1
|
Davenport C, Dubin A. Tadalafil therapy and severe chronic foot wound resolution. Int Wound J 2015; 12:733-6. [PMID: 25649683 DOI: 10.1111/iwj.12378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 09/01/2014] [Indexed: 12/20/2022] Open
Abstract
We report an unanticipated medication effect resulting in near-complete wound healing in a patient after beginning tadalafil therapy of 20 mg thrice daily. This patient was treated for 50 years with state-of-the art acute and then chronic wound interventions after a traumatic farm machinery accident which resulted in a devascularised foot wound. This infection was both life- and limb-threatening. The patient had undergone multiple vascular and plastic surgeries and antimicrobial therapies in addition to hospitalisations for sepsis. Limb amputation was being considered, when his urologist placed him on daily phosphodiesterase-5 inhibitor therapy, prior to unrelated urologic surgery. Remarkably, his foot wound underwent near-complete resolution and has been stable for 2 years.
Collapse
Affiliation(s)
- Claire Davenport
- Department of Internal Medicine, Albany Medical College, Albany, NY, USA
| | - Andrew Dubin
- Department of PM&R, Albany Medical College, Albany, NY, USA
| |
Collapse
|
2
|
Baker BA, Pine PS, Chatterjee K, Kumar G, Lin NJ, McDaniel JH, Salit ML, Simon CG. Ontology analysis of global gene expression differences of human bone marrow stromal cells cultured on 3D scaffolds or 2D films. Biomaterials 2014; 35:6716-26. [DOI: 10.1016/j.biomaterials.2014.04.075] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 04/22/2014] [Indexed: 01/07/2023]
|
3
|
Guerreiro SG, Oliveira MJ, Barbosa MA, Soares R, Granja PL. Neonatal Human Dermal Fibroblasts Immobilized in RGD–Alginate Induce Angiogenesis. Cell Transplant 2014; 23:945-57. [PMID: 23866308 DOI: 10.3727/096368913x670183] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Promoting angiogenesis in a damaged tissue is a major challenge for tissue regeneration. Recent findings in tissue engineering suggest that fibroblasts (FBs) play an important role in orchestrating the angiogenic process. Fibroblasts maintain the structural integrity of connective tissue by continuously secreting growth factors and extracellular matrix precursors, which are essential for endothelial cell (EC) adhesion and spreading, thus playing a crucial role in angiogenesis. We hypothesized that FBs immobilized in alginate gels grafted with the RGD peptidic sequence could influence the recruitment of ECs to improve vascularization. In this work, the modulation of immobilized human FBs within the 3D synthetic extracellular matrix was assessed. Experiments using cocultures of ECs and FBs in indirect contact as well as angiogenic assays were performed to assess the influence of FBs immobilized in RGD–alginate in ECs' viability, stabilization, sprouting, and assembly into capillary-like structures. This study demonstrates the ability of FBs immobilized within RGD–alginate microspheres to modulate and support capillary-like structures' assembly. These findings indicate that the microenvironment created by these stromal cells in the scaffold modulates capillary morphogenesis, thus stimulating angiogenesis in situ and can potentially be used in regenerative medicine in clinical scenarios where vascularization is essential.
Collapse
Affiliation(s)
- Susana G. Guerreiro
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Medicina (FMUP), Departamento de Bioquímica, Porto, Portugal
- Universidade do Porto, Faculdade de Engenharia (FEUP), Porto, Portugal
| | - Maria J. Oliveira
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Medicina (FMUP), Departamento de Anatomia Patológica, Porto, Portugal
| | - Mário A. Barbosa
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Engenharia (FEUP), Porto, Portugal
- Universidade do Porto, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Porto, Portugal
| | - Raquel Soares
- Universidade do Porto, Faculdade de Medicina (FMUP), Departamento de Bioquímica, Porto, Portugal
| | - Pedro L. Granja
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Universidade do Porto, Faculdade de Engenharia (FEUP), Porto, Portugal
| |
Collapse
|
4
|
Liu X, Claus P, Wu M, Reyns G, Verhamme P, Pokreisz P, Vandenwijngaert S, Dubois C, Vanhaecke J, Verbeken E, Bogaert J, Janssens S. Placental growth factor increases regional myocardial blood flow and contractile function in chronic myocardial ischemia. Am J Physiol Heart Circ Physiol 2013; 304:H885-94. [PMID: 23316060 DOI: 10.1152/ajpheart.00587.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Placental growth factor (PlGF) has a distinct biological phenotype with a predominant proangiogenic role in disease without affecting quiescent vessels in healthy organs. We tested whether systemic administration of recombinant human (rh)PlGF improves regional myocardial blood flow (MBF) and systolic function recovery in a porcine chronic myocardial ischemia model. We implanted a flow-limiting stent in the proximal left anterior descending coronary artery and measured systemic hemodynamics, regional myocardial function using MRI, and blood flow using colored microspheres 4 wk later. Animals were then randomized in a blinded way to receive an infusion of rhPlGF (15 μg·kg(-1)·day(-1), n = 9) or PBS (control; n = 10) for 2 wk. At 8 wk, myocardial perfusion and function were reassessed. Infusion of rhPlGF transiently increased PlGF serum levels >30-fold (1,153 ± 180 vs. 33 ± 18 pg/ml at baseline, P < 0.001) without affecting systemic hemodynamics. From 4 to 8 wk, rhPlGF increased regional MBF from 0.46 ± 0.11 to 0.85 ± 0.16 ml·min(-1)·g(-1), with a concomitant increase in systolic wall thickening from 11 ± 3% to 26 ± 5% in the ischemic area. In control animals, no significant changes from 4 to 8 wk were observed (MBF: 0.45 ± 0.07 to 0.49 ± 0.08 ml·min(-1)·g(-1) and systolic wall thickening: 14 ± 4% to 18 ± 1%). rhPlGF-induced functional improvement was accompanied by increased myocardial neovascularization, enhanced glycogen utilization, and reduced oxidative stress and cardiomyocyte apoptosis in the ischemic zone. In conclusion, systemic rhPlGF infusion significantly enhances regional blood flow and contractile function of the chronic ischemic myocardium without adverse effects. PlGF protein infusion may represent an attractive therapeutic strategy to increase myocardial perfusion and energetics in chronic ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Xiaoshun Liu
- Division of Clinical Cardiology and Department of Cardiovascular Sciences, Gasthuisberg University Hospitals, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Maina JN. Comparative molecular developmental aspects of the mammalian- and the avian lungs, and the insectan tracheal system by branching morphogenesis: recent advances and future directions. Front Zool 2012; 9:16. [PMID: 22871018 PMCID: PMC3502106 DOI: 10.1186/1742-9994-9-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 06/18/2012] [Indexed: 02/07/2023] Open
Abstract
Gas exchangers fundamentally form by branching morphogenesis (BM), a mechanistically profoundly complex process which derives from coherent expression and regulation of multiple genes that direct cell-to-cell interactions, differentiation, and movements by signaling of various molecular morphogenetic cues at specific times and particular places in the developing organ. Coordinated expression of growth-instructing factors determines sizes and sites where bifurcation occurs, by how much a part elongates before it divides, and the angle at which branching occurs. BM is essentially induced by dualities of factors where through feedback- or feed forward loops agonists/antagonists are activated or repressed. The intricate transactions between the development orchestrating molecular factors determine the ultimate phenotype. From the primeval time when the transformation of unicellular organisms to multicellular ones occurred by systematic accretion of cells, BM has been perpetually conserved. Canonical signalling, transcriptional pathways, and other instructive molecular factors are commonly employed within and across species, tissues, and stages of development. While much still remain to be elucidated and some of what has been reported corroborated and reconciled with rest of existing data, notable progress has in recent times been made in understanding the mechanism of BM. By identifying and characterizing the morphogenetic drivers, and markers and their regulatory dynamics, the elemental underpinnings of BM have been more precisely explained. Broadening these insights will allow more effective diagnostic and therapeutic interventions of developmental abnormalities and pathologies in pre- and postnatal lungs. Conservation of the molecular factors which are involved in the development of the lung (and other branched organs) is a classic example of nature's astuteness in economically utilizing finite resources. Once purposefully formed, well-tested and tried ways and means are adopted, preserved, and widely used to engineer the most optimal phenotypes. The material and time costs of developing utterly new instruments and routines with every drastic biological change (e.g. adaptation and speciation) are circumvented. This should assure the best possible structures and therefore functions, ensuring survival and evolutionary success.
