1
|
Libby JR, Royce H, Walker SR, Li L. The role of extracellular matrix in angiogenesis: Beyond adhesion and structure. BIOMATERIALS AND BIOSYSTEMS 2024; 15:100097. [PMID: 39129826 PMCID: PMC11315062 DOI: 10.1016/j.bbiosy.2024.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/19/2024] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
While the extracellular matrix (ECM) has long been recognized for its structural contributions, anchoring cells for adhesion, providing mechanical support, and maintaining tissue integrity, recent efforts have elucidated its dynamic, reciprocal, and diverse properties on angiogenesis. The ECM modulates angiogenic signaling and mechanical transduction, influences the extent and degree of receptor activation, controls cellular behaviors, and serves as a reservoir for bioactive macromolecules. Collectively, these factors guide the formation, maturation, and stabilization of a functional vascular network. This review aims to shed light on the versatile roles of the ECM in angiogenesis, transcending its traditional functions as a mere structural material. We will explore its engagement and synergy in signaling modulation, interactions with various angiogenic factors, and highlight its importance in both health and disease. By capturing the essence of the ECM's diverse functionalities, we highlight the significance in the broader context of vascular biology, enabling the design of novel biomaterials to engineer vascularized tissues and their potential therapeutic implications.
Collapse
Affiliation(s)
- Jaxson R. Libby
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Haley Royce
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
| | - Sarah R. Walker
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Linqing Li
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
- Department of Chemistry, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
2
|
Gudenschwager-Basso EK, Frydman G, Weerakoon S, Andargachew H, Piltaver CM, Huckle WR. Morphological evaluation of the feline placenta correlates with gene expression of vascular growth factors and receptors†. Biol Reprod 2024; 110:569-582. [PMID: 38092011 DOI: 10.1093/biolre/ioad167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 09/06/2023] [Accepted: 11/30/2023] [Indexed: 03/16/2024] Open
Abstract
Placental angiogenesis is critical for normal development. Angiogenic factors and their receptors are key regulators of this process. Dysregulated placental vascular development is associated with pregnancy complications. Despite their importance, vascular growth factor expression has not been thoroughly correlated with placental morphologic development across gestation in cats. We postulate that changes in placental vessel morphology can be appreciated as consequences of dynamic expression of angiogenic signaling agents. Here, we characterized changes in placental morphology alongside expression analysis of angiogenic factor splice variants and receptors throughout pregnancy in domestic shorthair cats. We observed increased vascular and lamellar density in the lamellar zone during mid-pregnancy. Immunohistochemical analysis localized the vascular endothelial growth factor A (VEGF-A) receptor KDR to endothelial cells of the maternal and fetal microvasculatures. PlGF and its principal receptor Flt-1 were localized to the trophoblasts and fetal vasculature. VEGF-A was found in trophoblast cells and associated with endothelial cells. We detected expression of two Plgf splice variants and four Vegf-a variants. Quantitative real-time polymerase chain reaction analysis showed upregulation of mRNAs encoding pan Vegf-a and all Vegf-a splice forms at gestational days 30-35. Vegf-A showed a marked relative increase in expression during mid-pregnancy, consistent with the pro-angiogenic changes seen in the lamellar zone at days 30-35. Flt-1 was upregulated during late pregnancy. Plgf variants showed stable expression during the first two-thirds of pregnancy, followed by a marked increase toward term. These findings revealed specific spatiotemporal expression patterns of VEGF-A family members consistent with pivotal roles during normal placental development.
Collapse
Affiliation(s)
- Erwin K Gudenschwager-Basso
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Galit Frydman
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Shaneke Weerakoon
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
- Virginia Tech Carilion School of Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Hariyat Andargachew
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Cassandra M Piltaver
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - William R Huckle
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| |
Collapse
|
3
|
Ternifi R, Wang Y, Gu J, Polley EC, Carter JM, Pruthi S, Boughey JC, Fazzio RT, Fatemi M, Alizad A. Ultrasound high-definition microvasculature imaging with novel quantitative biomarkers improves breast cancer detection accuracy. Eur Radiol 2022; 32:7448-7462. [PMID: 35486168 PMCID: PMC9616967 DOI: 10.1007/s00330-022-08815-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/24/2022] [Accepted: 04/12/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To overcome the limitations of power Doppler in imaging angiogenesis, we sought to develop and investigate new quantitative biomarkers of a contrast-free ultrasound microvasculature imaging technique for differentiation of benign from malignant pathologies of breast lesion. METHODS In this prospective study, a new high-definition microvasculature imaging (HDMI) was tested on 521 patients with 527 ultrasound-identified suspicious breast masses indicated for biopsy. Four new morphological features of tumor microvessels, microvessel fractal dimension (mvFD), Murray's deviation (MD), bifurcation angle (BA), and spatial vascularity pattern (SVP) as well as initial biomarkers were extracted and analyzed, and the results correlated with pathology. Multivariable logistic regression analysis was used to study the performance of different prediction models, initial biomarkers, new biomarkers, and combined new and initial biomarkers in differentiating benign from malignant lesions. RESULTS The new HDMI biomarkers, mvFD, BA, MD, and SVP, were statistically significantly different in malignant and benign lesions, regardless of tumor size. Sensitivity and specificity of the new biomarkers in lesions > 20 mm were 95.6% and 100%, respectively. Combining the new and initial biomarkers together showed an AUC, sensitivity, and specificity of 97% (95% CI: 95-98%), 93.8%, and 89.2%, respectively, for all lesions regardless of mass size. The classification was further improved by adding the Breast Imaging Reporting and Data System (BI-RADS) score to the prediction model, showing an AUC, sensitivity, and specificity of 97% (95% CI: 95-98%), 93.8%, and 89.2%, respectively. CONCLUSION The addition of new quantitative HDMI biomarkers significantly improved the accuracy in breast lesion characterization when used as a complementary imaging tool to the conventional ultrasound. KEY POINTS • Novel quantitative biomarkers extracted from tumor microvessel images increase the sensitivity and specificity in discriminating malignant from benign breast masses. • New HDMI biomarkers Murray's deviation, bifurcation angles, microvessel fractal dimension, and spatial vascularity pattern outperformed the initial biomarkers. • The addition of BI-RADS scores based on US descriptors to the multivariable analysis using all biomarkers remarkably increased the sensitivity, specificity, and AUC in all size groups.
