1
|
Schulz JA, Hartz AMS, Bauer B. ABCB1 and ABCG2 Regulation at the Blood-Brain Barrier: Potential New Targets to Improve Brain Drug Delivery. Pharmacol Rev 2023; 75:815-853. [PMID: 36973040 PMCID: PMC10441638 DOI: 10.1124/pharmrev.120.000025] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
The drug efflux transporters ABCB1 and ABCG2 at the blood-brain barrier limit the delivery of drugs into the brain. Strategies to overcome ABCB1/ABCG2 have been largely unsuccessful, which poses a tremendous clinical problem to successfully treat central nervous system (CNS) diseases. Understanding basic transporter biology, including intracellular regulation mechanisms that control these transporters, is critical to solving this clinical problem.In this comprehensive review, we summarize current knowledge on signaling pathways that regulate ABCB1/ABCG2 at the blood-brain barrier. In Section I, we give a historical overview on blood-brain barrier research and introduce the role that ABCB1 and ABCG2 play in this context. In Section II, we summarize the most important strategies that have been tested to overcome the ABCB1/ABCG2 efflux system at the blood-brain barrier. In Section III, the main component of this review, we provide detailed information on the signaling pathways that have been identified to control ABCB1/ABCG2 at the blood-brain barrier and their potential clinical relevance. This is followed by Section IV, where we explain the clinical implications of ABCB1/ABCG2 regulation in the context of CNS disease. Lastly, in Section V, we conclude by highlighting examples of how transporter regulation could be targeted for therapeutic purposes in the clinic. SIGNIFICANCE STATEMENT: The ABCB1/ABCG2 drug efflux system at the blood-brain barrier poses a significant problem to successful drug delivery to the brain. The article reviews signaling pathways that regulate blood-brain barrier ABCB1/ABCG2 and could potentially be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Julia A Schulz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Anika M S Hartz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| |
Collapse
|
2
|
Chen Y, Jiang L, Lyu K, Lu J, Long L, Wang X, Liu T, Li S. A Promising Candidate in Tendon Healing Events—PDGF-BB. Biomolecules 2022; 12:biom12101518. [PMID: 36291727 PMCID: PMC9599567 DOI: 10.3390/biom12101518] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/24/2022] Open
Abstract
Tendon injuries are one of the most common musculoskeletal disorders for which patients seek medical aid, reducing not only the quality of life of the patient but also imposing a significant economic burden on society. The administration of growth factors at the wound site is a feasible solution for enhancing tendon healing. Platelet-derived growth factor-BB (PDGF-BB) has a well-defined safety profile compared to other growth factors and has been approved by the Food and Drug Administration (FDA). The purpose of this review is to summarize the role of PDGF-BB in tendon healing through a comprehensive review of the published literature. Experimental studies suggest that PDGF-BB has a positive effect on tendon healing by enhancing inflammatory responses, speeding up angiogenesis, stimulating tendon cell proliferation, increasing collagen synthesis and increasing the biomechanics of the repaired tendon. PDGF-BB is regarded as a promising candidate in tendon healing. However, in order to realize its full potential, we still need to carefully consider and study key issues such as dose and application time in the future, so as to explore further applications of PDGF-BB in the tendon healing process.
Collapse
Affiliation(s)
- Yixuan Chen
- School of Physical Education, Southwest Medical University, Luzhou 646000, China
| | - Li Jiang
- School of Physical Education, Southwest Medical University, Luzhou 646000, China
| | - Kexin Lyu
- School of Physical Education, Southwest Medical University, Luzhou 646000, China
| | - Jingwei Lu
- School of Physical Education, Southwest Medical University, Luzhou 646000, China
| | - Longhai Long
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiaoqiang Wang
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tianzhu Liu
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
- Correspondence: (T.L.); (S.L.)
| | - Sen Li
- Spinal Surgery Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
- Correspondence: (T.L.); (S.L.)
| |
Collapse
|
3
|
Vascularization Strategies in Bone Tissue Engineering. Cells 2021; 10:cells10071749. [PMID: 34359919 PMCID: PMC8306064 DOI: 10.3390/cells10071749] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is a highly vascularized tissue, and its development, maturation, remodeling, and regeneration are dependent on a tight regulation of blood vessel supply. This condition also has to be taken into consideration in the context of the development of artificial tissue substitutes. In classic tissue engineering, bone-forming cells such as primary osteoblasts or mesenchymal stem cells are introduced into suitable scaffolds and implanted in order to treat critical-size bone defects. However, such tissue substitutes are initially avascular. Because of the occurrence of hypoxic conditions, especially in larger tissue substitutes, this leads to the death of the implanted cells. Therefore, it is necessary to devise vascularization strategies aiming at fast and efficient vascularization of implanted artificial tissues. In this review article, we present and discuss the current vascularization strategies in bone tissue engineering. These are based on the use of angiogenic growth factors, the co-implantation of blood vessel forming cells, the ex vivo microfabrication of blood vessels by means of bioprinting, and surgical methods for creating surgically transferable composite tissues.
Collapse
|
4
|
Wu K, Tang H, Lin R, Carr SG, Wang Z, Babicheva A, Ayon RJ, Jain PP, Xiong M, Rodriguez M, Rahimi S, Balistrieri F, Rahimi S, Valdez-Jasso D, Simonson TS, Desai AA, Garcia JG, Shyy JYJ, Thistlethwaite PA, Wang J, Makino A, Yuan JXJ. Endothelial platelet-derived growth factor-mediated activation of smooth muscle platelet-derived growth factor receptors in pulmonary arterial hypertension. Pulm Circ 2020; 10:2045894020948470. [PMID: 33294172 PMCID: PMC7707860 DOI: 10.1177/2045894020948470] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/15/2020] [Indexed: 12/22/2022] Open
Abstract
Platelet-derived growth factor is one of the major growth factors found in human and mammalian serum and tissues. Abnormal activation of platelet-derived growth factor signaling pathway through platelet-derived growth factor receptors may contribute to the development and progression of pulmonary vascular remodeling and obliterative vascular lesions in patients with pulmonary arterial hypertension. In this study, we examined the expression of platelet-derived growth factor receptor isoforms in pulmonary arterial smooth muscle and pulmonary arterial endothelial cells and investigated whether platelet-derived growth factor secreted from pulmonary arterial smooth muscle cell or pulmonary arterial endothelial cell promotes pulmonary arterial smooth muscle cell proliferation. Our results showed that the protein expression of platelet-derived growth factor receptor α and platelet-derived growth factor receptor β in pulmonary arterial smooth muscle cell was upregulated in patients with idiopathic pulmonary arterial hypertension compared to normal subjects. Platelet-derived growth factor activated platelet-derived growth factor receptor α and platelet-derived growth factor receptor β in pulmonary arterial smooth muscle cell, as determined by phosphorylation of platelet-derived growth factor receptor α and platelet-derived growth factor receptor β. The platelet-derived growth factor-mediated activation of platelet-derived growth factor receptor α/platelet-derived growth factor receptor β was enhanced in idiopathic pulmonary arterial hypertension-pulmonary arterial smooth muscle cell compared to normal cells. Expression level of platelet-derived growth factor-AA and platelet-derived growth factor-BB was greater in the conditioned media collected from idiopathic pulmonary arterial hypertension-pulmonary arterial endothelial cell than from normal pulmonary arterial endothelial cell. Furthermore, incubation of idiopathic pulmonary arterial hypertension-pulmonary arterial smooth muscle cell with conditioned culture media from normal pulmonary arterial endothelial cell induced more platelet-derived growth factor receptor α activation than in normal pulmonary arterial smooth muscle cell. Accordingly, the conditioned media from idiopathic pulmonary arterial hypertension-pulmonary arterial endothelial cell resulted in more pulmonary arterial smooth muscle cell proliferation than the media from normal pulmonary arterial endothelial cell. These data indicate that (a) the expression and activity of platelet-derived growth factor receptor are increased in idiopathic pulmonary arterial hypertension-pulmonary arterial smooth muscle cell compared to normal pulmonary arterial smooth muscle cell, and (b) pulmonary arterial endothelial cell from idiopathic pulmonary arterial hypertension patients secretes higher level of platelet-derived growth factor than pulmonary arterial endothelial cell from normal subjects. The enhanced secretion (and production) of platelet-derived growth factor from idiopathic pulmonary arterial hypertension-pulmonary arterial endothelial cell and upregulated platelet-derived growth factor receptor expression (and function) in idiopathic pulmonary arterial hypertension-pulmonary arterial smooth muscle cell may contribute to enhancing platelet-derived growth factor/platelet-derived growth factor receptor-associated pulmonary vascular remodeling in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Kang Wu
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- State Key Laboratory of Respiratory
Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical
University, Guangzhou, China
| | - Haiyang Tang
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- State Key Laboratory of Respiratory
Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical
University, Guangzhou, China
| | - Ruizhu Lin
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Department of Genetics and
Endocrinology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical
University, Guangzhou, China
| | - Shane G. Carr
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
| | - Ziyi Wang
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- State Key Laboratory of Respiratory
Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical
University, Guangzhou, China
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Aleksandra Babicheva
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Ramon J. Ayon
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Department of Molecular Physiology and
Biological Physics, University of Virginia, Charlottesville, USA
| | - Pritesh P. Jain
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Mingmei Xiong
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
- Department of Critical Medicine, The
Third Affiliated Hospital of Guangzhou Medical
University, Guangzhou, China
| | - Marisela Rodriguez
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Shamin Rahimi
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Francesca Balistrieri
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Shayan Rahimi
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University
of California, San Diego, La Jolla, USA
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Ankit A. Desai
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Department of Medicine, Indiana
University, Indinappolis, IN, USA
| | - Joe G.N. Garcia
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
| | - John Y.-J. Shyy
- Division of Cardiovascular Medicine,
University of California, San Diego, La Jolla, USA
| | | | - Jian Wang
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- State Key Laboratory of Respiratory
Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical
University, Guangzhou, China
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| | - Ayako Makino
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Division of Endocrinology
and Metabolism, Department of Medicine, University of California, San Diego, La
Jolla, USA
| | - Jason X.-J. Yuan
- Departments of Medicine and Physiology,
The University of Arizona, Tucson, USA
- Division of Pulmonary, Critical Care and
Sleep Medicine (Section of Physiology), University of California, San Diego, La
Jolla, USA
| |
Collapse
|
5
|
Single-Cell Receptor Quantification of an In Vitro Coculture Angiogenesis Model Reveals VEGFR, NRP1, Tie2, and PDGFR Regulation and Endothelial Heterogeneity. Processes (Basel) 2019. [DOI: 10.3390/pr7060356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing ones, is essential for both normal development and numerous pathologies. Systems biology has offered a unique approach to study angiogenesis by profiling tyrosine kinase receptors (RTKs) that regulate angiogenic processes and computationally modeling RTK signaling pathways. Historically, this systems biology approach has been applied on ex vivo angiogenesis assays, however, these assays are difficult to quantify and limited in their potential of temporal analysis. In this study, we adopted a simple two-dimensional angiogenesis assay comprised of human umbilical vein endothelial cells (HUVECs) and human dermal fibroblasts (HDFs) and examined temporal dynamics of a panel of six RTKs and cell heterogeneity up to 17 days. We observed ~2700 VEGFR1 (vascular endothelial growth factor receptor 1) per cell on 24-h-old cocultured HDF plasma membranes, which do not express VEGFR when cultured alone. We observed 4000–8100 VEGFR2 per cell on cocultured HUVEC plasma membranes throughout endothelial tube formation. We showed steady increase of platelet-derived growth factor receptors (PDGFRs) on cocultured HDF plasma membranes, and more interestingly, 1900–2900 PDGFRβ per plasma membrane were found on HUVECs within the first six hours of coculturing. These quantitative findings will offer us insights into molecular regulation during angiogenesis and help assess in vitro tube formation models and their physiological relevance.