Collapse
Affiliation(s)
- John N Maina
- Department of Zoology, University of Johannesburg, Auckland Park 2006, P,O, Box 524, Johannesburg, South Africa.
| |
Collapse
|
6
|
Blakely C, Jahan T. Emerging antiangiogenic therapies for non-small-cell lung cancer. Expert Rev Anticancer Ther 2012; 11:1607-18. [PMID: 21999134 DOI: 10.1586/era.11.146] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lung cancer remains the leading cause of cancer-related deaths. Antiangiogenic therapy has increasingly been studied for advanced non-small-cell lung cancer (NSCLC). Bevacizumab is the only approved antiangiogenic agent for NSCLC and has shown progression-free survival benefits in large Phase III studies and an overall survival benefit in the Phase III E4599 trial in advanced nonsquamous NSCLC. New antiangiogenic treatment strategies are being evaluated that target multiple receptors within a family (VEGF receptor [VEGFR]-1, VEGFR-2) or multiple angiogenic pathways (targets VEGFR and PDGF receptor pathways), and agents that inhibit alternative mediators of angiogenesis (integrins and established vasculature). As data become available from ongoing studies, it will be important to determine how these new antiangiogenic agents will best fit into the current NSCLC treatment paradigm.
Collapse
Affiliation(s)
- Collin Blakely
- University of California, San Francisco, Box 1705, San Francisco, CA 94143-1705, USA
| | | |
Collapse
|
7
|
Ma Q, Wang W, Chu PK, Mei S, Ji K, Jin L, Zhang Y. Concentration- and time-dependent response of human gingival fibroblasts to fibroblast growth factor 2 immobilized on titanium dental implants. Int J Nanomedicine 2012; 7:1965-76. [PMID: 22619534 PMCID: PMC3356224 DOI: 10.2147/ijn.s29538] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Titanium (Ti) implants are widely used clinically, but peri-implantitis remains one of the most common and serious complications. Healthy integration between gingival tissue and the implant surface is critical to long-term success in dental implant therapy. The objective of this study was to investigate how different concentrations of immobilized fibroblast growth factor 2 (FGF2) on the titania nanotubular surface influence the response of human gingival fibroblasts (HGFs). METHODS Pure Ti metal was anodized at 20 V to form a vertically organized titanium dioxide nanotube array on which three concentrations of FGF2 (250 ng/mL, 500 ng/mL, or 1000 ng/mL) were immobilized by repeated lyophilization. Surface topography was observed and FGF2 elution was detected using enzyme-linked immunosorbent assay. The bioactivity changes of dissolvable immobilized FGF2 were measured by methyl-thiazolyl-tetrazolium assay. Behavior of HGFs was evaluated using adhesion and methyl-thiazolyl-tetrazolium bromide assays. RESULTS The FGF2 remained for several days on the modified surface on which HGFs were cultured. Over 90% of the dissolvable immobilized FGF2 had been eluted by Day 9, whereas the FGF2 activity was found to diminish gradually from Day 1 to Day 9. The titania nanotubular surface with an optimal preparing concentration (500 ng/mL) of FGF2 immobilization exhibited improved HGF functions such as cellular attachment, proliferation, and extracellular matrix-related gene expression. Moreover, significant bidirectional as well as concentration- and time-dependent bioactivity was observed. CONCLUSION Synergism of the FGF2-impregnated titanium dioxide nanotubular surface revealed good gingival-implant integration, indicating that these materials might have promising applications in dentistry and other biomedical devices.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Prosthetic Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
8
|
Semrad TJ, Mack PC. Fibroblast growth factor signaling in non-small-cell lung cancer. Clin Lung Cancer 2011; 13:90-5. [PMID: 21959109 DOI: 10.1016/j.cllc.2011.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 07/27/2011] [Accepted: 08/01/2011] [Indexed: 11/25/2022]
Abstract
Despite recent progress in the treatment on non-small cell lung cancer (NSCLC), outcomes remain suboptimal. Treatment advances that target the epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) signaling pathways highlight the need to understand the multiple convergent growth factor signaling pathways involved in the pathogenesis of NSCLC. Signaling through fibroblast growth factors (FGF), long recognized for its pro-angiogenic activity, has recently emerged as a contributing factor in the pathogenesis and progression of NSCLC through an autocrine signaling loop. In addition, this pathway may function as a mechanism of resistance to anti-EGFR and anti-VEGF treatment. Clinical experience with FGF receptor (FGFR) inhibitors is mounting, and more specific inhibitors of this signaling pathway are in development. This review describes the structure of the FGF signaling pathway, delineates its dual roles in angiogenesis and proliferation in NSCLC, evaluates FGF ligand and receptor expression as prognostic biomarkers in NSCLC, and discusses the development of FGF pathway inhibitors for the treatment of lung malignancies.
Collapse
Affiliation(s)
- Thomas J Semrad
- Division of Hematology/Oncology, Department of Internal Medicine, University of California, Davis, Sacramento, CA 95817, USA.
| | | |
Collapse
|
9
|
Effect of native and NH3 plasma-functionalized polymeric membranes on the gene expression profiles of primary hepatocytes. J Tissue Eng Regen Med 2011; 6:486-96. [DOI: 10.1002/term.453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 05/23/2011] [Indexed: 11/07/2022]
|
10
|
Ma Q, Mei S, Ji K, Zhang Y, Chu PK. Immobilization of Ag nanoparticles/FGF-2 on a modified titanium implant surface and improved human gingival fibroblasts behavior. J Biomed Mater Res A 2011; 98:274-86. [DOI: 10.1002/jbm.a.33111] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 02/27/2011] [Accepted: 03/10/2011] [Indexed: 11/12/2022]
|
11
|
Moldenhauer A, Futschik M, Lu H, Helmig M, Götze P, Bal G, Zenke M, Han W, Salama A. Interleukin 32 promotes hematopoietic progenitor expansion and attenuates bone marrow cytotoxicity. Eur J Immunol 2011; 41:1774-86. [DOI: 10.1002/eji.201040986] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 01/23/2011] [Accepted: 03/15/2011] [Indexed: 11/07/2022]
|
12
|
Abstract
Angiogenesis is the formation of new blood vessels from pre-existing vasculature. Pathologic angiogenesis in the eye can lead to severe visual impairment. In our review, we discuss the roles of both pro-angiogenic and anti-angiogenic molecular players in corneal angiogenesis, proliferative diabetic retinopathy, exudative macular degeneration and retinopathy of prematurity, highlighting novel targets that have emerged over the past decade.
