxins. The blood-brain barrier (BBB) is very tightly regulated by specialized endothelial cells, two basement membranes, and astrocytic endfeet. The proximity of astrocytes to the vessel makes them perfect candidates to influence the function of the BBB. Moreover, other glial cells are also known to contribute to either BBB quiescence or breakdown. In this review, we summarize the knowledge on glial regulation of the BBB during development, in homeostatic conditions in the adult, and during neuroinflammatory responses.
Collapse
Affiliation(s)
- Bieke Broux
- Neuroimmunology Unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint Denis Street, Room R9.912, Montréal, Québec, Canada, H2X 0A9
- Department of Neuroscience, Faculté de Médecine, Université de Montréal, Montréal, Canada
- Hasselt University, Biomedical Research Institute and transnationale Universiteit Limburg, School of Life Sciences, Agoralaan, Building C, 3590, Diepenbeek, Belgium
| | - Elizabeth Gowing
- Neuroimmunology Unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint Denis Street, Room R9.912, Montréal, Québec, Canada, H2X 0A9
- Department of Neuroscience, Faculté de Médecine, Université de Montréal, Montréal, Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint Denis Street, Room R9.912, Montréal, Québec, Canada, H2X 0A9.
- Department of Neuroscience, Faculté de Médecine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
131
|
Li M, Zhao Y, Hao H, Han W, Fu X. Mesenchymal stem cell-based therapy for nonhealing wounds: today and tomorrow. Wound Repair Regen 2015; 23:465-82. [PMID: 25877885 DOI: 10.1111/wrr.12304] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/09/2015] [Accepted: 03/30/2015] [Indexed: 12/31/2022]
Abstract
Although advancements have been made with traditional therapies, the treatment of chronic nonhealing wounds still remains a tough challenge. In the past two decades, mesenchymal stem cell (MSC)-based therapy has emerged as a promising therapeutic strategy for nonhealing wounds because of their characteristics including self-renewal and a multidirectional differentiation ability and their easy collection and weak immunogenicity. There is a growing body of basic scientific studies that shed light on the functional mechanism of MSCs in modulating nonhealing wounds. Furthermore, critical advances have been achieved using MSC-based therapy in preclinical animal models as well as in clinics trials. In this present review, we summarize the mechanisms of MSCs and highlight the important preclinical and clinical trials of MSC therapy for nonhealing wounds. In particular, the combination of MSCs transplantation and tissue-engineered skin is addressed as a new strategy to optimize the delivery efficiency and therapeutic potential. Additionally, the current drawbacks of MSC therapy and the potential to further optimize the use of MSCs are implied.
Collapse
Affiliation(s)
- Meirong Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China.,Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya, Peoples Republic of China
| | - Yali Zhao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China.,Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya, Peoples Republic of China
| | - Haojie Hao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China
| | - Weidong Han
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China
| |
Collapse
|
132
|
Behl T, Kaur I, Goel H, Kotwani A. Significance of the antiangiogenic mechanisms of thalidomide in the therapy of diabetic retinopathy. Vascul Pharmacol 2015. [PMID: 26196302 DOI: 10.1016/j.vph.2015.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Diabetic retinopathy is an ocular complication associated with the chronic endocrine disorder of diabetes mellitus. Angiogenesis is adjudged as a prime modulatory event in this complication. The formation of new blood vessels on the pre-existing vasculature gives rise to an abundance of anatomical and physiological alterations which ultimately results in vision loss. The drastic consequences of this complication prompt the obligation of developing effective therapies for its cure. The existing therapy mainly includes destructive techniques such as laser photocoagulation. Owing to the various drawbacks associated with this technique, there is a need to develop alternative therapies which could halt the progression of diabetic retinopathy without causing considerable damage to the retinal cells. One such possible alternative treatment being researched upon is the antiangiogenic therapy. Since angiogenesis is a critical event during the progression of this disorder, targeting this event may perhaps prove effective in its treatment. Amongst several antiangiogenic agents, thalidomide holds a reputable position due to its effectiveness in terminating angiogenesis during various pathological conditions. This review focuses on the diverse molecular mechanisms proposed to explain the antiangiogenic properties of thalidomide and their applicability in diabetic retinopathy.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India.