Collapse
|
6
|
Ghali MGZ, Srinivasan VM, Johnson J, Kan P, Britz G. Therapeutically Targeting Platelet-Derived Growth Factor-Mediated Signaling Underlying the Pathogenesis of Subarachnoid Hemorrhage-Related Vasospasm. J Stroke Cerebrovasc Dis 2018; 27:2289-2295. [PMID: 30037648 DOI: 10.1016/j.jstrokecerebrovasdis.2018.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/10/2018] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Vasospasm accounts for a large fraction of the morbidity and mortality burden in patients sustaining subarachnoid hemorrhage (SAH). Platelet-derived growth factor (PDGF)-β levels rise following SAH and correlate with incidence and severity of vasospasm. METHODS The literature was reviewed for studies investigating the role of PDGF in the pathogenesis of SAH-related vasospasm and efficacy of pharmacological interventions targeting the PDGF pathway in ameliorating the same and improving clinical outcomes. RESULTS Release of blood under high pressure into the subarachnoid space activates the complement cascade, which results in release of PDGF. Abluminal contact of blood with cerebral vessels increases their contractile response to PDGF-β and thrombin, with the latter upregulating PDGF-β receptors and augmenting effects of PDGF-β. PDGF-β figures prominently in the early and late phases of post-SAH vasospasm. PDGF-β binding to the PDGF receptor-β results in receptor tyrosine kinase domain activation and consequent stimulation of intracellular signaling pathways, including p38 mitogen-activated protein kinase, phosphatidylinositol-3-kinase, Rho-associated protein kinase, and extracellular regulated kinase 1 and 2. Consequent increases in intracellular calcium and increased expression of genes mediating cellular growth and proliferation mediate PDGF-induced augmentation of vascular smooth muscle cell contractility, hypertrophy, and proliferation. CONCLUSION Treatments with statins, serine protease inhibitors, and small molecular pathway inhibitors have demonstrated varying degrees of efficacy in prevention of cerebral vasospasm, which is improved with earlier institution.
Collapse
Affiliation(s)
- Michael George Zaki Ghali
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas; Department of Neurosurgery, Houston Methodist Hospital, Houston, Texas.
| | | | - Jeremiah Johnson
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Peter Kan
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Gavin Britz
- Department of Neurosurgery, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
7
|
Folestad E, Kunath A, Wågsäter D. PDGF-C and PDGF-D signaling in vascular diseases and animal models. Mol Aspects Med 2018; 62:1-11. [PMID: 29410092 DOI: 10.1016/j.mam.2018.01.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/14/2017] [Accepted: 01/22/2018] [Indexed: 01/06/2023]
Abstract
Members of the platelet-derived growth factor (PDGF) family are well known to be involved in different pathological conditions. The cellular and molecular mechanisms induced by the PDGF signaling have been well studied. Nevertheless, there is much more to discover about their functions and some important questions to be answered. This review summarizes the known roles of two of the PDGFs, PDGF-C and PDGF-D, in vascular diseases. There are clear implications for these growth factors in several vascular diseases, such as atherosclerosis and stroke. The PDGF receptors are broadly expressed in the cardiovascular system in cells such as fibroblasts, smooth muscle cells and pericytes. Altered expression of the receptors and the ligands have been found in various cardiovascular diseases and current studies have shown important implications of PDGF-C and PDGF-D signaling in fibrosis, neovascularization, atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Erika Folestad
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anne Kunath
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Dick Wågsäter
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
8
|
Noskovičová N, Petřek M, Eickelberg O, Heinzelmann K. Platelet-Derived Growth Factor Signaling in the Lung. From Lung Development and Disease to Clinical Studies. Am J Respir Cell Mol Biol 2015; 52:263-84. [DOI: 10.1165/rcmb.2014-0294tr] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
9
|
Multifarious functions of PDGFs and PDGFRs in tumor growth and metastasis. Trends Mol Med 2013; 19:460-73. [PMID: 23773831 DOI: 10.1016/j.molmed.2013.05.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/03/2013] [Accepted: 05/09/2013] [Indexed: 01/06/2023]
Abstract
Platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) are frequently expressed in various tumors and their expression levels correlate with tumor growth, invasiveness, drug resistance, and poor clinical outcomes. Emerging experimental evidence demonstrates that PDGFs exhibit multiple functions in modulation of tumor growth, metastasis, and the tumor microenvironment by targeting malignant cells, vascular cells, and stromal cells. Understanding PDGF-PDGFR-mediated molecular signaling may provide new mechanistic rationales for optimizing current cancer therapies and the development of future novel therapeutic modalities.
Collapse
|
10
|
Yanamoto H, Kataoka H, Nakajo Y, Iihara K. The Role of the Host Defense System in the Development of Cerebral Vasospasm: Analogies between Atherosclerosis and Subarachnoid Hemorrhage. Eur Neurol 2012; 68:329-43. [DOI: 10.1159/000341336] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/24/2012] [Indexed: 01/13/2023]
|
11
|
Abstract
The family of platelet-derived growth factors (PDGFs) plays a number of critical roles in normal embryonic development, cellular differentiation, and response to tissue damage. Not surprisingly, as it is a multi-faceted regulatory system, numerous pathological conditions are associated with aberrant activity of the PDGFs and their receptors. As we and others have shown, human gliomas, especially glioblastoma, express all PDGF ligands and both the two cell surface receptors, PDGFR-α and -β. The cellular distribution of these proteins in tumors indicates that glial tumor cells are stimulated via PDGF/PDGFR-α autocrine and paracrine loops, while tumor vessels are stimulated via the PDGFR-β. Here we summarize the initial discoveries on the role of PDGF and PDGF receptors in gliomas and provide a brief overview of what is known in this field.
Collapse
Affiliation(s)
- Inga Nazarenko
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - Sanna-Maria Hede
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
- (currently) Uppsala University, Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, SE-751 85 Uppsala, Sweden
| | - Xiaobing He
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - Anna Hedrén
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - James Thompson
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
- Karolinska Healthcare Research Biobank (KHRBB), Clinical Pathology/Cytology, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| | - Mikael S. Lindström
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
| | - Monica Nistér
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:04, Karolinska University Hospital Solna, SE-17176 Stockholm, Sweden
- Karolinska Healthcare Research Biobank (KHRBB), Clinical Pathology/Cytology, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
12
|
Khachigian LM, Chesterman CN. Platelet-derived Growth Factor and its Receptor: Structure and Roles in Normal Growth and Pathology. Platelets 2009; 4:304-15. [DOI: 10.3109/09537109309013233] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
13
|
Chan-Ling T, Chu Y, Baxter L, Weible II M, Hughes S. In vivocharacterization of astrocyte precursor cells (APCs) and astrocytes in developing rat retinae: Differentiation, proliferation, and apoptosis. Glia 2009; 57:39-53. [DOI: 10.1002/glia.20733] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
14
|
Kazlauskas A. Platelet-Derived Growth Factor. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
15
|
Mottet D, Bellahcène A, Pirotte S, Waltregny D, Deroanne C, Lamour V, Lidereau R, Castronovo V. Histone Deacetylase 7 Silencing Alters Endothelial Cell Migration, a Key Step in Angiogenesis. Circ Res 2007; 101:1237-46. [DOI: 10.1161/circresaha.107.149377] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Global inhibition of class I and II histone deacetylases (HDACs) impairs angiogenesis. Herein, we have undertaken the identification of the specific HDAC(s) with activity that is necessary for the development of blood vessels. Using small interfering RNAs, we observed that HDAC7 silencing in endothelial cells altered their morphology, their migration, and their capacity to form capillary tube-like structures in vitro but did not affect cell adhesion, proliferation, or apoptosis. Among several factors known to be involved in angiogenesis, platelet-derived growth factor-B (
PDGF-B
) and its receptor (
PDGFR
-β) were the most upregulated genes following HDAC7 silencing. We demonstrated that their increased expression induced by HDAC7 silencing was partially responsible for the inhibition of endothelial cell migration. In addition, we have also shown that treatment of endothelial cells with phorbol 12-myristate 13-acetate resulted in the exportation of HDAC7 out of the nucleus through a protein kinase C/protein kinase D activation pathway and induced, similarly to HDAC7 silencing, an increase in PDGF-B expression, as well as a partial inhibition of endothelial cell migration. Collectively, these data identified HDAC7 as a key modulator of endothelial cell migration and hence angiogenesis, at least in part, by regulating PDGF-B/PDGFR-β gene expression. Because angiogenesis is required for tumor progression, HDAC7 may represent a rational target for therapeutic intervention against cancer.