Collapse
Affiliation(s)
- Yureeda Qazi
- Department of Ophthalmology, John Moran Eye Center, University of Utah, Salt Lake City, UT-84132, USA
| | | | | |
Collapse
|
13
|
Moberg C, Catalano RD, Charnock-Jones DS, Olovsson M. VEGF-A and Serum Withdrawal Induced Changes in the Transcript Profile in Human Endometrial Endothelial Cells. Reprod Sci 2010; 17:590-611. [DOI: 10.1177/1933719110364550] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Christian Moberg
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden,
| | - Rob D. Catalano
- Department of Pathology, University of Cambridge and the National Institute for Health Research, Cambridge Biomedical Centre, UK
| | - D. Stephen Charnock-Jones
- Department of Obstetrics and Gynaecology, University of Cambridge and the National Institute for Health Research, Cambridge Biomedical Centre, UK
| | - Matts Olovsson
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Butler JM, Kobayashi H, Rafii S. Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat Rev Cancer 2010; 10:138-46. [PMID: 20094048 PMCID: PMC2944775 DOI: 10.1038/nrc2791] [Citation(s) in RCA: 440] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The precise mechanisms whereby anti-angiogenesis therapy blocks tumour growth or causes vascular toxicity are unknown. We propose that endothelial cells establish a vascular niche that promotes tumour growth and tissue repair not only by delivering nutrients and O2 but also through an 'angiocrine' mechanism by producing stem and progenitor cell-active trophogens. Identification of endothelial-derived instructive angiocrine factors will allow direct tumour targeting, while diminishing the unwanted side effects associated with the use of anti-angiogenic agents.
Collapse
Affiliation(s)
- Jason M Butler
- Hideki Kobayashi and Shahin Rafii are at the Howard Hughes Medical Institute, Ansary Stem Cell Institute, Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | |
Collapse
|
15
|
Grimm D, Infanger M, Westphal K, Ulbrich C, Pietsch J, Kossmehl P, Vadrucci S, Baatout S, Flick B, Paul M, Bauer J. A delayed type of three-dimensional growth of human endothelial cells under simulated weightlessness. Tissue Eng Part A 2009; 15:2267-75. [PMID: 19226201 DOI: 10.1089/ten.tea.2008.0576] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Endothelial cells (ECs) form three-dimensional (3D) aggregates without any scaffold when they are exposed to microgravity simulated by a random positioning machine (RPM) but not under static conditions at gravity. Here we describe a delayed type of formation of 3D structures of ECs that was initiated when ECs cultured on a desktop RPM remained adherent for the first 5 days but spread over neighboring adherent cells, forming little colonies. After 2 weeks, tube-like structures (TSs) became visible in these cultures. They included a lumen, and they elongated during another 2 weeks of culturing. The walls of these TSs consisted mainly of single-layered ECs, which had produced significantly more beta(1)-integrin, laminin, fibronectin, and alpha-tubulin than ECs simultaneously grown adhering to the culture dishes under microgravity or normal gravity. The amount of actin protein was similar in ECs incorporated in TSs and in ECs growing at gravity. The ratio of tissue inhibitor of metalloproteinases-1 to matrix metalloproteinase-2 found in the supernatants was lower at the seventh than at the 28th day of culturing. These results suggest that culturing ECs under conditions of modeled gravitational unloading represents a new technique for studying the formation of tubes that resemble vascular intimas.
Collapse
Affiliation(s)
- Daniela Grimm
- Institute of Clinical Pharmacology and Toxicology, CBF/CCM, Charité-Universitätsmedizin , Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Rusnati M, Bugatti A, Mitola S, Leali D, Bergese P, Depero LE, Presta M. Exploiting Surface Plasmon Resonance (SPR) Technology for the Identification of Fibroblast Growth Factor-2 (FGF2) Antagonists Endowed with Antiangiogenic Activity. SENSORS (BASEL, SWITZERLAND) 2009; 9:6471-503. [PMID: 22454596 PMCID: PMC3312455 DOI: 10.3390/s90806471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 08/17/2009] [Accepted: 08/19/2009] [Indexed: 12/31/2022]
Abstract
Angiogenesis, the process of new blood vessel formation, is implicated in various physiological/pathological conditions, including embryonic development, inflammation and tumor growth. Fibroblast growth factor-2 (FGF2) is a heparin-binding angiogenic growth factor involved in various physiopathological processes, including tumor neovascularization. Accordingly, FGF2 is considered a target for antiangiogenic therapies. Thus, numerous natural/synthetic compounds have been tested for their capacity to bind and sequester FGF2 in the extracellular environment preventing its interaction with cellular receptors. We have exploited surface plasmon resonance (SPR) technique in search for antiangiogenic FGF2 binders/antagonists. In this review we will summarize our experience in SPR-based angiogenesis research, with the aim to validate SPR as a first line screening for the identification of antiangiogenic compounds.
Collapse
Affiliation(s)
- Marco Rusnati
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Antonella Bugatti
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Stefania Mitola
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Daria Leali
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Paolo Bergese
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Laura E. Depero
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| |
Collapse
|
17
|
Crosstalk between osteoblasts and endothelial cells co-cultured on a polycaprolactone-starch scaffold and the in vitro development of vascularization. Biomaterials 2009; 30:4407-15. [PMID: 19487022 DOI: 10.1016/j.biomaterials.2009.05.004] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 05/05/2009] [Indexed: 11/23/2022]
Abstract
The reconstruction of bone defects based on cell-seeded constructs requires a functional microvasculature that meets the metabolic demands of the engineered tissue. Therefore, strategies that augment neovascularization need to be identified. We propose an in vitro strategy consisting of the simultaneous culture of osteoblasts and endothelial cells on a starch-based scaffold for the formation of pre-vascular structures, with the final aim of accelerating the establishment of a vascular bed in the implanted construct. Human dermal microvascular endothelial cells (HDMECs) were co-cultured with human osteoblasts (hOBs) on a 3D starch-based scaffold and after 21 days of culture HDMEC aligned and organized into microcapillary-like structures. These vascular-like structures evolved from a cord-like configuration to a more complex branched morphology, had a lumen and stained in the perivascular region for type IV collagen. Genetic profiling of 84 osteogenesis-related genes was performed on co-culture vs. monoculture. Osteoblasts in co-culture showed a significant up-regulation of type I collagen and immunohistochemistry revealed that the scaffold was filled with a dense matrix stained for type I collagen. In direct contact with HDMEC hOBs secreted higher amounts of VEGF in relation to monoculture and the highest peak in the release profile correlated with the formation of microcapillary-like structures. The heterotypic communication between the two cell types was also assured by direct cell-cell contact as shown by the expression of the gap junction connexin 43. In summary, by making use of heterotypic cellular crosstalk this co-culture system is a strategy to form vascular-like structures in vitro on a 3D scaffold.