| | - Ishneet Kaur
- Department of Pharmacy, Chandigarh College of Pharmacy, Mohali, Punjab, India
| | - Heena Goel
- Department of Animal Husbandry, Junga, Shimla, Himachal Pradesh, India
| | - Anita Kotwani
- Department of Pharmacology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| |
Collapse
|
133
|
Prager B, Spampinato SF, Ransohoff RM. Sphingosine 1-phosphate signaling at the blood–brain barrier. Trends Mol Med 2015; 21:354-63. [DOI: 10.1016/j.molmed.2015.03.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/24/2015] [Accepted: 03/27/2015] [Indexed: 12/20/2022]
|
134
|
Abstract
The endothelium forms a selective semi-permeable barrier controlling bidirectional transfer between blood vessel and irrigated tissues. This crucial function relies on the dynamic architecture of endothelial cell–cell junctions, and in particular, VE -cadherin-mediated contacts. VE -cadherin indeed chiefly organizes the opening and closing of the endothelial barrier, and is central in permeability changes. In this review, the way VE -cadherin-based contacts are formed and maintained is first presented, including molecular traits of its expression, partners, and signaling. In a second part, the mechanisms by which VE -cadherin adhesion can be disrupted, leading to cell–cell junction weakening and endothelial permeability increase, are described. Overall, the molecular basis for VE -cadherin control of the endothelial barrier function is of high interest for biomedical research, as vascular leakage is observed in many pathological conditions and human diseases.
Collapse
|
135
|
Abstract
Blood vascular networks in vertebrates are essential to tissue survival. Establishment of a fully functional vasculature is complex and requires a number of steps including vasculogenesis and angiogenesis that are followed by differentiation into specialized vascular tissues (i.e., arteries, veins, and lymphatics) and organ-specific differentiation. However, an equally essential step in this process is the pruning of excessive blood vessels. Recent studies have shown that pruning is critical for the effective perfusion of blood into tissues. Despite its significance, vessel pruning is the least understood process in vascular differentiation and development. Two recently published PLOS Biology papers provide important new information about cellular dynamics of vascular regression.
Collapse
Affiliation(s)
- Nicolas Ricard
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Michael Simons
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
136
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1360] [Impact Index Per Article: 151.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|
137
|
Pigment epithelium-derived factor regulates microvascular permeability through adipose triglyceride lipase in sepsis. Clin Sci (Lond) 2015; 129:49-61. [PMID: 25700221 DOI: 10.1042/cs20140631] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PEDF induces vascular hyperpermeability by targeting the ATGL receptor, causing actin rearrangement and intercellular junctions disruption through activating RhoA. This damage can be arrested by PEDF-mAb or ATGL-shRNA, which may provide new potential therapeutic strategies for hyperpermeability in sepsis.
Collapse
|
138
|
Abstract
The arrival of multicellularity in evolution facilitated cell-cell signaling in conjunction with adhesion. As the ectodomains of cadherins interact with each other directly in trans (as well as in cis), spanning the plasma membrane and associating with multiple other entities, cadherins enable the transduction of "outside-in" or "inside-out" signals. We focus this review on signals that originate from the larger family of cadherins that are inwardly directed to the nucleus, and thus have roles in gene control or nuclear structure-function. The nature of cadherin complexes varies considerably depending on the type of cadherin and its context, and we will address some of these variables for classical cadherins versus other family members. Substantial but still fragmentary progress has been made in understanding the signaling mediators used by varied cadherin complexes to coordinate the state of cell-cell adhesion with gene expression. Evidence that cadherin intracellular binding partners also localize to the nucleus is a major point of interest. In some models, catenins show reduced binding to cadherin cytoplasmic tails favoring their engagement in gene control. When bound, cadherins may serve as stoichiometric competitors of nuclear signals. Cadherins also directly or indirectly affect numerous signaling pathways (e.g., Wnt, receptor tyrosine kinase, Hippo, NFκB, and JAK/STAT), enabling cell-cell contacts to touch upon multiple biological outcomes in embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center; Program in Genes & Development, Graduate School in Biomedical Sciences, Houston, Texas, USA.