Collapse
Affiliation(s)
- Redouane Ternifi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Yinong Wang
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Juanjuan Gu
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Eric C Polley
- Department of Health Science, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sandhya Pruthi
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Robert T Fazzio
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Mostafa Fatemi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Azra Alizad
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
4
|
Characterization of the Expression of Angiogenic Factors in Cutaneous Squamous Cell Carcinoma of Domestic Cats. Vet Sci 2022; 9:vetsci9070375. [PMID: 35878392 PMCID: PMC9351683 DOI: 10.3390/vetsci9070375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is a common malignant skin cancer with a significant impact on health, and it is important to determine the degree of reliance of CSCC on angiogenesis for growth and metastasis. Major regulators of angiogenesis are the vascular endothelial growth factor (VEGF) family and their associated receptors. Alternative pre-mRNA splicing produces multiple isoforms of VEGF-A and PLGF with distinct biological properties. Several studies highlight the function of VEGF-A in CSCC, but there are no studies of the different isoforms of VEGF-A and PLGF for this neoplasm. We characterized the expression of three isoforms of VEGF-A, two isoforms of PLGF, and their receptors in cat CSCC biopsies compared to normal haired skin (NHS). Although our results revealed no significant changes in transcript levels of panVEGF-A or their isoforms, the mRNA levels of PLGF I and the receptors Flt-1 and KDR were downregulated in CSCC compared to NHS. Differences were observed in ligand:receptor mRNA expression ratio, with the expression of VEGF-A relative to its receptor KDR higher in CSCC, which is consistent with our hypothesis and prior human SCC studies. Immunolocalization in tissue showed increased expression of all measured factors and receptors in tumor cells compared to NHS and surrounding vasculature. We conclude that the factors measured may play a pivotal role in CSCC growth, although further studies are needed to clarify the role of angiogenic factors in feline CSCC.
Collapse
|
5
|
Pahapale GJ, Tao J, Nikolic M, Gao S, Scarcelli G, Sun SX, Romer LH, Gracias DH. Directing Multicellular Organization by Varying the Aspect Ratio of Soft Hydrogel Microwells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104649. [PMID: 35434926 PMCID: PMC9189654 DOI: 10.1002/advs.202104649] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/08/2022] [Indexed: 06/03/2023]
Abstract
Multicellular organization with precise spatial definition is essential to various biological processes, including morphogenesis, development, and healing in vascular and other tissues. Gradients and patterns of chemoattractants are well-described guides of multicellular organization, but the influences of 3D geometry of soft hydrogels are less well defined. Here, the discovery of a new mode of endothelial cell self-organization guided by combinatorial effects of stiffness and geometry, independent of protein or chemical patterning, is described. Endothelial cells in 2 kPa microwells are found to be ≈30 times more likely to migrate to the edge to organize in ring-like patterns than in stiff 35 kPa microwells. This organization is independent of curvature and significantly more pronounced in 2 kPa microwells with aspect ratio (perimeter/depth) < 25. Physical factors of cells and substrates that drive this behavior are systematically investigated and a mathematical model that explains the organization by balancing the dynamic interaction between tangential cytoskeletal tension, cell-cell, and cell-substrate adhesion is presented. These findings demonstrate the importance of combinatorial effects of geometry and stiffness in complex cellular organization that can be leveraged to facilitate the engineering of bionics and integrated model organoid systems with customized nutrient vascular networks.
Collapse
Affiliation(s)
- Gayatri J. Pahapale
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiaxiang Tao
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Milos Nikolic
- Maryland Biophysics ProgramInstitute for Physical Science and TechnologyUniversity of MarylandCollege ParkMD20742USA
| | - Sammy Gao
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Giuliano Scarcelli
- Maryland Biophysics ProgramInstitute for Physical Science and Technology and Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Sean X. Sun
- Department of Mechanical EngineeringCell Biologyand Institute of NanoBioTechnology (INBT)Johns Hopkins UniversityBaltimoreMD21218USA
| | - Lewis H. Romer
- Department of Cell BiologyAnesthesiology and Critical Care MedicineBiomedical EngineeringPediatricsand Center for Cell DynamicsJohns Hopkins School of MedicineBaltimoreMD21205USA
| | - David H. Gracias
- Department of Chemical and Biomolecular EngineeringMaterials Science and EngineeringChemistry and Laboratory for Computational Sensing and Robotics (LCSR)Johns Hopkins UniversityBaltimoreMD21218USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins School of MedicineBaltimoreMD21205USA
| |
Collapse
|
6
|
Angiogenesis and Functional Vessel Formation Induced by Interstitial Flow and Vascular Endothelial Growth Factor Using a Microfluidic Chip. MICROMACHINES 2022; 13:mi13020225. [PMID: 35208349 PMCID: PMC8876009 DOI: 10.3390/mi13020225] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
Abstract
Angiogenesis occurs during both physiological and pathological processes. In this study, a microfluidic chip for the development of angiogenesis was utilized to assess angiogenic sprouting and functional vessel formation. We also found that vascular endothelial growth factor (VEGF) was a determinant of the initiation of vascular sprouts, while the direction of these sprouts was greatly influenced by interstitial flow. Isoforms of VEGF such as VEGF121, VEGF165, and VEGF189 displayed different angiogenic properties on the chip as assessed by sprout length and number, vessel perfusion, and connectivity. VEGF165 had the highest capacity to induce vascular sprouting among the three isoforms assessed and furthermore, also induced functional vessel formation. This chip could be used to analyze the effect of different angiogenic factors and drugs, as well as to explore the mechanism of angiogenesis induced by such factors.