Collapse
Affiliation(s)
- Denis Mottet
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| | - Akeila Bellahcène
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| | - Sophie Pirotte
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| | - David Waltregny
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| | - Christophe Deroanne
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| | - Virginie Lamour
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| | - Rosette Lidereau
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| | - Vincent Castronovo
- From the Metastasis Research Laboratory (D.M., A.B., S.P., D.W., V.L., V.C.) and Laboratory of Connective Tissue Biology (C.D.), University of Liège, Belgium; and Laboratory of Oncology (R.L.), INSERM E0017, Centre René Huguenin and Institut National de la Santé et de la Recherche Médicale, U735, St Cloud, France
| |
Collapse
|
16
|
Uchida K, Urabe K, Naruse K, Ujihira M, Mabuchi K, Itoman M. Comparison of the cytokine-induced migratory response between primary and subcultured populations of rat mesenchymal bone marrow cells. J Orthop Sci 2007; 12:484-92. [PMID: 17909935 DOI: 10.1007/s00776-007-1159-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Accepted: 06/15/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND The aim of this study was to investigate the phenotypic differences of primary rat mesenchymal bone marrow cells (MBMCs) and subcultured cells, the influence of subculture and cell density on the cellular phenotypes, and the difference in the migratory responses of these cells to cytokines. METHODS MBMCs were isolated from 8-week-old Wistar rats, and the cells were cultured for 1 week (passage 0, P0) or 3 weeks (P0-3W). P0 cells were subcultured for 1 week (P1). P1 cells were subcultured at several cell densities for 1 week (P2). Cell size and granularity were analyzed by flow cytometry. The gene expression characteristics of these cells were analyzed by reverse transcription polymerase chain reaction. Cell migration to bone morphogenetic protein-2 (BMP-2), fibroblast growth factor-2 (FGF-2), and platelet-derived growth factor-bb (PDGF-bb) was evaluated using a Boyden chamber. RESULTS Three morphologically distinct populations in P0 and two in P2 were detected. The levels of human rapidly self-renewing cell-related marker genes in P0 were more highly expressed than in P2. Mesenchymal stem cell-associated markers were expressed at the same level in P0 and P2. The gene expression levels of immature oligodendrocyte precursor cell markers in P0 were higher than those in P2, whereas those of smooth muscle cell markers and osteoblastic cell markers in P0 were lower than those in P2. Subculture decreased the gene expression levels of human rapidly self-renewing cell-associated markers. Cell migration of P0 cells was stimulated by PDGF-bb but not by BMP-2 or FGF-2. In contrast, PDGF-bb, BMP-2, and FGF-2 all stimulated cell migration of P2. CONCLUSION The types of cells in populations of primary and subcultured rat MBMCs were different, and the distribution of each cell population appeared to be changed by the culture conditions. The cell migration effect by PDGF-bb, BMP-2, and FGF-2 differed between the primary and subcultured MBMCs.
Collapse
Affiliation(s)
- K Uchida
- Department of Biomedical Engineering, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Hasebe R, Kimura T, Nakamura K, Ochiai K, Okazaki K, Wada R, Umemura T. Differential susceptibility of equine and mouse brain microvascular endothelial cells to equine herpesvirus 1 infection. Arch Virol 2005; 151:775-86. [PMID: 16328147 DOI: 10.1007/s00705-005-0653-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 08/16/2005] [Indexed: 10/25/2022]
Abstract
Equine herpesvirus 1 (EHV-1) shows endotheliotropism in the central nervous system (CNS) of infected horses. However, infection of endothelial cells has not been observed in the CNS of infected mice. To explore the basis for this difference in endotheliotropism, we compared the susceptibility of equine brain microvascular endothelial cells (EBMECs) and mouse brain microvascular endothelial cells (MBMECs) to EHV-1 infection. The kinetics of viral growth in EBMECs was typical of a fully productive infection whereas viral infection in MBMECs seemed to be nonproductive. Immunofluorescence microscopy using anti-EHV-1 polyclonal antibody demonstrated viral antigen in infected EBMECs, but not infected MBMECs. EHV-1 immediate early (IE), early (ICP0), and late (gB, gD and gK) transcripts were expressed in infected EBMECs. However, none of these genes was detected in infected MBMECs by reverse transcription-polymerase chain reaction. Electron microscopic examination at the stage of viral entry showed that viral particles were present within uncoated vesicles in the cytoplasm of EBMECs, but absent from those of MBMECs. These results suggest that viral entry is an important determinant of the susceptibility of EBMECs and MBMECs to EHV-1 infection.
Collapse
Affiliation(s)
- R Hasebe
- Laboratory of Comparative Pathology, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
18
|
Hoffmann J, Feng Y, vom Hagen F, Hillenbrand A, Lin J, Erber R, Vajkoczy P, Gourzoulidou E, Waldmann H, Giannis A, Wolburg H, Shani M, Jaeger V, Weich HA, Preissner KT, Hoffmann S, Deutsch U, Hammes HP. Endothelial survival factors and spatial completion, but not pericyte coverage of retinal capillaries determine vessel plasticity. FASEB J 2005; 19:2035-6. [PMID: 16215210 DOI: 10.1096/fj.04-2109fje] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pericyte loss and capillary regression are characteristic for incipient diabetic retinopathy. Pericyte recruitment is involved in vessel maturation, and ligand-receptor systems contributing to pericyte recruitment are survival factors for endothelial cells in pericyte-free in vitro systems. We studied pericyte recruitment in relation to the susceptibility toward hyperoxia-induced vascular remodeling using the pericyte reporter X-LacZ mouse and the mouse model of retinopathy of prematurity (ROP). Pericytes were found in close proximity to vessels, both during formation of the superficial and the deep capillary layers. When exposure of mice to the ROP was delayed by 24 h, i.e., after the deep retinal layer had formed [at postnatal (p) day 8], preretinal neovascularizations were substantially diminished at p18. Mice with a delayed ROP exposure had 50% reduced avascular zones. Formation of the deep capillary layers at p8 was associated with a combined up-regulation of angiopoietin-1 and PDGF-B, while VEGF was almost unchanged during the transition from a susceptible to a resistant capillary network. Inhibition of Tie-2 function either by soluble Tie-2 or by a sulindac analog, an inhibitor of Tie-2 phosphorylation, resensitized retinal vessels to neovascularizations due to a reduction of the deep capillary network. Inhibition of Tie-2 function had no effect on pericyte recruitment. Our data indicate that the final maturation of the retinal vasculature and its resistance to regressive signals such as hyperoxia depend on the completion of the multilayer structure, in particular the deep capillary layers, and are independent of the coverage by pericytes.
Collapse
Affiliation(s)
- J Hoffmann
- Medical Clinic and Policlinic 3, Justus-Liebig University Giessen, Giessen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Li X, Tjwa M, Moons L, Fons P, Noel A, Ny A, Zhou JM, Lennartsson J, Li H, Luttun A, Pontén A, Devy L, Bouché A, Oh H, Manderveld A, Blacher S, Communi D, Savi P, Bono F, Dewerchin M, Foidart JM, Autiero M, Herbert JM, Collen D, Heldin CH, Eriksson U, Carmeliet P. Revascularization of ischemic tissues by PDGF-CC via effects on endothelial cells and their progenitors. J Clin Invest 2005; 115:118-27. [PMID: 15630451 PMCID: PMC535797 DOI: 10.1172/jci19189] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2003] [Accepted: 11/09/2004] [Indexed: 12/14/2022] Open
Abstract
The angiogenic mechanism and therapeutic potential of PDGF-CC, a recently discovered member of the VEGF/PDGF superfamily, remain incompletely characterized. Here we report that PDGF-CC mobilized endothelial progenitor cells in ischemic conditions; induced differentiation of bone marrow cells into ECs; and stimulated migration of ECs. Furthermore, PDGF-CC induced the differentiation of bone marrow cells into smooth muscle cells and stimulated their growth during vessel sprouting. Moreover, delivery of PDGF-CC enhanced postischemic revascularization of the heart and limb. Modulating the activity of PDGF-CC may provide novel opportunities for treating ischemic diseases.