| | - Meghan T Maher
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Cara J Gottardi
- Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
139
|
Lin R, Cai J, Nathan C, Wei X, Schleidt S, Rosenwasser R, Iacovitti L. Neurogenesis is enhanced by stroke in multiple new stem cell niches along the ventricular system at sites of high BBB permeability. Neurobiol Dis 2015; 74:229-39. [DOI: 10.1016/j.nbd.2014.11.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/03/2014] [Accepted: 11/24/2014] [Indexed: 02/06/2023] Open
|
140
|
Schäker K, Bartsch S, Patry C, Stoll SJ, Hillebrands JL, Wieland T, Kroll J. The bipartite rac1 Guanine nucleotide exchange factor engulfment and cell motility 1/dedicator of cytokinesis 180 (elmo1/dock180) protects endothelial cells from apoptosis in blood vessel development. J Biol Chem 2015; 290:6408-18. [PMID: 25586182 DOI: 10.1074/jbc.m114.633701] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Engulfment and cell motility 1/dedicator of cytokinesis 180 (Elmo1/Dock180) is a bipartite guanine nucleotide exchange factor for the monomeric GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1). Elmo1/Dock180 regulates Rac1 activity in a specific spatiotemporal manner in endothelial cells (ECs) during zebrafish development and acts downstream of the Netrin-1/Unc5-homolog B (Unc5B) signaling cascade. However, mechanistic details on the pathways by which Elmo1/Dock180 regulates endothelial function and vascular development remained elusive. In this study, we aimed to analyze the vascular function of Elmo1 and Dock180 in human ECs and during vascular development in zebrafish embryos. In vitro overexpression of Elmo1 and Dock180 in ECs reduced caspase-3/7 activity and annexin V-positive cell number upon induction of apoptosis. This protective effect of Elmo1 and Dock180 is mediated by activation of Rac1, p21-activated kinase (PAK) and AKT/protein kinase B (AKT) signaling. In zebrafish, Elmo1 and Dock180 overexpression reduced the total apoptotic cell and apoptotic EC number and promoted the formation of blood vessels during embryogenesis. In conclusion, Elmo1 and Dock180 protect ECs from apoptosis by the activation of the Rac1/PAK/AKT signaling cascade in vitro and in vivo. Thus, Elmo1 and Dock180 facilitate blood vessel formation by stabilization of the endothelium during angiogenesis.
Collapse
Affiliation(s)
- Kathrin Schäker
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany, and
| | - Susanne Bartsch
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and
| | - Christian Patry
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and
| | - Sandra J Stoll
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim of Heidelberg University, 68167 Mannheim, Germany
| | - Jens Kroll
- From the Department of Vascular Biology and Tumor Angiogenesis, Center for Biomedicine and Medical Technology Mannheim (CBTM) and Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany, and
| |
Collapse
|
141
|
Abstract
Blood vessels are critical to deliver oxygen and nutrients to all of the tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier, which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and also protects the neural tissue from toxins and pathogens, and alterations of these barrier properties are an important component of pathology and progression of different neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the endothelial cells (ECs) that form the walls of the blood vessels, and these properties are regulated by interactions with different vascular, immune, and neural cells. Understanding how these different cell populations interact to regulate the barrier properties is essential for understanding how the brain functions during health and disease.
Collapse
Affiliation(s)
- Richard Daneman
- Departments of Neuroscience and Pharmacology, University of California, San Diego, San Diego, California 92093
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| |
Collapse
|
142
|
|
143
|
Chen PY, Qin L, Tellides G, Simons M. Fibroblast growth factor receptor 1 is a key inhibitor of TGFβ signaling in the endothelium. Sci Signal 2014; 7:ra90. [PMID: 25249657 DOI: 10.1126/scisignal.2005504] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Abnormal vascular homeostasis can lead to increased proliferation of smooth muscle cells and deposition of extracellular matrix, resulting in neointima formation, which contributes to vascular lumen narrowing, a pathology that underlies diseases including transplant vasculopathy, the recurrence of stenosis, and atherosclerosis. Growth of neointima is in part due to endothelial-to-mesenchymal transition (EndMT), a transforming growth factor-β (TGFβ)-driven process, which leads to increased numbers of smooth muscle cells and fibroblasts and deposition of extracellular matrix. We reported that endothelial cell-specific knockout of fibroblast growth factor receptor 1 (FGFR1) led to activation of TGFβ signaling and development of EndMT in vitro and in vivo. Furthermore, EndMT in human diseased vasculature correlated with decreased abundance of FGFR1. These findings identify FGFR1 as the key regulator of TGFβ signaling and EndMT development.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520, USA
| | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
144
|
Kondo M, Kamiya H, Himeno T, Naruse K, Nakashima E, Watarai A, Shibata T, Tosaki T, Kato J, Okawa T, Hamada Y, Isobe KI, Oiso Y, Nakamura J. Therapeutic efficacy of bone marrow-derived mononuclear cells in diabetic polyneuropathy is impaired with aging or diabetes. J Diabetes Investig 2014; 6:140-9. [PMID: 25802721 PMCID: PMC4364848 DOI: 10.1111/jdi.12272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 07/04/2014] [Accepted: 07/27/2014] [Indexed: 12/14/2022] Open
Abstract
Aims/Introduction Recent studies have shown that cell transplantation therapies, such as endothelial precursor cells, bone marrow-derived mononuclear cells (BM-MNCs) and mesenchymal stem cells, are effective on diabetic polyneuropathy through ameliorating impaired nerve blood flow in diabetic rats. Here, we investigated the effects of BM-MNCs transplantation in diabetic polyneuropathy using BM-MNCs derived from adult (16-week-old) diabetic (AD), adult non-diabetic (AN) or young (8-week-old) non-diabetic (YN) rats. Materials and Methods BM-MNCs of AD and AN were isolated after an 8-week diabetes duration. The BM-MNCs were characterized using flow cytometry analysis of cell surface markers and reverse transcription polymerase chain reaction of several cytokines. BM-MNCs or saline were injected into hind limb muscles. Four weeks later, the thermal plantar test, nerve conduction velocity, blood flow of the sciatic nerve and capillary-to-muscle fiber ratio were evaluated. Results The number of CD29+/CD90+ cells that host mesenchymal stem cells in BM-MNCs decreased in AD compared with AN or YN, and transcript expressions of basic fibroblast growth factor and nerve growth factor in BM-MNCs decreased in AD compared with AN or YN. Impaired thermal sensation, decreased blood flow of the sciatic nerve and delayed nerve conduction velocity in 8-week-diabetic rats were significantly ameliorated by BM-MNCs derived from YN, whereas BM-MNCs from AD or AN rats did not show any beneficial effect in these functional tests. Conclusions These results show that cytokine production abilities and the mesenchymal stem cell population of BM-MNCs would be modified by aging and metabolic changes in diabetes, and that these differences could explain the disparity of the therapeutic efficacy of BM-MNCs between young and adult or diabetic and non-diabetic patients in diabetic polyneuropathy.
Collapse
Affiliation(s)
- Masaki Kondo
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan ; Department of Immunology, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Hideki Kamiya
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan ; Department of CKD Initiatives, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Tatsuhito Himeno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Keiko Naruse
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Eitaro Nakashima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Atsuko Watarai
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Taiga Shibata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Takahiro Tosaki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Jiro Kato
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Tetsuji Okawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Yoji Hamada
- Department of Metabolic Medicine, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Ken-Ichi Isobe
- Department of Immunology, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Yutaka Oiso
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| | - Jiro Nakamura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine Nagoya, Japan
| |
Collapse
|
145
|
Fibroblast growth factor signaling affects vascular outgrowth and is required for the maintenance of blood vessel integrity. ACTA ACUST UNITED AC 2014; 21:1310-1317. [PMID: 25200605 DOI: 10.1016/j.chembiol.2014.07.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 06/26/2014] [Accepted: 07/14/2014] [Indexed: 01/21/2023]
Abstract
Angiogenesis contributes to the development of numerous disorders. Even though fibroblast growth factors (FGFs) were discovered as mediators of angiogenesis more than 30 years ago, their role in developmental angiogenesis still remains elusive. We use a recently described chemical probe, SSR128129E (SSR), that selectively inhibits the action of multiple FGF receptors (FGFRs), in combination with the zebrafish model to examine the role of FGF signaling in vascular development. We observe that while FGFR signaling is less important for vessel guidance, it affects vascular outgrowth and is especially required for the maintenance of blood vessel integrity by ensuring proper cell-cell junctions between endothelial cells. In conclusion, our work illustrates the power of a small molecule probe to reveal insights into blood vessel formation and stabilization and thus of broad interest to the vascular biology community.
