1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Thymosin β4 protects against aortic aneurysm via endocytic regulation of growth factor signaling

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Vascular stability and tone are maintained by contractile smooth muscle cells (VSMCs). However, injury-induced growth factors stimulate a contractile-synthetic phenotypic modulation which increases susceptibility to abdominal aortic aneurysm (AAA). As a regulator of embryonic VSMC differentiation, we hypothesized that Thymosin β4 (Tβ4) may function to maintain healthy vasculature throughout postnatal life. This was supported by the identification of an interaction with low density lipoprotein receptor related protein 1 (LRP1), an endocytic regulator of platelet-derived growth factor BB (PDGF-BB) signaling and VSMC proliferation. LRP1 variants have been implicated by genome-wide association studies with risk of AAA and other arterial diseases. Tβ4-null mice displayed aortic VSMC and elastin defects that phenocopy those of LRP1 mutants, and their compromised vascular integrity predisposed them to Angiotensin II–induced aneurysm formation. Aneurysmal vessels were characterized by enhanced VSMC phenotypic modulation and augmented PDGFR-β signaling. In vitro, enhanced sensitivity to PDGF-BB upon loss of Tβ4 was associated with dysregulated endocytosis, with increased recycling and reduced lysosomal targeting of LRP1–PDGFR-β. Accordingly, the exacerbated aneurysmal phenotype in Tβ4-null mice was rescued upon treatment with the PDGFR-β antagonist Imatinib. Our study identifies Tβ4 as a key regulator of LRP1 for maintaining vascular health, and provides insights into the mechanisms of growth factor–controlled VSMC phenotypic modulation underlying aortic disease progression.

          Abstract

          Related collections

          Most cited references75

          • Record: found
          • Abstract: found
          • Article: not found

          A guided tour into subcellular colocalization analysis in light microscopy.

          It is generally accepted that the functional compartmentalization of eukaryotic cells is reflected by the differential occurrence of proteins in their compartments. The location and physiological function of a protein are closely related; local information of a protein is thus crucial to understanding its role in biological processes. The visualization of proteins residing on intracellular structures by fluorescence microscopy has become a routine approach in cell biology and is increasingly used to assess their colocalization with well-characterized markers. However, image-analysis methods for colocalization studies are a field of contention and enigma. We have therefore undertaken to review the most currently used colocalization analysis methods, introducing the basic optical concepts important for image acquisition and subsequent analysis. We provide a summary of practical tips for image acquisition and treatment that should precede proper colocalization analysis. Furthermore, we discuss the application and feasibility of colocalization tools for various biological colocalization situations and discuss their respective strengths and weaknesses. We have created a novel toolbox for subcellular colocalization analysis under ImageJ, named JACoP, that integrates current global statistic methods and a novel object-based approach.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Role of platelet-derived growth factors in physiology and medicine.

            Platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) have served as prototypes for growth factor and receptor tyrosine kinase function for more than 25 years. Studies of PDGFs and PDGFRs in animal development have revealed roles for PDGFR-alpha signaling in gastrulation and in the development of the cranial and cardiac neural crest, gonads, lung, intestine, skin, CNS, and skeleton. Similarly, roles for PDGFR-beta signaling have been established in blood vessel formation and early hematopoiesis. PDGF signaling is implicated in a range of diseases. Autocrine activation of PDGF signaling pathways is involved in certain gliomas, sarcomas, and leukemias. Paracrine PDGF signaling is commonly observed in epithelial cancers, where it triggers stromal recruitment and may be involved in epithelial-mesenchymal transition, thereby affecting tumor growth, angiogenesis, invasion, and metastasis. PDGFs drive pathological mesenchymal responses in vascular disorders such as atherosclerosis, restenosis, pulmonary hypertension, and retinal diseases, as well as in fibrotic diseases, including pulmonary fibrosis, liver cirrhosis, scleroderma, glomerulosclerosis, and cardiac fibrosis. We review basic aspects of the PDGF ligands and receptors, their developmental and pathological functions, principles of their pharmacological inhibition, and results using PDGF pathway-inhibitory or stimulatory drugs in preclinical and clinical contexts.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Pathways and mechanisms of endocytic recycling.

              Endocytic recycling is coordinated with endocytic uptake to control the composition of the plasma membrane. Although much of our understanding of endocytic recycling has come from studies on the transferrin receptor, a protein internalized through clathrin-dependent endocytosis, increased interest in clathrin-independent endocytosis has led to the discovery of new endocytic recycling systems. Recent insights into the regulatory mechanisms that control endocytic recycling have focused on recycling through tubular carriers and the return to the cell surface of cargoes that enter cells through clathrin-independent mechanisms. Recent work emphasizes the importance of regulated recycling in processes as diverse as cytokinesis, cell adhesion, morphogenesis, cell fusion, learning and memory.
                Bookmark

                Author and article information

                Contributors
                Journal
                J Clin Invest
                J Clin Invest
                J Clin Invest
                The Journal of Clinical Investigation
                American Society for Clinical Investigation
                0021-9738
                1558-8238
                17 May 2021
                17 May 2021
                17 May 2021
                17 May 2021
                : 131
                : 10
                : e127884
                Affiliations
                [1 ]Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom.
                [2 ]Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Munich, Germany.
                [3 ]Biomolecular NMR and Center for Integrated Protein Science Munich at Chemistry Department, Technical University of Munich, Garching, Munich, Germany.
                [4 ]BHF Centre of Research Excellence, Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.
                [5 ]UCL-Institute of Child Health, London, United Kingdom.
                [6 ]Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom.
                [7 ]Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom.
                Author notes
                Address correspondence to: Nicola Smart, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, United Kingdom. Phone: 44.0.1865.252365; Email: nicola.smart@ 123456dpag.ox.ac.uk .

                Authorship note: SM and SB contributed equally to this work; ANR and AJ contributed equally to this work.

                Author information
                http://orcid.org/0000-0003-4787-2749
                http://orcid.org/0000-0002-5814-7166
                http://orcid.org/0000-0002-3620-4983
                http://orcid.org/0000-0001-8621-5165
                http://orcid.org/0000-0002-3498-172X
                http://orcid.org/0000-0001-7888-7180
                http://orcid.org/0000-0002-1594-0527
                http://orcid.org/0000-0002-9715-5951
                http://orcid.org/0000-0002-1043-4342
                http://orcid.org/0000-0002-3501-7254
                Article
                127884
                10.1172/JCI127884
                8121525
                33784254
                c136068f-1ff2-4446-a0d5-6d46dd330729
                © 2021 Munshaw et al.

                This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 1 February 2019
                : 23 March 2021
                Funding
                Funded by: Medical Research Council, https://doi.org/10.13039/501100000265;
                Award ID: #18/19_MSD_1202143
                Funded by: British Heart Foundation, https://doi.org/10.13039/501100000274;
                Award ID: FS/13/4/30045
                Studentship from the Oxford Medical Research Council Doctoral Training Partnership to Sonali Munshaw
                Categories
                Research Article

                vascular biology,cardiovascular disease,signal transduction

                Comments

                Comment on this article