Collapse
|
7
|
Dahan S, Sharma A, Cohen K, Baker M, Taqatqa N, Bentata M, Engal E, Siam A, Kay G, Drier Y, Elias S, Salton M. VEGFA's distal enhancer regulates its alternative splicing in CML. NAR Cancer 2021; 3:zcab029. [PMID: 34316716 PMCID: PMC8276762 DOI: 10.1093/narcan/zcab029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/21/2021] [Accepted: 06/27/2021] [Indexed: 12/28/2022] Open
Abstract
Enhancer demethylation in leukemia has been shown to lead to overexpression of genes which promote cancer characteristics. The vascular endothelial growth factor A (VEGFA) enhancer, located 157 Kb downstream of its promoter, is demethylated in chronic myeloid leukemia (CML). VEGFA has several alternative splicing isoforms with different roles in cancer progression. Since transcription and splicing are coupled, we wondered whether VEGFA enhancer activity can also regulate the gene's alternative splicing to contribute to the pathology of CML. Our results show that mutating the VEGFA +157 enhancer promotes exclusion of exons 6a and 7 and activating the enhancer by tethering a chromatin activator has the opposite effect. In line with these results, CML patients present with high expression of +157 eRNA and inclusion of VEGFA exons 6a and 7. In addition, our results show that the positive regulator of RNAPII transcription elongation, CCNT2, binds VEGFA's promoter and enhancer, and its silencing promotes exclusion of exons 6a and 7 as it slows down RNAPII elongation rate. Thus our results suggest that VEGFA's +157 enhancer regulates its alternative splicing by increasing RNAPII elongation rate via CCNT2. Our work demonstrates for the first time a connection between an endogenous enhancer and alternative splicing regulation of its target gene.
Collapse
Affiliation(s)
- Sara Dahan
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Aveksha Sharma
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Klil Cohen
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Mai Baker
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Nadeen Taqatqa
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Mercedes Bentata
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Eden Engal
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Ahmad Siam
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Gillian Kay
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yotam Drier
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Shlomo Elias
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Maayan Salton
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
8
|
Ganta VC, Annex BH. Peripheral vascular disease: preclinical models and emerging therapeutic targeting of the vascular endothelial growth factor ligand-receptor system. Expert Opin Ther Targets 2021; 25:381-391. [PMID: 34098826 PMCID: PMC8573823 DOI: 10.1080/14728222.2021.1940139] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/04/2021] [Indexed: 10/21/2022]
Abstract
Introduction: Vascular endothelial growth factor (VEGF)-A is a sought therapeutic target for PAD treatment because of its potent role in angiogenesis. However, no therapeutic benefit was achieved in VEGF-A clinical trials, suggesting that our understanding of VEGF-A biology and ischemic angiogenic processes needs development. Alternate splicing in VEGF-A produces pro- and anti-angiogenic VEGF-A isoforms; the only difference being a 6-amino acid switch in the C-terminus of the final 8th exon of the gene. This finding has changed our understanding of VEGF-A biology and may explain the lack of benefit in VEGF-A clinical trials. It presents new therapeutic opportunities for peripheral arterial disease (PAD) treatment.Areas covered: Literature search was conducted to include: 1) predicted mechanism by which the anti-angiogenic VEGF-A isoform would inhibit angiogenesis, 2) unexpected mechanism of action, and 3) how this mechanism revealed novel signaling pathways that may enhance future therapeutics in PAD.Expert opinion: Inhibiting a specific anti-angiogenic VEGF-A isoform in ischemic muscle promotes perfusion recovery in preclinical PAD. Additional efforts focused on the production of these isoforms, and the pathways altered by modulating different VEGF receptor-ligand interactions, and how this new data may allow bedside progress offers new approaches to PAD are discussed.I.