Collapse
Affiliation(s)
- Xuri Li
- The Center for Transgene Technology and Gene Therapy, Flanders Interuniversitary Institute for Biotechnology (VIB), University of Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Langley RR, Fan D, Tsan RZ, Rebhun R, He J, Kim SJ, Fidler IJ. Activation of the platelet-derived growth factor-receptor enhances survival of murine bone endothelial cells. Cancer Res 2004; 64:3727-30. [PMID: 15172974 DOI: 10.1158/0008-5472.can-03-3863] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The activation of the microvascular endothelial cell platelet-derived growth factor (PDGF) receptor (PDGF-R) by PDGF has been implicated in neoplastic angiogenesis. Here, we established cultures of murine bone microvascular endothelial cells and examined their response to stimulation with PDGF BB ligand and to blockade of PDGF-R signaling with the tyrosine kinase inhibitor STI571 (Gleevec). The addition of STI571 to cultures of bone endothelial cells blocked PDGF BB-induced phosphorylation in a dose-dependent manner and completely abrogated the activation of downstream targets Akt and ERK1/2. Coadministration of STI571 and Taxol also induced the activation of procaspase-3 and significant apoptosis. These data suggest that phosphorylation of PDGF-R stimulates survival pathways in bone endothelial cells and that by selectively inhibiting PDGF-R signaling with STI571, the cells are rendered sensitive to Taxol treatment. The therapeutic combination of STI571 and Taxol may be a powerful tool for targeting tumor-associated endothelial cells in the skeletal compartment.
Collapse
Affiliation(s)
- Robert R Langley
- Department of Cancer Biology, The University of Texas M D Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Kastin AJ, Akerstrom V, Hackler L, Pan W. Different mechanisms influencing permeation of PDGF-AA and PDGF-BB across the blood-brain barrier. J Neurochem 2003; 87:7-12. [PMID: 12969247 DOI: 10.1046/j.1471-4159.2003.01933.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Platelet-derived growth factor (PDGF) exerts neurotrophic and neuromodulatory effects on the CNS. To determine the permeability of the blood-brain barrier (BBB) to PDGF, we examined the blood-to-brain influx of radioactively labeled PDGF isoforms (PDGF-AA and PDGF-BB) by multiple-time regression analysis after intravenous (i.v.) injection and by in-situ perfusion, and also determined the physicochemical characteristics which affect their permeation across the BBB, including lipophilicity (measured by octanol:buffer partition coefficient), hydrogen bonding (measured by differences in octanol : buffer and isooctane : buffer partition coefficients), serum protein binding (measured by capillary electrophoresis), and stability of PDGF in blood 10 min after i.v. injection (measured by HPLC). After i.v. bolus injection, neither 125I-PDGF-AA nor 125I-PDGF-BB crossed the BBB, their influx rates being similar to that of the vascular marker 99mTc-albumin. 125I-PDGF-AA degraded significantly faster in blood than 125I-PDGF-BB. PDGF-BB, however, was completely bound to a large protein in serum whereas PDGF-AA showed no binding. Thus, degradation might explain the poor blood-to-brain influx of PDGF-AA, whereas protein binding could explain the poor influx of circulating PDGF-BB. Despite their lack of permeation in the intact mouse, both 125I-PDGF-AA and 125I-PDGF-BB entered the brain by perfusion in blood-free buffer, and the significantly faster rate of 125I-PDGF-AA than 125I-PDGF-BB may be explained by the lower hydrogen bonding potential of 125I-PDGF-AA. Thus, the lack of significant distribution of PDGF from blood to brain is not because of the intrinsic barrier function of the BBB but probably because of degradation and protein binding. Information from these studies could be useful in the design of analogues for delivery of PDGF as a therapeutic agent.
Collapse
Affiliation(s)
- Abba J Kastin
- VA Medical Center, New Orleans, Louisiana 70112-1262, USA.
| | | | | | | |
Collapse
|
22
|
Uhl E, Rösken F, Sirsjö A, Messmer K. Influence of platelet-derived growth factor on microcirculation during normal and impaired wound healing. Wound Repair Regen 2003; 11:361-7. [PMID: 12950640 DOI: 10.1046/j.1524-475x.2003.11508.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The aim of the current study was to evaluate the influence of platelet-derived growth factor (PDGF) on skin microcirculation during normal and impaired wound healing. Secondary healing wounds were created on the ears of hairless mice and treated once with 3 microg of PDGF-BB immediately after wound creation. Intravital fluorescence microscopy was used to quantify reepithelialization, revascularization, vessel diameters, vascular permeability, and leukocyte-endothelium interactions up to 24 days after wound creation. Microvascular perfusion was assessed by laser Doppler flowmetry. Wound healing was studied in normal (n = 15) and ischemic skin tissue (n = 15) as well as in mice (n = 17) rendered hyperglycemic by an intravenous injection of streptozotocin 7 days prior to wound creation. Treatment with PDGF accelerated reepithelialization and reduced the time for complete wound closure in ischemic skin from 14.9 +/- 2.5 (control) to 12.3 +/- 1.8 days (p < 0.03), and in hyperglycemic animals from 15.0 +/- 2.4 (control) to 12.0 +/- 3.0 days (p < 0.04). Revascularization of these wounds was also significantly enhanced after PDFG application. No other parameters were influenced by the treatment. Normal wound healing was not affected. This study confirms the positive influence of PDGF on wound healing under pathophysiological conditions. The effects in this model seem to be primarily due to the mitogenic potency of PDGF on keratinocytes and endothelial cells. A significant effect on leukocyte activation during the inflammatory process was not observed.
Collapse
Affiliation(s)
- Eberhard Uhl
- Department of Neurosurgery, Ludwig-Maximilians-University, Grosshadern University Hospital, Marchioninistrasse 15, D-81377 Munich, Germany.
| | | | | | | |
Collapse
|
23
|
Vinores SA, Seo MS, Derevjanik NL, Campochiaro PA. Photoreceptor-specific overexpression of platelet-derived growth factor induces proliferation of endothelial cells, pericytes, and glial cells and aberrant vascular development: an ultrastructural and immunocytochemical study. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 140:169-83. [PMID: 12586423 DOI: 10.1016/s0165-3806(02)00581-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Platelet-derived growth factor (PDGF) is necessary for the normal development of the retinal vasculature and its overexpression is likely to contribute to proliferative retinal disorders, such as proliferative vitreoretinopathy. Transgenic mice that overexpress PDGF-B in the photoreceptors (rho/PDGF-B mice) develop traction retinal detachment. In the present study, a detailed histopathological analysis was performed in rho/PDGF-B mice. In these transgenic mice, endothelial cells, pericytes, and glial cells begin to proliferate at postnatal day 7 (P7). All three cell types increase in numbers, forming a highly vascularized cell mass, which reaches a maximum thickness at P14. Cords of endothelial cells and glia invade the retina and exert traction, generating retinal folds; however, the deep capillary bed never forms. Griffonia simplicifolia isolectin B4 (GSA)-positive endothelial cells form tubes and penetrate the retina to the level of the outer plexiform layer, but they never interconnect to form the deep capillary bed. The vessels within the cell mass are patent, but have a very immature morphology. They often are thin-walled with fenestrations. Pericytes and glial cells are usually found in clusters and are not associated with the abnormal vessels. The lack of this association may account for the failure to form a mature vasculature.
Collapse
Affiliation(s)
- Stanley A Vinores
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287-9289, USA.
| | | | | | | |
Collapse
|
24
|
Faehling M, Kroll J, Föhr KJ, Fellbrich G, Mayr U, Trischler G, Waltenberger J. Essential role of calcium in vascular endothelial growth factor A-induced signaling: mechanism of the antiangiogenic effect of carboxyamidotriazole. FASEB J 2002; 16:1805-7. [PMID: 12354692 DOI: 10.1096/fj.01-0938fje] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Vascular endothelial growth factor-A (VEGF-A) plays a major role in tumor angiogenesis and raises the concentration of intracellular free calcium ([Ca2+]i). Carboxyamidotriazole (CAI), an inhibitor of calcium influx and of angiogenesis, is under investigation as a tumoristatic agent. We studied the effect of CAI and the role of [Ca2+]i in VEGF-A signaling in human endothelial cells. VEGF-A induced a biphasic [Ca2+]i signal. VEGF-A increased the level of intracellular inositol 1,4,5-trisphosphate (IP3), which suggests that VEGF-A releases Ca2+ from IP3-sensitive stores and induces store-operated calcium influx. Reduction of either extracellular or intracellular free Ca2+ inhibited VEGF-A-induced proliferation. CAI inhibited IP3 formation, both phases of the calcium signal, nitric oxide (NO) release, and proliferation induced by VEGF-A. CAI prevented neither activation of VEGF receptor-2 (VEGFR-2) (KDR/Flk-1), phospholipase C-g, or mitogen-activated protein kinase (MAP kinase) nor translocation of nuclear factor of activated T cells (NFAT). We conclude that calcium signaling is necessary for VEGF-A-induced proliferation. MAP kinase activation occurs independently of [Ca2+]i but is not sufficient to induce proliferation in the absence of calcium signaling. Inhibition of the VEGF-A-induced [Ca2+]i signal and proliferation by CAI can be explained by inhibition of IP3 formation and may contribute to the antiangiogenic action of CAI. Calcium-dependent NO formation may represent a link between calcium signaling and proliferation.