Collapse
|
18
|
Mellberg S, Dimberg A, Bahram F, Hayashi M, Rennel E, Ameur A, Westholm JO, Larsson E, Lindahl P, Cross MJ, Claesson-Welsh L. Transcriptional profiling reveals a critical role for tyrosine phosphatase VE-PTP in regulation of VEGFR2 activity and endothelial cell morphogenesis. FASEB J 2009; 23:1490-502. [PMID: 19136612 DOI: 10.1096/fj.08-123810] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To define molecular events accompanying formation of the 3-dimensional (3D) vascular tube, we have characterized gene expression during vascular endothelial growth factor (VEGF)-induced tubular morphogenesis of endothelial cells. Microarray analyses were performed comparing gene induction in growth-arrested, tube-forming endothelial cells harvested from 3D collagen cultures to that in proliferating endothelial cells cultured on fibronectin. Differentially expressed genes were clustered and analyzed for specific endothelial expression through publicly available datasets. We validated the contribution of one of the identified genes, vascular endothelial protein tyrosine phosphatase (VE-PTP), to endothelial morphogenesis. Silencing of VE-PTP expression was accompanied by increased VEGF receptor-2 (VEGFR2) tyrosine phosphorylation and activation of downstream signaling pathways. The increased VEGFR2 activity promoted endothelial cell cycle progression, overcoming the G(0)/G(1) arrest associated with organization into tubular structures in the 3D cultures. Proximity ligation showed close association between VEGFR2 and VE-PTP in resting cells. Activation of VEGFR2 by VEGF led to rapid loss of association, which was resumed with time in parallel with decreased receptor activity. In conclusion, we have identified genes, which may serve critical functions in formation of the vascular tube. One of these, VE-PTP, regulates VEGFR2 activity thereby modulating the VEGF-response during angiogenesis.
Collapse
Affiliation(s)
- Sofie Mellberg
- Department of Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, S-751 85 Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Su SC, Mendoza EA, Kwak HI, Bayless KJ. Molecular profile of endothelial invasion of three-dimensional collagen matrices: insights into angiogenic sprout induction in wound healing. Am J Physiol Cell Physiol 2008; 295:C1215-29. [DOI: 10.1152/ajpcell.00336.2008] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sprouting angiogenesis is a multistep process consisting of basement membrane degradation, endothelial cell (EC) activation, proliferation, invasion, lumen formation, and sprout stabilization. Such complexity is consistent with a requirement for orchestration of individual gene expression alongside multiple signaling pathways. To better understand the mechanisms that direct the transformation of adherent ECs on the surface of collagen matrices to develop multicellular invading sprouts, we analyzed differential gene expression with time using a defined in vitro model of EC invasion driven by the combination of sphingosine-1-phosphate, basic FGF, and VEGF. Gene expression changes were confirmed by real-time PCR and Western blot analyses. A cohort of cell adhesion molecule genes involved in adherens junction and cell-extracellular matrix (ECM) interactions were upregulated, whereas a set of genes associated with tight junctions were downregulated. Numerous genes encoding ECM proteins and proteases were induced, indicating that biosynthesis and remodeling of ECM is indispensable for sprouting angiogenesis. Knockdown of a highly upregulated gene, a disintegrin and metalloproteinase with thrombospondin-type repeats-1 (ADAMTS1), decreased invasion responses, confirming a role for ADAMTS1 in mediating EC invasion. Furthermore, differential expression of multiple members of the Wnt and Notch pathways was observed. Functional experiments indicated that inhibition and activation of the Notch signaling pathway stimulated and inhibited EC invasion responses, respectively. This study has enhanced the molecular road map of gene expression changes that occur during endothelial invasion and highlighted the utility of three-dimensional models to study EC morphogenesis.
Collapse
|
21
|
Klapperich C, Kaufman J, Wong J. Controlling and Assessing Cell–Biomaterial Interactions at the Micro- and Nanoscale. Biomaterials 2007. [DOI: 10.1201/9780849378898.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
22
|
Rennel E, Mellberg S, Dimberg A, Petersson L, Botling J, Ameur A, Westholm JO, Komorowski J, Lassalle P, Cross MJ, Gerwins P. Endocan is a VEGF-A and PI3K regulated gene with increased expression in human renal cancer. Exp Cell Res 2007; 313:1285-94. [PMID: 17362927 DOI: 10.1016/j.yexcr.2007.01.021] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2006] [Revised: 01/06/2007] [Accepted: 01/10/2007] [Indexed: 12/16/2022]
Abstract
An in vitro model of VEGF-A-induced angiogenesis was used to generate transcription profiles of human microvascular endothelial cells. Microarray analysis showed increased transcription of genes known to regulate angiogenesis, but also genes that previously have not been firmly associated with angiogenesis such as endocan, pinin, plakophilin, phosphodiesterase 4B and gelsolin. Increased endocan mRNA levels in response to VEGF-A in endothelial cells and in human renal cancer have previously been reported. We now show increased endocan protein levels in VEGF-A treated endothelial cells and in human renal clear cell carcinoma. Increased protein expression was observed both in tumor cells and in a subset of tumor vessels, while expression in normal kidney tissue was low. VEGF-A seemed to be a specific inducer of endocan transcription since FGF-2, PDGF-BB, HGF/SF and EGF did not alter expression levels. Inhibition of PI3K with LY294002 caused a 12-fold increase in endocan transcription suggesting a repressive function of PI3K. In contrast inhibition of Src or MEK, which are signaling pathways activated by VEGF-A, did not influence basal or VEGF-A-induced endocan levels. In conclusion our study shows that, among angiogenic growth factors, VEGF-A is a specific inducer of endocan transcription which is translated into increased protein levels in VEGF-A treated endothelial cells. Increased endocan protein expression in human renal cancer suggests a role in tumor growth.
Collapse
Affiliation(s)
- Emma Rennel
- Department of Genetics and Pathology, the Rudbeck Laboratory, Uppsala University, S-751 85 Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Anghelina M, Moldovan L, Zabuawala T, Ostrowski MC, Moldovan NL. A subpopulation of peritoneal macrophages form capillarylike lumens and branching patterns in vitro. J Cell Mol Med 2007; 10:708-15. [PMID: 16989730 PMCID: PMC3933152 DOI: 10.1111/j.1582-4934.2006.tb00430.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective: We have previously shown that monocytes/macrophages (MC/Mph) influence neovascularization by extracellular matrix degradation, and by direct incorporation into growing microvessels. To date, neither the phenotype of these cells, nor the stages of their capillary-like conversion were sufficiently characterized. Methods: We isolated mouse peritoneal Mph from transgenic mice expressing fluorescent proteins either ubiquitously, or specifically in the myelocytic lineage. These Mph were embedded in Matrigel which contained fluorescent protease substrates, exposed to an MCP-1 chemotactic gradient, and then examined by confocal microscopy after various intervals. Results: Within 3 hrs after gel embedding, we detected TIMP-1 and MMP-12 dependent proteolysis of the matrix surrounding Mph, mostly in the direction of high concentrations of MCP-1. After 2 days, Mph developed intracellular vacuoles containing degradation product. At 5 days these vacuoles were enlarged and/or fused to generate trans-cellular lumens in approximately 10% of cells or more (depending on animal’s genetic background). At this stage, Mph became tubular, and occasionally organized in three-dimensional structures resembling branched microvessels. Conclusion: Isolated mouse peritoneal Mph penetrate Matrigel and form tunnels via a metalloprotease-driven proteolysis and phagocytosis. Following a morphological adjustment driven by occurrence, enlargement and/or fusion process of intracellular vacuoles, similar to that described in bona fide endothelium, a subpopulation of these cells end up by lining a capillary-like lumen in vitro. Thus we show that adult Mph, not only the more primitive ‘endothelial progenitors’, have functional properties until now considered defining of the endothelial phenotype.