Collapse
Affiliation(s)
- Vijay Chaitanya Ganta
- Department of Medicine and Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Brian H Annex
- Department of Medicine and Vascular Biology Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
9
|
Usuelli M, Meyer T, Mezzenga R, Mitsi M. VEGF and VEGFR2 bind to similar pH-sensitive sites on fibronectin, exposed by heparin-mediated conformational changes. J Biol Chem 2021; 296:100584. [PMID: 33771558 PMCID: PMC8102423 DOI: 10.1016/j.jbc.2021.100584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Physical interactions between vascular endothelial growth factor (VEGF), a central player in blood endothelial cell biology, and fibronectin, a major fibrillar protein of the extracellular matrix, are important determinants of angiogenic activity in health and disease. Conditions signaling the need for new blood vessel growth, such as hypoxia and low extracellular pH, increase VEGF–fibronectin interactions. These interactions can be further fine-tuned through changes in the availability of the VEGF-binding sites on fibronectin, regulated by conformational changes induced by heparin and heparan sulfate chains within the extracellular matrix. These interactions may alter VEGF bioavailability, generate gradients, or alter the way VEGF is recognized by and activates its cell-surface receptors. Here, using equilibrium and kinetic studies, we discovered that fibronectin can also interact with the extracellular domain of the VEGF receptor 2 (VEGFR2). The VEGFR2-binding sites on fibronectin show great similarity to the VEGF-binding sites, as they were also exposed upon heparin-induced conformational changes in fibronectin, and the interaction was enhanced at acidic pH. Kinetic parameters and affinities for VEGF and VEGFR2 binding to fibronectin were determined by surface plasmon resonance measurements, revealing two populations of fibronectin-binding sites for each molecule. Our data also suggest that a VEGF/VEGFR2/fibronectin triple complex may be formed by VEGF or VEGFR2 first binding to fibronectin and subsequently recruiting the third binding partner. The formation of such a complex may lead to the activation of distinct angiogenic signaling pathways, offering new possibilities for clinical applications that target angiogenesis.
Collapse
Affiliation(s)
- Mattia Usuelli
- Laboratory of Food and Soft Materials, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Timmy Meyer
- Laboratory of Food and Soft Materials, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Raffaele Mezzenga
- Laboratory of Food and Soft Materials, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| | - Maria Mitsi
- Laboratory of Food and Soft Materials, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
Li J, Liu X, Zang S, Zhou J, Zhang F, Sun B, Qi D, Li X, Kong J, Jin D, Yang X, Luo Y, Lu Y, Lin B, Niu W, Liu T. Small extracellular vesicle-bound vascular endothelial growth factor secreted by carcinoma-associated fibroblasts promotes angiogenesis in a bevacizumab-resistant manner. Cancer Lett 2020; 492:71-83. [PMID: 32860852 DOI: 10.1016/j.canlet.2020.08.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 01/08/2023]
Abstract
The blood vessel growth inhibitor bevacizumab targets vascular endothelial growth factor (VEGF), a crucial regulator of angiogenesis. Recently, small extracellular vesicles (sEVs) have been demonstrated to be important vehicles in the transport of growth factors to target cells. In this study, we isolated primary carcinoma-associated fibroblasts (CAFs) from four human oral squamous cell carcinoma (OSCC) specimens. Compared with other non-extracellular vesicle components, CAF-derived sEVs were found to be the main regulators of angiogenesis. The ability of CAF sEVs to activate VEGF receptor 2 (VEGFR2) signaling in human umbilical vein endothelial cells (HUVEC) was dependent on the association between sEVs and VEGF. In addition, sEV-bound VEGF secreted by CAFs further activated VEGFR2 signaling in HUVEC in a bevacizumab-resistant manner. VEGF was found to interact with heparan sulfate proteoglycans on the CAF sEV surface and could be released by heparinase I/III. The bioactivity of the dissociated VEGF was retained in vitro and in vivo and could be neutralized by bevacizumab. These findings suggest that the combined use of heparinase and bevacizumab might inhibit angiogenesis in patients with high levels of sEV-bound VEGF.
Collapse
Affiliation(s)
- Jiao Li
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Xue Liu
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Shizhu Zang
- Biomedical Engineering Department, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Jiasheng Zhou
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Fuyin Zhang
- Department of Oral Surgery, The Second Affiliated Hospital, Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, China
| | - Bo Sun
- Department of Oral Surgery, The Second Affiliated Hospital, Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, China
| | - Dongyuan Qi
- Department of Oral Surgery, The First Affiliated Hospital of Dalian Medical University, No.222 Zhongshan Road, Dalian, 116023, China
| | - Xiaojie Li
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Jing Kong
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Dong Jin
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China
| | - Yong Luo
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, No.2 Linggong Road, Ganjingzi District, Dalian, 116023, China
| | - Yao Lu
- Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No.457 Zhongshan Road, Dalian, 116023, China
| | - Bingcheng Lin
- Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No.457 Zhongshan Road, Dalian, 116023, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China.
| | - Tingjiao Liu
- School of Stomatology, Dalian Medical University, No.9 West Section, South Road of Lvshun, Dalian, 116044, China.
| |
Collapse
|
11
|
Adachi Y, Matsuki M, Watanabe H, Takase K, Kodama K, Matsui J, Funahashi Y, Nomoto K. Antitumor and Antiangiogenic Activities of Lenvatinib in Mouse Xenograft Models of Vascular Endothelial Growth Factor-Induced Hypervascular Human Hepatocellular Carcinoma. Cancer Invest 2019; 37:185-198. [PMID: 31006280 DOI: 10.1080/07357907.2019.1601209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
High expression of vascular endothelial growth factor (VEGF) in patients with hepatocellular carcinoma (HCC) is associated with poor prognosis. Here, we investigated the antitumor activity of lenvatinib, a multiple receptor tyrosine kinase inhibitor, in VEGF-overexpressing HCC models. In human umbilical vein endothelial cells, lenvatinib showed potent inhibitory activities against VEGF-induced proliferation and VEGF/basic fibroblast growth factor-induced tube formation. In VEGF-overexpressing HCC xenograft models, characterized by aggressive tumor growth and hypervascularity, lenvatinib had significant antitumor and antiangiogenic activities. These results suggest that potent activity of lenvatinib against VEGF signaling underlies its antitumor and antiangiogenic activities in the hypervascular HCC models.