Collapse
Affiliation(s)
- Martin Faehling
- Department of Internal Medicine II, University of Ulm, 89081 Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Cao R, Bråkenhielm E, Li X, Pietras K, Widenfalk J, Ostman A, Eriksson U, Cao Y. Angiogenesis stimulated by PDGF-CC, a novel member in the PDGF family, involves activation of PDGFR-alphaalpha and -alphabeta receptors. FASEB J 2002; 16:1575-83. [PMID: 12374780 DOI: 10.1096/fj.02-0319com] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A newly discovered PDGF isoform, PDGF-CC, is expressed in actively angiogenic tissues such as placenta, some embryonic tissues, and tumors. We test the possibility that PDGF-CC promotes angiogenesis in vivo. The core domain (mature form) of human PDGF-CC is sufficiently potent to stimulate neovascularization in the mouse cornea. The corneal angiogenic response induced by PDGF-CC is robust although the area of neovascularization is smaller than those of FGF-2- and VEGF-stimulated angiogenesis. Similarly, PDGF-BB and PDGF-AB induce angiogenic responses virtually indistinguishable from PDGF-CC-stimulated vessels. In contrast, PDGF-AA displays only a weak angiogenic response in the mouse cornea. Although there was no significant difference in incorporation of mural cells to the newly formed blood vessels induced by PDGF-BB and -CC, the percentage of mural cell positive vessels induced by PDGF-AA was greater than those induced by FGF-2, PDGF-BB, and PDGF-CC. In the developing chick embryo, PDGF-CC induced branch sprouts from established blood vessels. In PDGF receptor-transfected endothelial cells, PDGF-CC activated the PDGF receptor alpha subunit (PDGFR-alpha). PDGF-CC, but not PDGF-AA, was able to activate PDGFR-beta receptor in endothelial cells that coexpress both alpha and beta forms of receptors. Thus, the PDGF-CC-mediated angiogenic response is most likely transduced by PDGF-alphaalpha and -alphabeta receptors. These data demonstrate that the PDGF family is a complex and important group of proangiogenic factors.
Collapse
MESH Headings
- Allantois/blood supply
- Allantois/drug effects
- Animals
- Blood Vessels/drug effects
- Blood Vessels/metabolism
- Chick Embryo
- Chorion/blood supply
- Chorion/drug effects
- Cornea/blood supply
- Cornea/drug effects
- Cornea/metabolism
- Endothelial Growth Factors/pharmacology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Gene Expression
- Humans
- Immunohistochemistry
- Intercellular Signaling Peptides and Proteins/pharmacology
- Lymphokines/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/chemically induced
- Neovascularization, Pathologic/metabolism
- Phosphorylation/drug effects
- Platelet Endothelial Cell Adhesion Molecule-1/analysis
- Platelet-Derived Growth Factor/metabolism
- Platelet-Derived Growth Factor/pharmacology
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Tyrosine/metabolism
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- Renhai Cao
- Microbiology and Tumor Biology Center, Karolinska Institute, S-171 77 Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Hyperglycaemia appears to be a critical factor in the aetiology of diabetic retinopathy and initiates downstream events including: basement membrane thickening, pericyte drop out and retinal capillary non-perfusion. More recently, focus has been directed to the molecular basis of the disease process in diabetic retinopathy. Of particular importance in the development and progression of diabetic retinopathy is the role of growth factors (eg vascular endothelial growth factor, placenta growth factor and pigment epithelium-derived factor) together with specific receptors and obligate components of the signal transduction pathway needed to support them. Despite these advances there are still a number of important questions that remain to be answered before we can confidently target pathological signals. How does hyperglycaemia regulate retinal vessels? Which growth factors are most important and at what stage of retinopathy do they operate? What is the preferred point in the growth factor signalling cascade for therapeutic intervention? Answers to these questions will provide the basis for new therapeutic interventions in a debilitating ocular condition.
Collapse
Affiliation(s)
- J Cai
- Department of Optometry and Vision Sciences, Cardiff University, UK
| | | |
Collapse
|
27
|
Nguyen LL, D'Amore PA. Cellular interactions in vascular growth and differentiation. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 204:1-48. [PMID: 11243594 DOI: 10.1016/s0074-7696(01)04002-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In nature, mammalian cells do not exist in isolation, but rather are involved in interactions with other cells and matrix. In this review, several aspects of cellular interactions that are important in vascular growth and development will be highlighted. The cardiovascular system is the earliest to develop in the embryo. A number of growth factors and their receptors mediate the complex stages of migration, assembly, organization, and stabilization of developing vessels. In the adult organism, normal angiogenesis is restricted primarily to tissue growth (such as muscle and fat), the wound healing process and the female reproductive system. However, pathological angiogenesis, such as with tumor growth, diabetic retinopathy, and arthritis, is of great concern. The identification and/or development of exogenous and endogenous angiogenesis inhibitors has added to the understanding of these pathological processes. In addition to cellular interactions via ligands and receptors, cells also interact directly through physical contacts. These interactions facilitate anchorage, communication, and permeability. Since vessels serve as non-leaky conduits for blood flow as well as interfaces for molecular diffusion, the physical interactions between the cells that make up vessels must be specific for the function at hand. Permeability is a specialized function of vessels and is mediated by intracellular mechanisms and intercellular interactions. Cells also interact with the surrounding extracellular matrix. Integrin-matrix interaction is a two-way exchange critical for angiogenesis. Matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases play major roles in embryonic remodeling, adult injury, and pathological conditions. Several experimental model systems have been useful in our understanding of cellular interactions. These in vitro models incorporate heterotypic cell-cell interactions and/or allow cell-matrix interactions to occur.
Collapse
Affiliation(s)
- L L Nguyen
- Schepens Eye Research Institute and Department of Surgery, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
28
|
HUGHES SUZANNE, CHAN-LING TAILOI. Roles of Endothelial Cell Migration and Apoptosis in Vascular Remodeling during Development of the Central Nervous System. Microcirculation 2000. [DOI: 10.1111/j.1549-8719.2000.tb00131.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Affiliation(s)
- M E Beckner
- Robert C. Byrd Health Science Center, West Virginia University, Morgantown, USA.
| |
Collapse
|
30
|
Abstract
Platelet-derived growth factor (PDGF) is a major mitogen for connective tissue cells and certain other cell types. It is a dimeric molecule consisting of disulfide-bonded, structurally similar A- and B-polypeptide chains, which combine to homo- and heterodimers. The PDGF isoforms exert their cellular effects by binding to and activating two structurally related protein tyrosine kinase receptors, denoted the alpha-receptor and the beta-receptor. Activation of PDGF receptors leads to stimulation of cell growth, but also to changes in cell shape and motility; PDGF induces reorganization of the actin filament system and stimulates chemotaxis, i.e., a directed cell movement toward a gradient of PDGF. In vivo, PDGF has important roles during the embryonic development as well as during wound healing. Moreover, overactivity of PDGF has been implicated in several pathological conditions. The sis oncogene of simian sarcoma virus (SSV) is related to the B-chain of PDGF, and SSV transformation involves autocrine stimulation by a PDGF-like molecule. Similarly, overproduction of PDGF may be involved in autocrine and paracrine growth stimulation of human tumors. Overactivity of PDGF has, in addition, been implicated in nonmalignant conditions characterized by an increased cell proliferation, such as atherosclerosis and fibrotic conditions. This review discusses structural and functional properties of PDGF and PDGF receptors, the mechanism whereby PDGF exerts its cellular effects, and the role of PDGF in normal and diseased tissues.
Collapse
Affiliation(s)
- C H Heldin
- Ludwig Institute for Cancer Research, Biomedical Center, and Department of Pathology, University Hospital, Uppsala, Sweden.
| | | |
Collapse
|
31
|
Selheim F, Holmsen H, Vassbotn FS. Platelet-derived growth factor inhibits platelet activation in heparinized whole blood. Thromb Res 1999; 95:185-96. [PMID: 10498388 DOI: 10.1016/s0049-3848(99)00038-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We previously have demonstrated that human platelets have functionally active platelet-derived growth factor alpha-receptors. Studies with gel-filtered platelets showed that an autocrine inhibition pathway is transduced through this tyrosine kinase receptor during platelet activation. The physiological significance of this inhibitory effect of platelet-derived growth factor on gel-filtered platelets activation is, however, not known. In the present study, we investigated whether platelet-derived growth factor inhibits platelet activation under more physiological conditions in heparinized whole blood, which represents a more physiological condition than gel-filtered platelets. Using flow cytometric assays, we demonstrate here that platelet-derived growth factor inhibits thrombin-, thrombin receptor agonist peptide SFLLRN-, and collagen-induced platelet aggregation and shedding of platelet-derived microparticles from the platelet plasma membrane during platelet aggregation in stirred heparinized whole blood. The inhibitory effect of platelet-derived growth factor was dose dependent. However, under nonaggregating conditions (no stirring), we could not demonstrate any significant effect of platelet-derived growth factor on thrombin- and thrombin receptor agonist peptide-induced platelet surface expression of P-selectin. Our results demonstrate that platelet-derived growth factor appears to be a true antithrombotic agent only under aggregating conditions in heparinized whole blood.