Collapse
Affiliation(s)
- Mirela Anghelina
- Department of Internal Medicine/Division of Cardiology, The Ohio State UniversityColumbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, OH, USA
| | - Leni Moldovan
- Department of Internal Medicine/Division of Cardiology, The Ohio State UniversityColumbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, OH, USA
| | - Tahera Zabuawala
- Department of Molecular Genetics and Molecular, Cellular and Developmental Biology Program, and Comprehensive Cancer, The Ohio State UniversityColumbus, OH, USA
| | - M C Ostrowski
- Department of Molecular Genetics and Molecular, Cellular and Developmental Biology Program, and Comprehensive Cancer, The Ohio State UniversityColumbus, OH, USA
| | - N L Moldovan
- Department of Internal Medicine/Division of Cardiology, The Ohio State UniversityColumbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State UniversityColumbus, OH, USA
- Biomedical Engineering Department, The Ohio State UniversityColumbus, OH, USA
- * Correspondence to: Nicanor I. MOLDOVAN, Ph. D. Departments of Internal Medicine/Cardiology and Biomedical Engineering, Davis Heart and Lung Research Institute, The Ohio State University, 473 W. 12th Ave, Columbus, OH, 43210, USA. Tel.: ++614-247-7801 Fax: ++614-293-5614 E-mail:
| |
Collapse
|
24
|
Glesne DA, Zhang W, Mandava S, Ursos L, Buell ME, Makowski L, Rodi DJ. Subtractive transcriptomics: establishing polarity drives in vitro human endothelial morphogenesis. Cancer Res 2006; 66:4030-40. [PMID: 16618722 DOI: 10.1158/0008-5472.can-05-3294] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although investigations of mature normal and tumor-derived capillaries have resulted in characterization of these structures at the phenotypic level, less is known regarding the initial molecular cues for cellular assembly of endothelial cells into human capillaries. Here, we employ a novel combination of microenvironmental manipulation and microarray data filtration over narrowly delineated temporal data series to identify the morphogenesis component apart from the proliferation component, as pooled human microvascular-derived endothelial cells are induced to form capillary-like structures in vitro in a murine tumor-derived matrix. The 217 morphogenesis-specific genes identified using this subtractive transcriptomics approach are mostly independent of the angiogenic proteins currently used as therapeutic targets for aberrant angiogenesis. Quantitative real-time PCR was used to validate 20% of these transcripts. Immunofluorescent analysis of proliferating and tube-forming cells validates at the protein level the morphogenesis-specific expression pattern of 16 of the 217 gene products identified. The transcripts that are selectively up-regulated in tube-forming endothelial cells reveal a temporal expression pattern of genes primarily associated with intracellular trafficking, guided migration, cytoskeletal reorganization, cellular adhesion, and proliferation inhibition. These data show that a sequential up-regulation of genes that establish and maintain polarity occurs during migration and morphogenesis of in vitro human endothelial cells undergoing tubulogenesis; some of which may well be effective as novel antiangiogenic drug targets.
Collapse
Affiliation(s)
- David A Glesne
- Biosciences Division, Argonne National Laboratory, Argonne, Illinois 60439, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Chute JP, Muramoto GG, Dressman HK, Wolfe G, Chao NJ, Lin S. Molecular Profile and Partial Functional Analysis of Novel Endothelial Cell-Derived Growth Factors that Regulate Hematopoiesis. Stem Cells 2006; 24:1315-27. [PMID: 16373696 DOI: 10.1634/stemcells.2005-0029] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent progress has been made in the identification of the osteoblastic cellular niche for hematopoietic stem cells (HSCs) within the bone marrow (BM). Attempts to identify the soluble factors that regulate HSC self-renewal have been less successful. We have demonstrated that primary human brain endothelial cells (HUBECs) support the ex vivo amplification of primitive human BM and cord blood cells capable of repopulating non-obese diabetic/severe combined immunodeficient repopulating (SCID) mice (SCID repopulating cells [SRCs]). In this study, we sought to characterize the soluble hematopoietic activity produced by HUBECs and to identify the growth factors secreted by HUBECs that contribute to this HSC-supportive effect. Extended noncontact HUBEC cultures supported an eight-fold increase in SRCs when combined with thrombopoietin, stem cell factor, and Flt-3 ligand compared with input CD34(+) cells or cytokines alone. Gene expression analysis of HUBEC biological replicates identified 65 differentially expressed, nonredundant transcripts without annotated hematopoietic activity. Gene ontology studies of the HUBEC transcriptome revealed a high concentration of genes encoding extracellular proteins with cell-cell signaling function. Functional analyses demonstrated that adrenomedullin, a vasodilatory hormone, synergized with stem cell factor and Flt-3 ligand to induce the proliferation of primitive human CD34(+)CD38(-)lin(-) cells and promoted the expansion of CD34(+) progenitors in culture. These data demonstrate the potential of primary HUBECs as a reservoir for the discovery of novel secreted proteins that regulate human hematopoiesis.
Collapse
Affiliation(s)
- John P Chute
- Division of Cellular Therapy, Department of Internal Medicine, Duke University, Durham, North Carolina 27710, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Anghelina M, Krishnan P, Moldovan L, Moldovan NI. Monocytes/macrophages cooperate with progenitor cells during neovascularization and tissue repair: conversion of cell columns into fibrovascular bundles. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:529-41. [PMID: 16436667 PMCID: PMC1606496 DOI: 10.2353/ajpath.2006.050255] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The potential of monocytes/macrophages (MC/Mph) to contribute to neovascularization has recently become a topic of intense scrutiny. Here, we characterized the behavior of MC/Mph in cellular infiltrates, with emphasis on their spatial organization and localization in newly formed microvessels. To this end, we studied MC/Mph migration and assembly in basic fibroblast growth factor-supplemented Matrigel plugs placed in transgenic Tie2-beta-galactosidase mice for up to 4 weeks. In these plugs, along with Nile Red-positive adipocytes, we found MC/Mph distributed in cell cords, also containing various mature and progenitor tissue cells; and functional Tie2-positive or -negative microvessels embedded in bundles of fibrillar collagen surrounded by F4/80-positive MC/Mph. At earlier stages of infiltration, we found tubular destruction of the matrix (tunnels) and MC/Mph-lined capillary-like structures occasionally containing erythrocytes, indicating their propensity for endothelial trans-differentiation. We also analyzed in vitro the MCP-1-induced chemotactic migration of fluorescently labeled peritoneal MC/Mph incorporated in Matrigel-containing fluorescent protease substrates. Many of these MC/Mph produced MMP-12- and TIMP-1-dependent tunnels coupled with acquisition of a lumen. In conclusion, long-term implantation of Matrigel plugs qualifies as a novel experimental model of tissue regeneration, in which neovascularization intimately couples with fibrosis and organogenesis and in which cells of MC/Mph phenotype play a key structural role.