Collapse
Affiliation(s)
- Yusuke Adachi
- a Tsukuba Research Laboratories , Eisai Co., Ltd , Ibaraki , Japan
| | - Masahiro Matsuki
- a Tsukuba Research Laboratories , Eisai Co., Ltd , Ibaraki , Japan
| | - Hideki Watanabe
- a Tsukuba Research Laboratories , Eisai Co., Ltd , Ibaraki , Japan
| | - Kazuma Takase
- a Tsukuba Research Laboratories , Eisai Co., Ltd , Ibaraki , Japan
| | - Kotaro Kodama
- a Tsukuba Research Laboratories , Eisai Co., Ltd , Ibaraki , Japan
| | | | | | | |
Collapse
|
12
|
KITAYAMA Y, TAKEUCHI T. Oriented Immobilization-based Molecular Imprinting for Constructing Nanocavities Capable of Precise Molecular Recognition. BUNSEKI KAGAKU 2019. [DOI: 10.2116/bunsekikagaku.68.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Kamon Y, Takeuchi T. Molecularly Imprinted Nanocavities Capable of Ligand-Binding Domain and Size/Shape Recognition for Selective Discrimination of Vascular Endothelial Growth Factor Isoforms. ACS Sens 2018; 3:580-586. [PMID: 29441779 DOI: 10.1021/acssensors.7b00622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Vascular endothelial growth factor 165 (VEGF165) is known to be predominantly expressed in the first stage of vascularization; therefore, the detection of VEGF165 is important in the stage diagnosis of cancers. Molecularly imprinted nanocavities, capable of the selective discrimination of VEGF165 from other VEGF isoforms, were prepared by surface-initiated atom transfer radical polymerization. VEGF165 was immobilized on a gold-coated glass substrate by anchored heparin moieties, where the immobilized heparin was able to capture VEGF165 by binding with the heparin-binding domain (HBD) on VEGF165. Molecular imprinting was conducted on the immobilized VEGF165 by using methacrylic acid (MAA) as a functional monomer to interact with basic amino acids outside of the HBD of VEGF165 by electrostatic interaction. After the removal of VEGF165 from the obtained polymer thin layer (ca. 7 nm), VEGF165-imprinted nanocavities remained, in which the heparin moiety and MAA residues were located in suitable positions for VEGF165 recognition. The molecularly imprinted polymer (MIP) thin layer showed a binding affinity for VEGF165 (dissociation constant: 3.4 nM) that was ten times higher than that of the substrate before polymerization (heparin-immobilized substrate). A much lower binding affinity for VEGF121, which contains no heparin-binding domain, was observed. Moreover, the MIP thin layer distinguished VEGF165 from VEGF189, which possesses a larger molecular size than VEGF165, an amino acid sequence homology of 87%, and contains HBDs, whereas the heparin-immobilized substrate showed almost no selectivity. These results suggested that the heparin moiety within the nanocavity provided HBD selectivity and the polymer matrix composed of the molecularly imprinted nanocavity provided size/shape selectivity, which resulted in the highly selective discrimination of VEGF isoforms.
Collapse
Affiliation(s)
- Yuri Kamon
- Graduate School of Engineering, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Toshifumi Takeuchi
- Graduate School of Engineering, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
14
|
Clegg LE, Mac Gabhann F. A computational analysis of pro-angiogenic therapies for peripheral artery disease. Integr Biol (Camb) 2018; 10:18-33. [PMID: 29327758 PMCID: PMC7017937 DOI: 10.1039/c7ib00218a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inducing therapeutic angiogenesis to effectively form hierarchical, non-leaky networks of perfused vessels in tissue engineering applications and ischemic disease remains an unmet challenge, despite extensive research and multiple clinical trials. Here, we use a previously-developed, multi-scale, computational systems pharmacology model of human peripheral artery disease to screen a diverse array of promising pro-angiogenic strategies, including gene therapy, biomaterials, and antibodies. Our previously-validated model explicitly accounts for VEGF immobilization, Neuropilin-1 binding, and weak activation of VEGF receptor 2 (VEGFR2) by the "VEGFxxxb" isoforms. First, we examine biomaterial-based delivery of VEGF engineered for increased affinity to the extracellular matrix. We show that these constructs maintain VEGF close to physiological levels and extend the duration of VEGFR2 activation. We demonstrate the importance of sub-saturating VEGF dosing to prevent angioma formation. Second, we examine the potential of ligand- or receptor-based gene therapy to normalize VEGF receptor signaling. Third, we explore the potential for antibody-based pro-angiogenic therapy. Our model supports recent observations that improvement in perfusion following treatment with anti-VEGF165b in mice is mediated by VEGF-receptor 1, not VEGFR2. Surprisingly, the model predicts that the approved anti-VEGF cancer drug, bevacizumab, may actually improve signaling of both VEGFR1 and VEGFR2 via a novel 'antibody swapping' effect that we demonstrate here. Altogether, this model provides insight into the mechanisms of action of several classes of pro-angiogenic strategies within the context of the complex molecular and physiological processes occurring in vivo. We identify molecular signaling similarities between promising approaches and key differences between promising and ineffective strategies.