Collapse
|
146
|
Endothelial cell FGF signaling is required for injury response but not for vascular homeostasis. Proc Natl Acad Sci U S A 2014; 111:13379-84. [PMID: 25139991 DOI: 10.1073/pnas.1324235111] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Endothelial cells (ECs) express fibroblast growth factor receptors (FGFRs) and are exquisitely sensitive to FGF signals. However, whether the EC or another vascular cell type requires FGF signaling during development, homeostasis, and response to injury is not known. Here, we show that Flk1-Cre or Tie2-Cre mediated deletion of FGFR1 and FGFR2 (Fgfr1/2(Flk1-Cre) or Fgfr1/2(Tie2-Cre) mice), which results in deletion in endothelial and hematopoietic cells, is compatible with normal embryonic development. As adults, Fgfr1/2(Flk1-Cre) mice maintain normal blood pressure and vascular reactivity and integrity under homeostatic conditions. However, neovascularization after skin or eye injury was significantly impaired in both Fgfr1/2(Flk1-Cre) and Fgfr1/2(Tie2-Cre) mice, independent of either hematopoietic cell loss of FGFR1/2 or vascular endothelial growth factor receptor 2 (Vegfr2) haploinsufficiency. Also, impaired neovascularization was associated with delayed cutaneous wound healing. These findings reveal a key requirement for cell-autonomous EC FGFR signaling in injury-induced angiogenesis, but not for vascular homeostasis, identifying the EC FGFR signaling pathway as a target for diseases associated with aberrant vascular proliferation, such as age-related macular degeneration, and for modulating wound healing without the potential toxicity associated with direct manipulation of systemic FGF or VEGF activity.
Collapse
|
147
|
Tchaicha JH, Akbay EA, Altabef A, Mikse OR, Kikuchi E, Rhee K, Liao RG, Bronson RT, Sholl LM, Meyerson M, Hammerman PS, Wong KK. Kinase domain activation of FGFR2 yields high-grade lung adenocarcinoma sensitive to a Pan-FGFR inhibitor in a mouse model of NSCLC. Cancer Res 2014; 74:4676-84. [PMID: 25035393 DOI: 10.1158/0008-5472.can-13-3218] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Somatic mutations in FGFR2 are present in 4% to 5% of patients diagnosed with non-small cell lung cancer (NSCLC). Amplification and mutations in FGFR genes have been identified in patients with NSCLCs, and clinical trials are testing the efficacy of anti-FGFR therapies. FGFR2 and other FGFR kinase family gene alterations have been found in both lung squamous cell carcinoma and lung adenocarcinoma, although mouse models of FGFR-driven lung cancers have not been reported. Here, we generated a genetically engineered mouse model (GEMM) of NSCLC driven by a kinase domain mutation in FGFR2. Combined with p53 ablation, primary grade 3/4 adenocarcinoma was induced in the lung epithelial compartment exhibiting locally invasive and pleiotropic tendencies largely made up of multinucleated cells. Tumors were acutely sensitive to pan-FGFR inhibition. This is the first FGFR2-driven lung cancer GEMM, which can be applied across different cancer indications in a preclinical setting.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma of Lung
- Animals
- Animals, Genetically Modified/genetics
- Animals, Genetically Modified/metabolism
- Antineoplastic Agents/pharmacology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Disease Models, Animal
- Female
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mutation/drug effects
- Mutation/genetics
- Protein Kinase Inhibitors/pharmacology
- Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Jeremy H Tchaicha
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Ludwig Institute for Cancer, Cambridge, Massachusetts
| | - Esra A Akbay
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Ludwig Institute for Cancer, Cambridge, Massachusetts
| | - Abigail Altabef
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Ludwig Institute for Cancer, Cambridge, Massachusetts
| | - Oliver R Mikse
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Ludwig Institute for Cancer, Cambridge, Massachusetts
| | - Eiki Kikuchi
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Ludwig Institute for Cancer, Cambridge, Massachusetts
| | - Kevin Rhee
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Ludwig Institute for Cancer, Cambridge, Massachusetts
| | - Rachel G Liao
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. The Broad Institute, Cambridge, Massachusetts
| | - Roderick T Bronson
- Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Matthew Meyerson
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. The Broad Institute, Cambridge, Massachusetts
| | - Peter S Hammerman
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. The Broad Institute, Cambridge, Massachusetts.