Collapse
Affiliation(s)
- Mirela Anghelina
- Department of Internal Medicine/Cardiology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
27
|
Davis GE, Senger DR. Endothelial extracellular matrix: biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ Res 2005; 97:1093-107. [PMID: 16306453 DOI: 10.1161/01.res.0000191547.64391.e3] [Citation(s) in RCA: 893] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The extracellular matrix (ECM) is critical for all aspects of vascular biology. In concert with supporting cells, endothelial cells (ECs) assemble a laminin-rich basement membrane matrix that provides structural and organizational stability. During the onset of angiogenesis, this basement membrane matrix is degraded by proteinases, among which membrane-type matrix metalloproteinases (MT-MMPs) are particularly significant. As angiogenesis proceeds, ECM serves essential functions in supporting key signaling events involved in regulating EC migration, invasion, proliferation, and survival. Moreover, the provisional ECM serves as a pliable scaffold wherein mechanical guidance forces are established among distal ECs, thereby providing organizational cues in the absence of cell-cell contact. Finally, through specific integrin-dependent signal transduction pathways, ECM controls the EC cytoskeleton to orchestrate the complex process of vascular morphogenesis by which proliferating ECs organize into multicellular tubes with functional lumens. Thus, the composition of ECM and therefore the regulation of ECM degradation and remodeling serves pivotally in the control of lumen and tube formation and, finally, neovessel stability and maturation.
Collapse
Affiliation(s)
- George E Davis
- Department of Pathology, Texas A&M University System Health Science Center, College Station, USA
| | | |
Collapse
|
28
|
van Beijnum JR, Griffioen AW. In silico analysis of angiogenesis associated gene expression identifies angiogenic stage related profiles. Biochim Biophys Acta Rev Cancer 2005; 1755:121-34. [PMID: 16038789 DOI: 10.1016/j.bbcan.2005.06.001] [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: 02/18/2005] [Accepted: 06/14/2005] [Indexed: 01/04/2023]
Abstract
In vitro models have been extensively used to map gene expression in ECs but few studies have used cells from in vivo sources directly. Here, we compare different gene expression surveys on both cultured and fresh tissue derived ECs, and it emerges that gene expression profiles can be paralleled with the angiogenic stage of the cells. ECs stimulated with different growth factors in monolayer cultures exhibit gene expression profiles indicative of an active proliferative state, whereas gene expression in tube forming cells in vitro involves genes implicated in cell adhesion processes. Genes overexpressed in tumor ECs are biased towards extracellular matrix remodeling, a late event in angiogenesis. The elucidation of gene expression profiles under these different conditions will contribute to a better understanding of the molecular mechanisms during angiogenesis in both pathological and physiological circumstances and will have implications for the development of angiogenesis interfering treatment strategies.
Collapse
Affiliation(s)
- Judy R van Beijnum
- Angiogenesis Laboratory, Research Institute for Growth and Development, Departments of Internal Medicine and Pathology, Maastricht University Hospital, PO Box 5800, 6202AZ Maastricht, The Netherlands
| | | |
Collapse
|
29
|
Sun XT, Zhang MY, Shu C, Li Q, Yan XG, Cheng N, Qiu YD, Ding YT. Differential gene expression during capillary morphogenesis in a microcarrier-based three-dimensional in vitro model of angiogenesis with focus on chemokines and chemokine receptors. World J Gastroenterol 2005; 11:2283-90. [PMID: 15818740 PMCID: PMC4305813 DOI: 10.3748/wjg.v11.i15.2283] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2004] [Revised: 03/14/2004] [Accepted: 04/13/2004] [Indexed: 02/06/2023] Open
Abstract
AIM To globally compare the gene expression profiles during the capillary morphogenesis of human microvascular endothelial cells (HMVECs) in an in vitro angiogenesis system with affymetrix oligonucleotide array. METHODS A microcarrier-based in vitro angiogenesis system was developed, in which ECs migrated into the matrix, proliferated, and formed capillary sprouts. The sprouts elongated, branched and formed networks. The total RNA samples from the HMVECs at the selected time points (0.5, 24, and 72 h) during the capillary morphogenesis were used for microarray analyses, and the data were processed with the softwares provided by the manufacturers. The expression patterns of some genes were validated and confirmed by semi-quantitative RT-PCR. The regulated genes were grouped based on their molecular functions and expression patterns, and among them the expression of chemokines and chemokine receptors was specially examined and their functional implications were analyzed. RESULTS A total of 1 961 genes were up- or down-regulated two-folds or above, and among them, 468 genes were up- or down-regulated three-folds or above. The regulated genes could be grouped into categories based on their molecular functions, and were also clustered into six groups based on their patterns of expression. As for chemokines and chemokine receptors, CXCL1/GRO-alpha, CXCL2/GRO-beta, CXCL5/ENA-78, CXCL6/GCP2, IL-8/CXCL8, CXCL12/SDF-1, CXCL9/Mig, CXC11/ITAC, CX3CL1/fractalkine, CCL2/MCP-1, CCL3, CCL5/RANTES, CCL7, CCL15, CCL21, CCL23, CCL28, and CCR1, CCR9, CXCR4 were identified. Moreover, these genes demonstrated different changing patterns during the capillary morphogenesis, which implied that they might have different roles in the sequential process. Among the chemokines identified, CCL2/MCP-1, CCL5/RANTES and CX3CL1 were specially up-regulated at the 24-h time point when the sprouting characterized the morphological change. It was thus suggested that they might exert crucial roles at the early stage of angiogenesis. CONCLUSION The present study demonstrates a global profile of gene expression during endothelial capillary morphogenesis, and the results provide us much information about the molecular mechanisms of angiogenesis, with which further evaluation of individual genes can be conducted.
Collapse
Affiliation(s)
- Xi-Tai Sun
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Medical College, Hepatobiliary Research Institute, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Paris D, Ait-Ghezala G, Mathura VS, Patel N, Quadros A, Laporte V, Mullan M. Anti-angiogenic activity of the mutant Dutch A(beta) peptide on human brain microvascular endothelial cells. ACTA ACUST UNITED AC 2005; 136:212-30. [PMID: 15893605 DOI: 10.1016/j.molbrainres.2005.02.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2004] [Revised: 01/13/2005] [Accepted: 02/05/2005] [Indexed: 01/13/2023]
Abstract
Cerebral amyloid angiopathy is a common pathological feature of patients with Alzheimer's disease (AD) and it is also the hallmark of individuals with a rare autosomal dominant disorder known as hereditary cerebral hemorrhage with amyloidosis-Dutch type. We have shown previously that wild type A(beta) peptides are anti-angiogenic both in vitro and in vivo and could contribute to the compromised cerebrovascular architecture observed in AD. In the present study, we investigated the potential anti-angiogenic activity of the Dutch A(beta)(1-40) (E22Q) peptide. We show that compared to wild type A(beta), freshly solubilized Dutch A(beta) peptide more potently inhibits the formation of capillary structures induced by plating human brain microvascular endothelial cells onto a reconstituted basement membrane. Aggregated/fibrillar preparations of wild type A(beta) and Dutch A(beta) do not appear to be anti-angiogenic in this assay. The stronger anti-angiogenic activity of the Dutch A(beta) compared to wild type A(beta) appears to be related to the increased formation of low molecular weight A(beta) oligomers in the culture medium surrounding human brain microvascular endothelial cells. Using oligonucleotide microarray analysis of human brain microvascular endothelial cells, followed by a genome-scale computational analysis with the Ingenuity Pathways Knowledge Base, networks of genes affected by an anti-angiogenic dose of Dutch A(beta) were identified. This analysis highlights that several biological networks involved in angiogenesis, tumorigenesis, atherosclerosis, cellular migration and proliferation are disrupted in human brain microvascular endothelial cells exposed to Dutch A(beta). Altogether, these data provide new molecular clues regarding the pathological activity of Dutch A(beta) peptide in the cerebrovasculature.