Collapse
Affiliation(s)
- F Selheim
- Department of Biochemistry and Molecular Biology, University of Bergen, Norway.
| | | | | |
Collapse
|
32
|
IRUELA-ARISPE MLUISA, RODRIGUEZ-MANZANEQUE JUANCARLOS, ABU-JAWDEH GRAZIELLA. Endometrial Endothelial Cells Express Estrogen and Progesterone Receptors and Exhibit a Tissue Specific Response to Angiogenic Growth Factors. Microcirculation 1999. [DOI: 10.1111/j.1549-8719.1999.tb00095.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
33
|
Kajihara T, Ohnishi T, Arakaki N, Semba I, Daikuhara Y. Expression of hepatocyte growth factor/scatter factor and c-Met in human dental papilla and fibroblasts from dental papilla. Arch Oral Biol 1999; 44:135-47. [PMID: 10206332 DOI: 10.1016/s0003-9969(98)00101-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hepatocyte growth factor/scatter factor (HGF/SF), a broad-spectrum and multifunctional cytokine, is essential for the development of tissues including tooth. Here it was found that the HGF/SF content of human dental papillae obtained from 8 to 16-year-old individuals decreased significantly with age. Cultured fibroblasts prepared from the dental papillae of individuals of different ages produced HGF/SF at almost the same rate, but the sensitivities of the cells to interleukin-1alpha and tumour necrosis factor-alpha for the production of HGF/SF increased with age. Generally, mesenchymal cells such as fibroblasts produce HGF/SF but do not express c-Met, a receptor for HGF/SF, yet fibroblasts in dental papilla and cultured fibroblasts prepared from dental papilla did express c-Met, as determined by immunohistochemistry, in situ hybridization and reverse transcription-polymerase chain reaction. Recombinant human [125I]iodo-HGF/SF specifically bound to cell-surface macromolecules with a mol. wt of 146,000, which is the same as that of the beta-subunit of c-Met. The physiological role of c-Met on fibroblasts in dental papilla is unknown, but the addition of 2 ng of HGF/SF per ml to the culture medium significantly stimulated DNA synthesis in the cells, as determined by pulse labelling with [3H]thymidine. Exogenous HGF/SF also stimulated secretion by the cells of vascular endothelial growth factor, a cytokine that induces blood vessel-formation. These results suggest that HGF/SF may be involved in tooth development via autocrine mechanisms.
Collapse
Affiliation(s)
- T Kajihara
- Department of Biochemistry, Kagoshima University Dental School, Japan
| | | | | | | | | |
Collapse
|
34
|
Nicosia RF, Villaschi S. Autoregulation of angiogenesis by cells of the vessel wall. INTERNATIONAL REVIEW OF CYTOLOGY 1998; 185:1-43. [PMID: 9750264 DOI: 10.1016/s0074-7696(08)60148-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The cells of the vessel wall can regulate angiogenesis by producing growth factors, proteolytic enzymes, extracellular matrix components, cell adhesion molecules, and vasoactive factors. This property enables preexisting blood vessels to generate new vessels in the absence of exogenous angiogenic stimuli. Vascular autoregulation of angiogenesis can be studied by culturing rat aortic or venous explants in collagen gels under serum-free conditions. In this system, the combined effect of injury and exposure of explants to collagen triggers a self-limited angiogenic response. Interactions among endothelial cells, smooth muscle cells, and fibroblasts play a critical role in the regulation of this process. This chapter reviews the literature on angiogenesis, focusing on the vessel wall as a highly specialized and plastic tissue capable of regenerating itself through autocrine, paracrine, and juxtacrine mechanisms.
Collapse
Affiliation(s)
- R F Nicosia
- Department of Pathology, Allegheny University of the Health Sciences, Philadelphia, Pennsylvania 19102, USA
| | | |
Collapse
|
35
|
Abstract
Angiogenic cytokines constitute a potentially novel form of therapy for patients with cardiovascular disease. The feasibility of using recombinant formulations of angiogenic growth factors to expedite and/or augment collateral artery development in animal models of myocardial and hindlimb ischemia--'therapeutic angiogenesis'--has now been well established. These studies have suggested that two angiogenic growth factors in particular--basic fibroblast growth factor and vascular endothelial growth factor--are sufficiently potent to merit further investigation. More recently, experiments performed in our laboratory have indicated that, in the case of vascular endothelial growth factor--a secreted protein--similar results may be achieved by percutaneous arterial gene transfer. Further laboratory and clinical studies may yield promising insights into the fundamental basis for native as well as therapeutic angiogenesis, and at the same time more explicitly define the manner in which therapeutic angiogenesis may be successfully incorporated into clinical practice.
Collapse
Affiliation(s)
- J M Isner
- Department of Medicine (Cardiology), St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135-2997, USA
| |
Collapse
|
36
|
Crosby JR, Seifert RA, Soriano P, Bowen-Pope DF. Chimaeric analysis reveals role of Pdgf receptors in all muscle lineages. Nat Genet 1998; 18:385-8. [PMID: 9537425 DOI: 10.1038/ng0498-385] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Blood vessels originate as simple endothelial cell tubes. It has been proposed that platelet-derived growth factor B polypeptide (Pdgfb) secreted by these endothelial cells drives the formation of the surrounding muscular wall by recruiting nearby mesenchymal cells. However, targetted inactivation of the Pdgfb gene or the Pdgf receptor beta (Pdgfrb) gene, by homologous recombination, does not prevent the development of apparently normal large arteries and connective tissue. We have used an in vivo competition assay in which we prepared chimaeric blastocysts, composed of a mixture of wild-type (Pdgfrb[+/+]) and Pdgfrb(+/-) or wild-type and Pdgfrb(-/-) cells, and quantified the relative success of cells of the two component genotypes in competing for representation in different cell lineages as the chimaeric embryos developed. This study revealed that the participation of Pdgfrb(-/-) cells in all muscle lineages (smooth, cardiac, skeletal and pericyte) was reduced by eightfold compared with Pdgfrb(+/+) cells, and that participation of Pdgfrb(+/-) cells was reduced by twofold (eightfold for pericytes). Pdgfrb inactivation did not affect cell contribution to non-muscle mesodermal lineages, including fibroblasts and endothelial cells. Chimaera competition is therefore a sensitive, quantitative method for determining developmental roles of specific genes, even when those roles are not apparent from analysis of purebred mutants; most likely because they are masked by homeostatic mechanisms.
Collapse
Affiliation(s)
- J R Crosby
- Department of Pathology, University of Washington, Seattle 98195-7470, USA
| | | | | | | |
Collapse
|
37
|
Sakata M, Yanamoto H, Hashimoto N, Iihara K, Tsukahara T, Taniguchi T, Kikuchi H. Induction of infarct tolerance by platelet-derived growth factor against reversible focal ischemia. Brain Res 1998; 784:250-5. [PMID: 9518639 DOI: 10.1016/s0006-8993(97)01345-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nerve growth factor, brain-derived neurotrophic factor, and other neurotrophic factors have been reported to have neuroprotective effects against global ischemia. To investigate whether the homodimer of platelet-derived growth factor B-chain (PDGF-BB) can protect neurons against focal temporary ischemia, PDGF-BB was administered to the rat brain for a prolonged period prior to, during, and after ischemia, since PDGF-BB protected rat neurons from global ischemia in our previous study. A total of 82 male Sprague-Dawley rats were used. Recombinant PDGF-BB, or saline was administered into the left neocortex via an implanted osmotic pump for 3 days (1.2 microg in total), 7 days (2 microgram or 4 microgram in total), or 14 days (4 microgram in total) pre-ischemia and 2 days post-ischemia. In an additional group, PDGF-BB (4 microgram in total) was administered for 14 days by osmotic pump and focal ischemia was induced after an additional 7-day interval following removal of the pump. Focal temporary ischemia was induced in the left MCA territory by bilateral CCA and MCA occlusion for 2 h. All rats were sacrificed 2 days after ischemia and the volume of cerebral infarct was analyzed using TTC staining. In a separate set of animals, regional cerebral blood flow (rCBF) was monitored by the hydrogen clearance method and laser Doppler flowmetry (LDF) of the neocortex after 14 days of intracerebral administration of PDGF-BB or saline. In the group receiving PDGF-BB (4 microgram in total) for 7 or 14 days pre-ischemia, there was a significant reduction of neocortical infarction compared to that in the control or saline-infused group. The size of cerebral infarct was smallest in the group that received PDGF-BB for 14 days, when ischemia was induced 7 days after removal of the pump. Regarding rCBF measurement, there were no significant differences in groups receiving PDGF-BB or saline infusion for 14 days. The potent neuroprotective effect of PDGF-BB on global ischemia was also demonstrated in the focal ischemia model. However, prolonged intracerebral infusion for 7 to 14 days was necessary to achieve a significant reduction of infarct volume. Neuroprotection was not due to increased collateral flow during ischemia.
Collapse
Affiliation(s)
- M Sakata
- Laboratory for Cerebrovascular Disorders, National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka, 565, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Woodard AS, García-Cardeña G, Leong M, Madri JA, Sessa WC, Languino LR. The synergistic activity of alphavbeta3 integrin and PDGF receptor increases cell migration. J Cell Sci 1998; 111 ( Pt 4):469-78. [PMID: 9443896 DOI: 10.1242/jcs.111.4.469] [Citation(s) in RCA: 132] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Integrins and growth factor receptors act synergistically to modulate cellular functions. The alphavbeta3 integrin and the platelet-derived growth factor receptor have both been shown to play a positive role in cell migration. We show here that a platelet derived growth factor-BB gradient stimulated migration of rat microvascular endothelial cells on vitronectin (9.2-fold increase compared to resting cells) in a alphavbeta3 and RGD-dependent manner. In contrast, this response was not observed on a beta1 integrin ligand, laminin; background levels of migration, in response to a platelet derived growth factor-BB gradient, were observed on this substrate or on bovine serum albumin (2.4- or 2.0-fold, respectively). Comparable results were obtained using NIH-3T3 cells. Platelet derived growth factor-BB did not change the cells' ability to adhere to vitronectin, nor did it stimulate a further increase in proliferation on vitronectin versus laminin. In addition, platelet derived growth factor-BB stimulation of NIH-3T3 cells did not alter the ability of alphavbeta3 to bind RGD immobilized on Sepharose. The alphavbeta3 integrin and the platelet derived growth factor receptor-beta associate in both microvascular endothelial cells and NIH-3T3 cells, since they coprecipitated using two different antibodies to either alphavbeta3 or to the platelet derived growth factor receptor-beta. In contrast, beta1 integrins did not coprecipitate with the platelet derived growth factor receptor-beta. These results point to a novel pathway, mediated by the synergistic activity of alphavbeta3 and the platelet derived growth factor receptor-beta, that regulates cell migration and, therefore, might play a role during neovessel formation and tissue infiltration.