Collapse
Affiliation(s)
- Lindsay E Clegg
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | | |
Collapse
|
15
|
Guo M, Shi JH, Wang PL, Shi DZ. Angiogenic Growth Factors for Coronary Artery Disease: Current Status and Prospects. J Cardiovasc Pharmacol Ther 2017; 23:130-141. [PMID: 29025278 DOI: 10.1177/1074248417735399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ming Guo
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun-He Shi
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Pei-Li Wang
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Da-Zhuo Shi
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Clegg LE, Mac Gabhann F. A computational analysis of in vivo VEGFR activation by multiple co-expressed ligands. PLoS Comput Biol 2017; 13:e1005445. [PMID: 28319199 PMCID: PMC5378411 DOI: 10.1371/journal.pcbi.1005445] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 04/03/2017] [Accepted: 03/08/2017] [Indexed: 12/16/2022] Open
Abstract
The splice isoforms of vascular endothelial growth A (VEGF) each have different affinities for the extracellular matrix (ECM) and the coreceptor NRP1, which leads to distinct vascular phenotypes in model systems expressing only a single VEGF isoform. ECM-immobilized VEGF can bind to and activate VEGF receptor 2 (VEGFR2) directly, with a different pattern of site-specific phosphorylation than diffusible VEGF. To date, the way in which ECM binding alters the distribution of isoforms of VEGF and of the related placental growth factor (PlGF) in the body and resulting angiogenic signaling is not well-understood. Here, we extend our previous validated cell-level computational model of VEGFR2 ligation, intracellular trafficking, and site-specific phosphorylation, which captured differences in signaling by soluble and immobilized VEGF, to a multi-scale whole-body framework. This computational systems pharmacology model captures the ability of the ECM to regulate isoform-specific growth factor distribution distinctly for VEGF and PlGF, and to buffer free VEGF and PlGF levels in tissue. We show that binding of immobilized growth factor to VEGF receptors, both on endothelial cells and soluble VEGFR1, is likely important to signaling in vivo. Additionally, our model predicts that VEGF isoform-specific properties lead to distinct profiles of VEGFR1 and VEGFR2 binding and VEGFR2 site-specific phosphorylation in vivo, mediated by Neuropilin-1. These predicted signaling changes mirror those observed in murine systems expressing single VEGF isoforms. Simulations predict that, contrary to the 'ligand-shifting hypothesis,' VEGF and PlGF do not compete for receptor binding at physiological concentrations, though PlGF is predicted to slightly increase VEGFR2 phosphorylation when over-expressed by 10-fold. These results are critical to design of appropriate therapeutic strategies to control VEGF availability and signaling in regenerative medicine applications.
Collapse
Affiliation(s)
- Lindsay E. Clegg
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Feilim Mac Gabhann
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
|
18
|
Yamamoto H, Rundqvist H, Branco C, Johnson RS. Autocrine VEGF Isoforms Differentially Regulate Endothelial Cell Behavior. Front Cell Dev Biol 2016; 4:99. [PMID: 27709112 PMCID: PMC5030275 DOI: 10.3389/fcell.2016.00099] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/26/2016] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF) is involved in all the essential biology of endothelial cells, from proliferation to vessel function, by mediating intercellular interactions and monolayer integrity. It is expressed as three major alternative spliced variants. In mice, these are VEGF120, VEGF164, and VEGF188, each with different affinities for extracellular matrices and cell surfaces, depending on the inclusion of heparin-binding sites, encoded by exons 6 and 7. To determine the role of each VEGF isoform in endothelial homeostasis, we compared phenotypes of primary endothelial cells isolated from lungs of mice expressing single VEGF isoforms in normoxic and hypoxic conditions. The differential expression and distribution of VEGF isoforms affect endothelial cell functions, such as proliferation, adhesion, migration, and integrity, which are dependent on the stability of and affinity to VEGF receptor 2 (VEGFR2). We found a correlation between autocrine VEGF164 and VEGFR2 stability, which is also associated with increased expression of proteins involved in cell adhesion. Endothelial cells expressing only VEGF188, which localizes to extracellular matrices or cell surfaces, presented a mesenchymal morphology and weakened monolayer integrity. Cells expressing only VEGF120 lacked stable VEGFR2 and dysfunctional downstream processes, rendering the cells unviable. Endothelial cells expressing these different isoforms in isolation also had differing rates of apoptosis, proliferation, and signaling via nitric oxide (NO) synthesis. These data indicate that autocrine signaling of each VEGF isoform has unique functions on endothelial homeostasis and response to hypoxia, due to both distinct VEGF distribution and VEGFR2 stability, which appears to be, at least partly, affected by differential NO production. This study demonstrates that each autocrine VEGF isoform has a distinct effect on downstream functions, namely VEGFR2-regulated endothelial cell homeostasis in normoxia and hypoxia.