Collapse
Affiliation(s)
- Daniel Paris
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Presta M, Dell'Era P, Mitola S, Moroni E, Ronca R, Rusnati M. Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev 2005; 16:159-78. [PMID: 15863032 DOI: 10.1016/j.cytogfr.2005.01.004] [Citation(s) in RCA: 948] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fibroblast growth factors (FGFs) are a family of heparin-binding growth factors. FGFs exert their pro-angiogenic activity by interacting with various endothelial cell surface receptors, including tyrosine kinase receptors, heparan-sulfate proteoglycans, and integrins. Their activity is modulated by a variety of free and extracellular matrix-associated molecules. Also, the cross-talk among FGFs, vascular endothelial growth factors (VEGFs), and inflammatory cytokines/chemokines may play a role in the modulation of blood vessel growth in different pathological conditions, including cancer. Indeed, several experimental evidences point to a role for FGFs in tumor growth and angiogenesis. This review will focus on the relevance of the FGF/FGF receptor system in adult angiogenesis and its contribution to tumor vascularization.
Collapse
Affiliation(s)
- Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Stanniocalcin was originally described as a hormone with calcitonin-like actions in fish. During the last decade, mammalian forms of stanniocalcin have been identified, and this discovery has led to important advances in our understanding of this enigmatic polypeptide hormone. This review briefly covers some early studies on stanniocalcin in fish and then provides a more in-depth look at some of the more intriguing, new aspects of its functions in mammals. The roles of stanniocalcin in renal function, metabolism, angiogenesis, pregnancy and lactation, bone formation, and neural protection are discussed, along with new information relating to its receptor-mediated sequestration and accumulation in target cell organelles.
Collapse
Affiliation(s)
- Mary E Gerritsen
- Department of Molecular Pharmacology, Exelixis Inc., San Francisco, California 94083, USA
| | | |
Collapse
|
33
|
Abstract
Few terms in the biomedical lexicon are as widely recognized as the phrase blood-brain barrier (BBB). Indeed, it immediately conjures up a "barricade" between the blood and the brain, a feature often considered more obstacle than safeguard. In truth, the BBB performs in both capacities, and it is precisely this duality that imparts such a vital role to the BBB in influencing physiological and pathophysiological processes in the CNS. Although the concept is more than a century old, the BBB continues to remain enigmatic in both substance and idea, with seemingly resolved issues once again beckoning for clarification. In this regard, recent technological advancements, such as sequencing of the human genome and development of microarray analysis, have illuminated novel aspects of vascular gene expression and provoked reconsideration of the cellular and biochemical makeup of the BBB. In light of the critical impact of the BBB in the realms of science and medicine, this Mini-Review will revisit the topic of the composition of the BBB, specifically highlighting how recent developments in endothelial biology have prompted a reevaluation of its precise vascular location. We have intentionally avoided discussing generalized features of the BBB, as these have been skillfully described elsewhere as noted.
Collapse
Affiliation(s)
- Shujun Ge
- Blood-Brain Barrier Laboratory, Department of Pharmacology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | |
Collapse
|
34
|
Abstract
The blood vessels of the pancreatic islets are of crucial importance for oxygen and metabolite supply, and dispersal of secreted hormones. In addition to this, endothelial cells have an important role in the revascularization process after islet transplantation. Studies have reported signs of poor engraftment of transplanted islets, presumably due to impaired revascularization. The aims of this study were to investigate islet endothelial cells and the revascularization process of transplanted islets. The lectin Bandeiraea simplicifolia was found to consistently stain endothelium of both endogenous and transplanted pancreatic islets. By using this marker, we investigated the vascular density of both endogenous and transplanted islets of C57BL/6 mice. One month post-transplantation, a time point when the implants are assumed to be completely revascularized, the graft vascular density was decreased at all investigated implantation sites when compared to endogenous islets. Furthermore, most of the blood vessels were located in the graft connective tissue stroma. Similar results were obtained six months post-transplantation and in cured diabetic animals after one month. In order to evaluate the function of intraportally transplanted islets, we developed a method to retrieve such islets. Enzymatic and mechanic treatment of the liver enabled us to re-isolate the transplanted islets for further in vitro studies. These islets had decreased insulin release, insulin content and glucose oxidation rate when compared to non-transplanted control islets. To understand the role of islet endothelium in the revascularization of transplanted islets we performed angiogenesis microarray studies on islet endothelial cells, from non-cultured, cultured and transplanted islets. We found that the islet endothelium expressed mRNA for both inhibitors and inducers of angiogenesis, and that this expression differed with time. In conclusion, these results provide a useful platform for further studies on the islet endothelium.
Collapse
Affiliation(s)
- Göran Mattsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
35
|
Netherton SJ, Maurice DH. Vascular endothelial cell cyclic nucleotide phosphodiesterases and regulated cell migration: implications in angiogenesis. Mol Pharmacol 2005; 67:263-72. [PMID: 15475573 DOI: 10.1124/mol.104.004853] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis is necessary during embryonic development and wound healing but can be detrimental in pathologies, including cancer. Because initiation of angiogenesis involves migration and proliferation of vascular endothelial cells (VECs) and cAMP-elevating agents inhibit these events, such agents may represent a novel therapeutic avenue to controlling angiogenesis. Intracellular cAMP levels are regulated by their synthesis by adenylyl cyclases and hydrolysis by cyclic nucleotide phosphodiesterases (PDEs). In this report, we show that human VECs express variants of PDE2, PDE3, PDE4, and PDE5 families and demonstrate that the levels of these enzymes differ in VECs derived from aorta, umbilical vein, and microvascular structures. Selective inhibition of PDE2 did not increase cAMP in any VECs, whether in the absence or presence of forskolin, but it did inhibit migration of all VECs studied. Inhibition of PDE4 activity decreased migration, and in conjunction with forskolin, increased cAMP in all VECs studied. PDE3 inhibition potentiated forskolin-induced increases in cAMP and inhibited migration in VECs derived from aorta and umbilical vein but not in microvascular VECs. In experiments with combinations of PDE2, PDE3, and PDE4 inhibitors, a complex interaction between the abilities of these agents to limit human VEC migration was observed. Overall, our data are consistent with the hypothesis that PDE subtype inhibition allows different effects in distinct VEC populations and indicate that these agents may represent novel therapeutic agents to limit angiogenesis in complex human diseases.