Collapse
Affiliation(s)
- A S Woodard
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | |
Collapse
|
39
|
Kawabe T, Wen TC, Matsuda S, Ishihara K, Otsuda H, Sakanaka M. Platelet-derived growth factor prevents ischemia-induced neuronal injuries in vivo. Neurosci Res 1997; 29:335-43. [PMID: 9527625 DOI: 10.1016/s0168-0102(97)00105-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Platelet-derived growth factor (PDGF) has been considered to be a neuroprotective factor candidate on the basis of several in vitro studies. However, the in vivo effect of PDGF on ischemic neurons has not been determined. In the present study, the effect of PDGF-BB on the ischemia-induced disability of passive avoidance task and hippocampal CA1 neuron death in normothermic gerbils, whose the brain temperature was kept at 37.0 +/- 0.2 degrees C during 3-min occlusion of the common carotid arteries was investigated. When PDGF-BB was continuously infused for 7 days into the cerebral ventricles of gerbils with transient forebrain ischemia, response latency time in a passive avoidance task was significantly prolonged. Subsequent histological examinations showed that PDGF-BB effectively increased the number of viable pyramidal neurons in the hippocampal CA1 region as well as synapses within the strata moleculare, radiatum and oriens of the region in comparison with the numbers of neurons and synapses in vehicle-treated ischemic gerbils. In situ detection of DNA fragmentation (TUNEL staining) revealed that TUNEL-positive neurons in the hippocampal CA1 field of vehicle-treated ischemic gerbils were much more numerous than those in the field of PDGF-BB-treated ischemic animals after 7 days ischemia. These findings suggest that the present ischemic animal model exhibits a more delayed neuronal degeneration of the hippocampal CA1 field than the conventional 5-min ischemic model and that the 7-day infusion of PDGF-BB, starting 2 h before ischemic insult, not only prevents delayed neuronal death in the hippocampal CA1 field at 7 days after forebrain ischemia but also inhibits a slowly progressive neuronal degeneration occurring thereafter.
Collapse
Affiliation(s)
- T Kawabe
- Department of Anatomy, Ehime University School of Medicine, Shigenobu, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Floege J, Hudkins KL, Seifert RA, Francki A, Bowen-Pope DF, Alpers CE. Localization of PDGF alpha-receptor in the developing and mature human kidney. Kidney Int 1997; 51:1140-50. [PMID: 9083280 DOI: 10.1038/ki.1997.157] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Using in situ hybridization and immunocytochemistry we describe the renal localization of the PDGF alpha-receptor. PDGF alpha-receptor mRNA was uniformly present in human metanephric kidney in interstitial cells and vascular arcades that course through the blastema. PDGF alpha-receptor mRNA was present in some mesangial structures in early glomeruli, but was largely lost as glomeruli matured. It was present in adventitial fibroblasts, but usually not in vascular smooth muscle cells or endothelial cells of the fetal vasculature. This pattern persisted in adult kidneys, with extensive expression of mRNA by interstitial cells and only occasional expression by mesangial cells. All in situ hybridization findings were corroborated by immunocytochemistry. Double immunolabeling confirmed the rare expression of the PDGF alpha-receptor protein by vascular smooth muscle cells and the absence of its expression by endothelial cells. Given that both PDGF A- and B-chain can promote smooth muscle cell and fibroblast migration and proliferation and that both signal through the PDGF alpha-receptor, these data suggest that PDGF alpha-receptor may play important roles in the early vasculogenesis of the fetal kidney as well as in the pathogenesis of renal interstitial fibrosis.
Collapse
MESH Headings
- Adult
- Endothelium, Vascular/cytology
- Endothelium, Vascular/embryology
- Endothelium, Vascular/metabolism
- Fetus/cytology
- Fetus/metabolism
- Glomerular Mesangium/cytology
- Glomerular Mesangium/embryology
- Glomerular Mesangium/metabolism
- Humans
- Immunohistochemistry
- In Situ Hybridization
- Kidney/cytology
- Kidney/embryology
- Kidney/metabolism
- Kidney Diseases/etiology
- Kidney Diseases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Platelet-Derived Growth Factor alpha
- Receptors, Platelet-Derived Growth Factor/genetics
- Receptors, Platelet-Derived Growth Factor/metabolism
Collapse
Affiliation(s)
- J Floege
- Division of Nephrology, Medical School, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Thommen R, Humar R, Misevic G, Pepper MS, Hahn AW, John M, Battegay EJ. PDGF-BB increases endothelial migration and cord movements during angiogenesis in vitro. J Cell Biochem 1997. [DOI: 10.1002/(sici)1097-4644(19970301)64:3<403::aid-jcb7>3.0.co;2-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
42
|
|
43
|
Fruttiger M, Calver AR, Krüger WH, Mudhar HS, Michalovich D, Takakura N, Nishikawa S, Richardson WD. PDGF mediates a neuron-astrocyte interaction in the developing retina. Neuron 1996; 17:1117-31. [PMID: 8982160 DOI: 10.1016/s0896-6273(00)80244-5] [Citation(s) in RCA: 189] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Astrocytes invade the developing retina from the optic nerve head, over the axons of retinal ganglion cells (RGCs). RGCs express the platelet-derived growth factor A-chain (PDGF-A) and retinal astrocytes the PDGF alpha-receptor (PDGFR alpha), suggesting that PDGF mediates a paracrine interaction between these cells. To test this, we inhibited PDGF signaling in the eye with a neutralizing anti-PDGFR alpha antibody or a soluble extracellular fragment of PDGFR alpha. These treatments inhibited development of the astrocyte network. We also generated transgenic mice that overexpress PDGF-A in RGCs. This resulted in hyperproliferation of astrocytes, which in turn induced excessive vasculogenesis. Thus, PDGF appears to be a link in the chain of cell-cell interactions responsible for matching numbers of neurons, astrocytes, and blood vessels during retinal development.
Collapse
Affiliation(s)
- M Fruttiger
- MRC Laboratory for Molecular Cell Biology, University College London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Waltenberger J, Akyürek ML, Aurivillius M, Wanders A, Larsson E, Fellström B, Funa K. Ischemia-induced transplant arteriosclerosis in the rat. Induction of peptide growth factor expression. Arterioscler Thromb Vasc Biol 1996; 16:1516-23. [PMID: 8977457 DOI: 10.1161/01.atv.16.12.1516] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Peptide growth factors have been reported to contribute to the atherogenic process, and they are known to mediate signals for vascular remodeling. Using syngeneic and allogeneic rat aorta transplant models, we analyzed the impact of cold ischemia time up to 24 hours and reperfusion injury on development of transplant arteriosclerosis during the first 2 months after transplantation. The expression of the transforming growth factor-beta (TGF-beta) family as well as the platelet-derived growth factor (PDGF) and its receptors was studied by use of immunohistochemistry, followed by semiquantitative evaluation and multivariate analysis. In the syngeneically transplanted aortas, the expression of TGF-beta 1, PDGF, and the two PDGF receptors in the neointima increased significantly with the extent of cold ischemia time. Furthermore, there was a significant induction of the latent TGF-beta binding protein in the neointima as well as TGF-beta 2 in the media, both correlating with the observation time after transplantation. In the allogeneic grafts, all examined proteins were already induced strongly 2 weeks after transplantation, even at the shortest ischemic period studied (1 hour). However, no positive correlation between growth factor expression and cold ischemia or observation time could be found. Double immunohistochemistry revealed that macrophages express PDGF and its receptors as well as TGF-beta 1. Smooth muscle cells express both types of PDGF receptors, and a few T cells express TGF-beta 1 as well as PDGF receptors. In summary, TGF-beta and PDGF are induced by allogeneic as well as ischemic stimuli in transplanted aortas, suggesting a role in the pathogenesis of transplant arteriosclerosis and representing a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- J Waltenberger
- Department of Internal Medicine II, Ulm University Medical Center, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Kobayashi S, Kimura I, Kimura M. Diabetic state-modified macrophages in GK rat release platelet-derived growth factor-BB for tube formation of endothelial cells in rat aorta. IMMUNOPHARMACOLOGY 1996; 35:171-80. [PMID: 8956981 DOI: 10.1016/s0162-3109(96)00143-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cultured peritoneal macrophages (M phi) of GK rats, a non insulin-dependent diabetes mellitus model established from normal Wistar rats, increased the tube formation of aortic endothelial cells (EC). A polyclonal anti-platelet-derived growth factor (PDGF)-BB (0.21-3.3 micrograms/ml) inhibited (by 66.5 +/- 6.6%) the tube-forming activity of conditioned medium (CM) from M phi in diabetic GK rats, but not that in age-matched normal Wistar rats. A polyclonal anti-interleukin (IL)-1 alpha (0.16-0.33%) also inhibited (by 37.7 +/- 5.8%) the activity of diabetic M phi-CM, and its inhibitory effect was smaller than that of anti-PDGF-BB. A monoclonal anti-basic fibroblast growth factor (bFGF) (0.6-60 ng/ml) inhibited the activities of CM from both M phi, respectively. The tube-forming activity of the normal M phi-CM was increased by the serum isolated from the diabetic GK rat more than by that from the age-matched normal Wistar rat. The activity of normal M phi-CM was also increased by 16.7-50 mM glucose-exposed serum. These tube-forming activities of the CM of M phi stimulated by diabetic serum and by the high concentration of glucose-treated serum were completely inhibited by anti-PDGF-BB. In conclusion, PDGF-BB was selectively released from the diabetic state-modified M phi in the GK rat to increase tube formation, suggesting a key role in the cause of angiogenesis. The diabetic state-induced activation may depend on advanced glycosylation endproducts produced in the serum in the diabetic rat.