| | - Kwok-Kin Wong
- Department of Medicine, Dana Farber Cancer Institute, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Ludwig Institute for Cancer, Cambridge, Massachusetts. Belfer Institute for Applied Cancer Science, Boston, Massachusetts.
| |
Collapse
|
148
|
Abstract
The formation of vasculature is essential for tissue maintenance and regeneration. During development, the vasculature forms via the dual processes of vasculogenesis and angiogenesis, and is regulated at multiple levels: from transcriptional hierarchies and protein interactions to inputs from the extracellular environment. Understanding how vascular formation is coordinated in vivo can offer valuable insights into engineering approaches for therapeutic vascularization and angiogenesis, whether by creating new vasculature in vitro or by stimulating neovascularization in vivo. In this Review, we will discuss how the process of vascular development can be used to guide approaches to engineering vasculature. Specifically, we will focus on some of the recently reported approaches to stimulate therapeutic angiogenesis by recreating the embryonic vascular microenvironment using biomaterials for vascular engineering and regeneration.
Collapse
Affiliation(s)
- Kyung Min Park
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, and The Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, and The Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21208, USA
| |
Collapse
|
149
|
Sakurai T, Lanahan A, Woolls MJ, Li N, Tirziu D, Murakami M. Live cell imaging of primary rat neonatal cardiomyocytes following adenoviral and lentiviral transduction using confocal spinning disk microscopy. J Vis Exp 2014:e51666. [PMID: 24998400 PMCID: PMC4209952 DOI: 10.3791/51666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Primary rat neonatal cardiomyocytes are useful in basic in vitro cardiovascular research because they can be easily isolated in large numbers in a single procedure. Due to advances in microscope technology it is relatively easy to capture live cell images for the purpose of investigating cellular events in real time with minimal concern regarding phototoxicity to the cells. This protocol describes how to take live cell timelapse images of primary rat neonatal cardiomyocytes using a confocal spinning disk microscope following lentiviral and adenoviral transduction to modulate properties of the cell. The application of two different types of viruses makes it easier to achieve an appropriate transduction rate and expression levels for two different genes. Well focused live cell images can be obtained using the microscope's autofocus system, which maintains stable focus for long time periods. Applying this method, the functions of exogenously engineered proteins expressed in cultured primary cells can be analyzed. Additionally, this system can be used to examine the functions of genes through the use of siRNAs as well as of chemical modulators.
Collapse
Affiliation(s)
- Takashi Sakurai
- Max-Planck-Institute for Molecular Biomedicine and Institute of Cell Biology; Department of Internal Medicine, Yale Cardiovascular Research Center and Section of Cardiovascular Medicine;
| | - Anthony Lanahan
- Department of Internal Medicine, Yale Cardiovascular Research Center and Section of Cardiovascular Medicine
| | - Melissa J Woolls
- Department of Internal Medicine, Yale Cardiovascular Research Center and Section of Cardiovascular Medicine
| | - Na Li
- Department of Internal Medicine, Yale Cardiovascular Research Center and Section of Cardiovascular Medicine
| | - Daniela Tirziu
- Department of Internal Medicine, Yale Cardiovascular Research Center and Section of Cardiovascular Medicine
| | - Masahiro Murakami
- Department of Internal Medicine, Yale Cardiovascular Research Center and Section of Cardiovascular Medicine
| |
Collapse
|
150
|
Zhang X, Simons M. Receptor tyrosine kinases endocytosis in endothelium: biology and signaling. Arterioscler Thromb Vasc Biol 2014; 34:1831-7. [PMID: 24925972 DOI: 10.1161/atvbaha.114.303217] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Receptor tyrosine kinases are involved in regulation of key processes in endothelial biology, including proliferation, migration, and angiogenesis. It is now generally accepted that receptor tyrosine kinase signaling occurs intracellularly and on the plasma membrane, although many important details remain to be worked out. Endocytosis and subsequent intracellular trafficking spatiotemporally regulate receptor tyrosine kinase signaling, whereas signaling endosomes provide a platform for the compartmentalization of signaling events. This review summarizes recent advances in our understanding of endothelial receptor tyrosine kinase endocytosis and signaling using vascular endothelial growth factor receptor-2 as a paradigm.
Collapse
Affiliation(s)
- Xi Zhang
- From the Department of Cell Biology, and Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Michael Simons
- From the Department of Cell Biology, and Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|