Collapse
MESH Headings
- 1-Methyl-3-isobutylxanthine/pharmacology
- 2',3'-Cyclic-Nucleotide Phosphodiesterases/genetics
- 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism
- 3',5'-Cyclic-AMP Phosphodiesterases/genetics
- 3',5'-Cyclic-AMP Phosphodiesterases/metabolism
- 3',5'-Cyclic-GMP Phosphodiesterases/genetics
- 3',5'-Cyclic-GMP Phosphodiesterases/metabolism
- Aorta
- Cell Movement/physiology
- Cyclic AMP/metabolism
- Cyclic AMP/physiology
- Cyclic Nucleotide Phosphodiesterases, Type 2
- Cyclic Nucleotide Phosphodiesterases, Type 3
- Cyclic Nucleotide Phosphodiesterases, Type 4
- Cyclic Nucleotide Phosphodiesterases, Type 5
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiology
- Humans
- Kinetics
- Microcirculation/physiology
- Neovascularization, Physiologic/physiology
- Phosphoric Diester Hydrolases/genetics
- Phosphoric Diester Hydrolases/metabolism
- Quinolones/pharmacology
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Umbilical Veins
Collapse
Affiliation(s)
- Stuart J Netherton
- Department of Pharmacology and Toxicology, Botterell Hall, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | | |
Collapse
|
36
|
Maina JN. Systematic analysis of hematopoietic, vasculogenetic, and angiogenetic phases in the developing embryonic avian lung, Gallus gallus variant domesticus. Tissue Cell 2004; 36:307-22. [PMID: 15385148 DOI: 10.1016/j.tice.2004.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2004] [Revised: 04/14/2004] [Accepted: 05/05/2004] [Indexed: 10/26/2022]
Abstract
In the embryonic lung of the domestic fowl, Gallus gallus variant domesticus, hematogenetic and vasculogenetic cells become ultrastructurally clear from day 4 of development. In the former group of cells, filopodial extensions coalesce, cytoplasm thickens, and accumulating hemoglobin displaces the nucleus peripherally while in the latter, conspicuous filopodial extensions and large nuclei develop as the cells assume a rather stellate appearance. From day 5, erythrocytes and granular leukocytes begin forming from cytoarchitecturally cognate hematogenetic cells. The cells become distinguishable when hemoglobin starts to accumulate in the erythroblasts and electron dense bodies form in the leukoblasts. Vasculogenesis begins from day 7 in different areas of the developing lung: erthrocytes (but not granular leukocytes) appear to attract committed vasculogenetic cells (angioblasts) that form an endothelial lining and vessel wall. Arrangement of angioblasts around forming blood vessels sets the direction along which the vessels sprout (angiogenesis). In some areas of the developing lung, through what seems like an inductive erythropoietic process, arcades of erythrocytes organize. Once endothelial cells surround such continuities, discrete vascular units organize. By day 10, the major parts of the in-built (intrinsic) pulmonary vasculature are assembled. Complete pulmonary circulation (i.e., through the exchange tissue) is not established until after day 18 when the blood capillaries start to develop. Since the precursory erythrocytes do not have a respiratory role, it is imperative that de novo erythropoiesis is essential for vasculogenesis. Diffuse (fragmentary) development and subsequent piecemeal assembly of the pulmonary vascular system may explicate the fabrication of a complex circulatory architecture that grants cross-current, counter-current, and multicapillary serial arterialization designs in the exchange tissue of the avian lung. The exceptional respiratory efficiency of the avian lung is largely attributable to the geometries (physical interfacing) between the bronchial and vascular elements at different levels of morphological organization.
Collapse
Affiliation(s)
- J N Maina
- School of Anatomical Sciences, Faculty of Health Sciences, The University of the Witwatersrand, 7 York Road, Parktown 2193, Johannesburg, South Africa.
| |
Collapse
|
37
|
Peirce SM, Price RJ, Skalak TC. Spatial and temporal control of angiogenesis and arterialization using focal applications of VEGF164 and Ang-1. Am J Physiol Heart Circ Physiol 2003; 286:H918-25. [PMID: 14604856 DOI: 10.1152/ajpheart.00833.2003] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Microvascular networks undergo patterning changes that determine and reflect functional adaptations during tissue remodeling. Alterations in network architectures are a result of complex and integrated signaling events. To understand how two growth factor signals interact to stimulate angiogenesis and arterialization, we engineered spatially directed microvascular pattern changes in vivo by using combinations of focally delivered exogenous growth factors. We implanted microdelivery beads containing recombinant vascular endothelial growth factor-164 (VEGF(164)) and recombinant angiopoietin-1* (Ang-1*) into the dorsal subcutaneous tissue of fully anesthetized male Fischer 344 rats implanted with backpack window chambers, and we quantified vascular patterning changes by using intravital microscopy, a combination of architectural metrics, and immunohistochemistry. Focal delivery of VEGF(164) caused spatially directed increases in both the total number and the density of vessels with diameters <25 microm 7 days after microbead implantation. Increases were maintained out to 14 days but were reduced to control values by day 21. The addition of Ang-1* on day 7 maintained these increases out to day 21, induced vessel order ratios comparable to control levels, and was accompanied by increases in the length density of smooth muscle alpha-actin-positive vessels. We achieved spatial control of patterning changes in vivo by using multisignal stimulation via focal delivery of exogenous growth factor combinations and conclude that Ang-1* administered subsequent to VEGF(164) stimulation induces vascular growth while maintaining a network pattern consistent with native patterns that persist in the presence of vehicle control stimulation.
Collapse
Affiliation(s)
- Shayn M Peirce
- Dept. of Biomedical Engineering, Univ. of Virginia, Box 800759, Health System, Charlottesville, VA 22908, USA.
| | | | | |
Collapse
|
38
|
Gerritsen ME, Tomlinson JE, Zlot C, Ziman M, Hwang S. Using gene expression profiling to identify the molecular basis of the synergistic actions of hepatocyte growth factor and vascular endothelial growth factor in human endothelial cells. Br J Pharmacol 2003; 140:595-610. [PMID: 14504135 PMCID: PMC1574080 DOI: 10.1038/sj.bjp.0705494] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatocyte growth factor (HGF) and vascular endothelial cell growth factor (VEGF) are two potent endothelial mitogens with demonstrated angiogenic activities in animal models of therapeutic angiogenesis. Several recent studies suggest that these growth factors may act synergistically, although the mechanism of this interaction is not understood. Changes in the gene expression profile of human umbilical vein endothelial cells treated with HGF, VEGF or the combination of the two were analyzed with high-density oligonucleotide arrays, representing approximately 22000 genes. Notably, the genes significantly up- and downregulated by VEGF versus HGF exhibited very little overlap, indicating distinct signal transduction pathways. The combination of HGF and VEGF markedly increased the number of significantly up- and downregulated genes. At 4 h, the combination of the two growth factors induced a number of chemokine and cytokines and their receptors (IL-8, IL-6, IL-11, CCR6, CXCR1,CXC1 and IL17RC), numerous genes involved in growth factor signal transduction (egr-1, fosB, grb10, grb14,MAP2K3,MAP3K8, MAPKAP2,MPK3, DUSP4 and DUSP6), as well as a number of other growth factors (PDGFA, BMP2, Hb-EGF, FGF16, heuregulin beta 1, c-kit ligand, angiopoietin 2 and angiopoietin 4 and VEGFC). In addition, the VEGF receptors neuropilin-1 and flt-1 were also upregulated. At 24 h, a clear 'cell cycle' signature is noted, with the upregulated expression of various cell cycle control proteins and gene involved in the regulation of mitosis and mitotic spindle assembly. The receptor for HGF, c-met, is also upregulated. These data are consistent with the hypothesis that the combination of HGF and VEGF results in the cooperative upregulation of a number of different molecular pathways leading to a more robust proliferative response, that is, growth factor(s), receptors, molecules involved in growth factor signal transduction, as well as, at later time points, upregulation of the necessary cellular proteins required for cells to escape cell cycle arrest and enter the cell cycle.
Collapse
Affiliation(s)
- Mary E Gerritsen
- Department of Vascular Biology, Millennium Pharmaceuticals, South San Francisco, CA 94080, USA.
| | | | | | | | | |
Collapse
|