Collapse
Affiliation(s)
- S Kobayashi
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, Japan
| | | | | |
Collapse
|
46
|
Funa K, Yamada N, Brodin G, Pietz K, Ahgren A, Wictorin K, Lindvall O, Odin P. Enhanced synthesis of platelet-derived growth factor following injury induced by 6-hydroxydopamine in rat brain. Neuroscience 1996; 74:825-33. [PMID: 8884778 DOI: 10.1016/0306-4522(96)00152-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The kinetics of platelet-derived growth factor messenger RNA synthesis in the substantia nigra and in the striatum, before and after unilateral intranigral 6-hydroxydopamine injection, was studied and compared with that after sham operation by a quantitative reverse transcription-polymerase chain reaction. The kinetics of brain-derived neurotrophic factor messenger RNA was studied as a comparison. Furthermore, the expression of platelet-derived growth factor A- and B-chain proteins was analysed by enzyme-linked immunosorbent assay and immunohistochemistry. In the ipsilateral striatum of 6-hydroxydopamine-lesioned rats, the signal density of messenger RNA for both A- and B-chains had already increased at one day and remained at an elevated level during the observation period of four weeks. In the substantia nigra ipsilateral to the lesion, a strongly increased level of B-chain and, to a lesser extent, of A-chain messenger RNA was already detected at 4h, reaching a maximal level at one day. No significant increase was seen either in sham-operated rats or in the contralateral striatum and substantia nigra. Amounts of platelet-derived growth factor proteins were examined separately by enzyme-linked immunosorbent assay in both sides of the substantia nigra, striatum and cortex. Three days after 6-hydroxydopamine lesions the levels of both platelet-derived growth factor A- and B-chains increased in the ipsilateral striatum, substantia nigra, and cortex. An increase in the A-chain was also observed in the contralateral side of the brain. The signal for brain-derived neurotrophic factor messenger RNA increased in the striatum in the lesioned side and, to a lesser extent, in the contralateral side, as well as in the substantia nigra, where a significant difference was observed when compared with the contralateral side. Semiquantitative immunohistochemical analysis on the substantia nigra confirmed the enhanced platelet-derived growth factor expression, revealing that the majority of the platelet-derived growth factor-producing cells were neurons. In summary, we have shown that platelet-derived growth factor messenger RNA as well as its protein are induced after injury to dopaminergic cells. These data indicate an important role of platelet-derived growth factor in the dopaminergic system.
Collapse
Affiliation(s)
- K Funa
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Horner A, Bord S, Kemp P, Grainger D, Compston JE. Distribution of platelet-derived growth factor (PDGF) A chain mRNA, protein, and PDGF-alpha receptor in rapidly forming human bone. Bone 1996; 19:353-62. [PMID: 8894141 DOI: 10.1016/s8756-3282(96)00217-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Platelet-derived growth factors (PDGFs) are potent bone cell mitogens which stimulate the proliferation of osteoblastic cells, may also be involved in the regulation of osteoclastic bone resorption, and indirectly induce vascular endothelial cell proliferation and angiogenesis. In view of the established relationship between angiogenesis and osteogenesis, the production of PDGFs by both osteoblastic and vascular endothelial cells suggests that they may play a role in bone formation during skeletal development. We have used two human models of rapid bone formation, heterotopic bone and osteophytic bone, to investigate the expression of PDGF-A mRNA and protein and the PDGF-alpha receptor protein in vivo using in situ hybridization and immunohistochemistry. PDGF-A mRNA and protein were widely distributed throughout heterotopic and osteophytic bone. Within the cartilaginous tissue PDGF-A mRNA and protein were most strongly expressed by mature chondrocytes with decreased expression in the hypertrophic zone and almost no staining in the mineralizing and mineralized zones. PDGF mRNA and protein were also expressed in cells of small blood vessels within fibrous and cartilaginous tissue. In contrast, PDGF-alpha receptor expression was restricted to a minority of hypertrophic chondrocytes and sites of vascular invasion. Within the bone and fibrous tissue the growth factor and the receptor were widely distributed, being detected on most cells at sites of bone formation or in remodeling sites; no receptor was detected on osteoclasts. These data demonstrate the widespread expression of PDGF-A and its receptor in forming human bone and indicate that this growth factor may exert autocrine and paracrine effects to regulate osteogenesis during skeletal development.
Collapse
Affiliation(s)
- A Horner
- University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, UK.
| | | | | | | | | |
Collapse
|
48
|
Iihara K, Sasahara M, Hashimoto N, Hazama F. Induction of platelet-derived growth factor beta-receptor in focal ischemia of rat brain. J Cereb Blood Flow Metab 1996; 16:941-9. [PMID: 8784238 DOI: 10.1097/00004647-199609000-00018] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Our previous study on the ischemia-induced expression of platelet-derived growth factor (PDGF)-B chain in the rat brain prompted us to examine expression of PDGF beta-receptor in the ischemic brain. Focal ischemia was induced by permanent tandem occlusion of middle cerebral and common carotid arteries in spontaneously hypertensive rats. Northern analysis revealed that ischemia significantly increased expression of the receptor in the ischemic neocortex at 4 and 7 days (328 +/- 109%; 323 +/- 119%, respectively, over control: n = 4, p < 0.05 versus sham). Neurons in infarct transiently showed increased immunostaining for the receptor at 1 day, whereas neurons in periinfarct area showed sustained and increased immunoreactivity from 1 to 14 days post-ischemia. Reactive glial cells in the external capsule and in molecular layer of the neocortex adjacent to infarct possessed enhanced immunoreactivity from 1 to 21 days. Furthermore, marked immunoreactivity was observed on brain macrophages in infarct and on the abluminal side of capillaries surrounding infarct from 4 to 7 days. These results demonstrated that ischemic insult increases expression of the PDGF beta-receptor at both the mRNA and protein level in the brain, suggesting its important role in cellular cascade of the ischemic brain.
Collapse
Affiliation(s)
- K Iihara
- Department of Cerebrovascular Surgery, National Cardiovascular Center, Osaka, Japan
| | | | | | | |
Collapse
|
49
|
Tanizawa S, Ueda M, van der Loos CM, van der Wal AC, Becker AE. Expression of platelet derived growth factor B chain and beta receptor in human coronary arteries after percutaneous transluminal coronary angioplasty: an immunohistochemical study. HEART (BRITISH CARDIAC SOCIETY) 1996; 75:549-56. [PMID: 8697155 PMCID: PMC484375 DOI: 10.1136/hrt.75.6.549] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To evaluate whether expression of platelet derived growth factor B (PDGF-B) protein is associated with expression of its receptor protein in human coronary arteries after angioplasty and to identify cells involved. BACKGROUND PDGF is considered an important growth factor in the repair process of the vessel wall after angioplasty. In situ hybridisation has revealed expression of PDGF-A and -B chain messenger ribonucleic acid (mRNA) in human coronary arteries at sites of postangioplasty injury. METHODS Target and non-target sites of eight coronary arteries were studied immunohistochemically for PDGF-B and PDGF-beta receptor proteins in relation to macrophages, T lymphocytes, smooth muscle cells, and HLA-DR positive cells. RESULTS The PDGF-B and PDGF-beta receptor proteins were expressed in areas with distinct repair, containing alpha actin negative spindle cells, macrophages and, at later stages, alpha actin positive smooth muscle cells as well. When the neointima was composed mainly of alpha actin smooth muscle cells, PDGF-B expression was rare and PDGF-beta receptor expression was negative. CONCLUSIONS There is expression of PDGF-B and PDGF-beta receptor proteins at sites of postangioplasty repair in human coronary arteries. The associated cells are mainly macrophages and alpha actin negative spindle cells; the latter may be dedifferentiated smooth muscle cells. A link between PDGF expression and the postangioplasty time interval suggests a relation with cell differentiation as part of the maturation of the repair tissue. Mutual expression of both the growth factor and its receptor protein strongly suggests that in humans a PDGF mediated repair process occurs, with involvement of smooth muscle cells and macrophages.
Collapse
Affiliation(s)
- S Tanizawa
- Department of Cardiovascular Pathology, University of Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|
50
|
Wang JL, Nistér M, Bongcam-Rudloff E, Pontén J, Westermark B. Suppression of platelet-derived growth factor alpha- and beta-receptor mRNA levels in human fibroblasts by SV40 T/t antigen. J Cell Physiol 1996; 166:12-21. [PMID: 8557760 DOI: 10.1002/(sici)1097-4652(199601)166:1<12::aid-jcp2>3.0.co;2-j] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
It is known that down-regulation of cell surface platelet-derived growth factor (PDGF) receptors accompanies transformation by SV40. In this work human embryonic lung fibroblasts were used as a model system to study the effects of SV 40 on PDGF receptor expression. It is shown that transformation by SV 40 early region leads to a total loss of PDGF alpha-receptor and partial loss of beta-receptor mRNA. Microinjection experiments revealed that receptor down-regulation was a primary effect, and not only secondary to transformation and clonal selection. Total loss of PDGF alpha-receptor expression requires both large T and small t, and down-regulation of the PDGF alpha-receptor occurs independently of p53 and Rb binding to large T.
Collapse
Affiliation(s)
- J L Wang
- Department of Pathology, University Hospital, Uppsala, Sweden
| | | | | | | | | |
Collapse
|