Collapse
Affiliation(s)
- Hideki Yamamoto
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
| | - Helene Rundqvist
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholm, Sweden
| | - Cristina Branco
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
| | - Randall S. Johnson
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholm, Sweden
| |
Collapse
|
19
|
Kareva I. Escape from tumor dormancy and time to angiogenic switch as mitigated by tumor-induced stimulation of stroma. J Theor Biol 2016; 395:11-22. [PMID: 26826487 DOI: 10.1016/j.jtbi.2016.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 11/28/2022]
Abstract
A variety of mechanisms have been proposed to explain "cancer without disease", the state of tumor dormancy, characterized by balance in cell proliferation and cell death within a tumor. Here we have investigated a theoretical construct, whereby one of such mechanisms, the time to induction of angiogenesis, or "angiogenic switch", is mitigated by the degree of stromal stimulation by the tumor. We tested this hypothesis and its implications by introducing a mathematical model that captures how angiogenesis regulators, released from the platelet clot, contribute to formation of normal vasculature. We then modified the model to introduce tumor-induced increase in production of angiogenesis regulators and were able to simulate pathological angiogenesis. Through varying parameters governing the degree of tumor-induced stromal stimulation, we were able to qualitatively replicate experimentally observed growth curves for both dormant and actively growing tumors of breast cancer and liposarcoma. In fact, variation of very few parameters was sufficient to replicate any experimentally observed time to angiogenic switch in the available data. Finally, we investigated the effects of tighter binding isoforms of angiogenesis stimulators on neovasculature formation and tumor growth, which may provide an explanation for variations in angiogenesis -dependence in tumors of different tissue origin.
Collapse
Affiliation(s)
- Irina Kareva
- Floating Hospital for Children at Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA; Mathematical, Computational and Modeling Sciences Center, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
20
|
Sargent KM, McFee RM, Spuri Gomes R, Cupp AS. Vascular endothelial growth factor A: just one of multiple mechanisms for sex-specific vascular development within the testis? J Endocrinol 2015; 227:R31-50. [PMID: 26562337 PMCID: PMC4646736 DOI: 10.1530/joe-15-0342] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/16/2015] [Indexed: 01/25/2023]
Abstract
Testis development from an indifferent gonad is a critical step in embryogenesis. A hallmark of testis differentiation is sex-specific vascularization that occurs as endothelial cells migrate from the adjacent mesonephros into the testis to surround Sertoli-germ cell aggregates and induce seminiferous cord formation. Many in vitro experiments have demonstrated that vascular endothelial growth factor A (VEGFA) is a critical regulator of this process. Both inhibitors to VEGFA signal transduction and excess VEGFA isoforms in testis organ cultures impaired vascular development and seminiferous cord formation. However, in vivo models using mice which selectively eliminated all VEGFA isoforms: in Sertoli and germ cells (pDmrt1-Cre;Vegfa(-/-)); Sertoli and Leydig cells (Amhr2-Cre;Vegfa(-/-)) or Sertoli cells (Amh-Cre;Vegfa(-/-) and Sry-Cre;Vegfa(-/-)) displayed testes with observably normal cords and vasculature at postnatal day 0 and onwards. Embryonic testis development may be delayed in these mice; however, the postnatal data indicate that VEGFA isoforms secreted from Sertoli, Leydig or germ cells are not required for testis morphogenesis within the mouse. A Vegfa signal transduction array was employed on postnatal testes from Sry-Cre;Vegfa(-/-) versus controls. Ptgs1 (Cox1) was the only upregulated gene (fivefold). COX1 stimulates angiogenesis and upregulates, VEGFA, Prostaglandin E2 (PGE2) and PGD2. Thus, other gene pathways may compensate for VEGFA loss, similar to multiple independent mechanisms to maintain SOX9 expression. Multiple independent mechanism that induce vascular development in the testis may contribute to and safeguard the sex-specific vasculature development responsible for inducing seminiferous cord formation, thus ensuring appropriate testis morphogenesis in the male.
Collapse
Affiliation(s)
- Kevin M Sargent
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| | - Renee M McFee
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| | - Renata Spuri Gomes
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| | - Andrea S Cupp
- Department of Animal ScienceUniversity of Nebraska-Lincoln, Animal Science Building, 3940 Fair Street, Lincoln, Nebraska 68583-0908, USA
| |
Collapse
|
21
|
Rao SR, Shelton SE, Dayton PA. The "Fingerprint" of Cancer Extends Beyond Solid Tumor Boundaries: Assessment With a Novel Ultrasound Imaging Approach. IEEE Trans Biomed Eng 2015; 63:1082-6. [PMID: 26394410 DOI: 10.1109/tbme.2015.2479590] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
GOAL Abnormalities of microvascular morphology have been associated with tumor angiogenesis for more than a decade, and are believed to be intimately related to both tumor malignancy and response to treatment. However, the study of these vascular changes in-vivo has been challenged due to the lack of imaging approaches which can assess the microvasculature in 3-D volumes noninvasively. Here, we use contrast-enhanced "acoustic angiography" ultrasound imaging to observe and quantify heterogeneity in vascular morphology around solid tumors. METHODS Acoustic angiography, a recent advance in contrast-enhanced ultrasound imaging, generates high-resolution microvascular images unlike anything possible with standard ultrasound imaging techniques. Acoustic angiography images of a genetically engineered mouse breast cancer model were acquired to develop an image acquisition and processing routine that isolated radially expanding regions of a 3-D image from the tumor boundary to the edge of the imaging field for assessment of vascular morphology of tumor and surrounding vessels. RESULTS Quantitative analysis of vessel tortuosity for the tissue surrounding tumors 3 to 7 mm in diameter revealed that tortuosity decreased in a region 6 to 10 mm from the tumor boundary, but was still significantly elevated when compared to control vasculature. CONCLUSION Our analysis of angiogenesis-induced changes in the vasculature outside the tumor margin reveals that the extent of abnormal tortuosity extends significantly beyond the primary tumor mass. SIGNIFICANCE Visualization of abnormal vascular tortuosity may make acoustic angiography an invaluable tool for early tumor detection based on quantifying the vascular footprint of small tumors and a sensitive method for understanding changes in the vascular microenvironment during tumor progression.
Collapse
|
22
|
MDA-MB-231 breast cancer cells overexpressing single VEGF isoforms display distinct colonisation characteristics. Br J Cancer 2015. [PMID: 26196186 PMCID: PMC4559830 DOI: 10.1038/bjc.2015.267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background: Vascular endothelial growth factor (VEGF) is a multifunctional cytokine that has important roles in angiogenesis. Our knowledge of the significance of VEGF isoforms in human cancer remains incomplete. Methods: Bioluminescence imaging and transcriptomic analysis were used to study the colonisation capacity of the human breast cancer cells MDA-MB-231 controlling or overexpressing the VEGF165 or VEGF189 isoform (named cV-B, V165-B and V189-B, respectively) in nude mice. Results: When injected into the bloodstream, V189-B cells induced less metastasis in the lungs and bone than V165-B and cV-B control cells, consistent with longer survival of these mice and delay in tumour uptake in the mice injected with a V189-B clone. Histological analysis confirmed that there were less αSMA-positive cells in the lungs of the mice injected with V189-B. In vitro V189-B cells decreased both cell invasion and survival. Using transcriptomic analysis, we identified a subset of 18 genes expressed differentially between V189 and V165 cell lines and in 120 human breast tumours. V165 was associated with poor prognosis, whereas V189 was not, suggesting a complex regulation by VEGF isoforms. Our results showed a negative correlation between the expression pattern of VEGF189 and the levels of expression of seven genes that influence metastasis. Conclusion: Our findings provide the first evidence that VEGF isoforms have different effects on breast cancer cell line colonisation in vivo.
Collapse
|
23
|
Clegg LW, Mac Gabhann F. Site-Specific Phosphorylation of VEGFR2 Is Mediated by Receptor Trafficking: Insights from a Computational Model. PLoS Comput Biol 2015; 11:e1004158. [PMID: 26067165 PMCID: PMC4466579 DOI: 10.1371/journal.pcbi.1004158] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/25/2015] [Indexed: 02/05/2023] Open
Abstract
Matrix-binding isoforms and non-matrix-binding isoforms of vascular endothelial growth factor (VEGF) are both capable of stimulating vascular remodeling, but the resulting blood vessel networks are structurally and functionally different. Here, we develop and validate a computational model of the binding of soluble and immobilized ligands to VEGF receptor 2 (VEGFR2), the endosomal trafficking of VEGFR2, and site-specific VEGFR2 tyrosine phosphorylation to study differences in induced signaling between these VEGF isoforms. In capturing essential features of VEGFR2 signaling and trafficking, our model suggests that VEGFR2 trafficking parameters are largely consistent across multiple endothelial cell lines. Simulations demonstrate distinct localization of VEGFR2 phosphorylated on Y1175 and Y1214. This is the first model to clearly show that differences in site-specific VEGFR2 activation when stimulated with immobilized VEGF compared to soluble VEGF can be accounted for by altered trafficking of VEGFR2 without an intrinsic difference in receptor activation. The model predicts that Neuropilin-1 can induce differences in the surface-to-internal distribution of VEGFR2. Simulations also show that ligated VEGFR2 and phosphorylated VEGFR2 levels diverge over time following stimulation. Using this model, we identify multiple key levers that alter how VEGF binding to VEGFR2 results in different coordinated patterns of multiple downstream signaling pathways. Specifically, simulations predict that VEGF immobilization, interactions with Neuropilin-1, perturbations of VEGFR2 trafficking, and changes in expression or activity of phosphatases acting on VEGFR2 all affect the magnitude, duration, and relative strength of VEGFR2 phosphorylation on tyrosines 1175 and 1214, and they do so predictably within our single consistent model framework. Vascular endothelial growth factor (VEGF) is an important regulator of blood vessel growth. To date, therapies attempting to harness the VEGF system to promote blood vessel growth (e.g. for wound healing or ischemic disease) have achieved only limited success. To improve VEGF-based therapies, we need to better understand how VEGF promotes development of functional blood vessels. We have developed a computational model of VEGF binding to the receptor VEGFR2, trafficking of VEGFR2 through endosomal compartments in the cell, and activation of VEGFR2 on several tyrosine residues. The pattern of tyrosines activated on VEGFR2 influences cell behavior, promoting cell survival, proliferation, or migration. The combination of these cues influences the diameter of vessels, degree of branching, and leakiness of the resultant vessel network. Our model shows that changes in VEGFR2 trafficking as a result of VEGF immobilization to the extracellular matrix are sufficient to describe observed changes in the pattern of VEGFR2 activation compared to stimulation with purely soluble VEGF. This model can be used to predict how VEGF immobilization, interactions with co-receptors or proteins that deactivate VEGFR2, and changes to VEGFR2 trafficking can be tuned to promote development of functional blood vessel networks for tissue engineering applications.
Collapse
Affiliation(s)
- Lindsay Wendel Clegg
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| | - Feilim Mac Gabhann
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
24
|
Kantari-Mimoun C, Castells M, Klose R, Meinecke AK, Lemberger UJ, Rautou PE, Pinot-Roussel H, Badoual C, Schrödter K, Österreicher CH, Fandrey J, Stockmann C. Resolution of liver fibrosis requires myeloid cell-driven sinusoidal angiogenesis. Hepatology 2015; 61:2042-55. [PMID: 25475053 DOI: 10.1002/hep.27635] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 12/01/2014] [Indexed: 12/12/2022]
Abstract
UNLABELLED Angiogenesis is a key feature of liver fibrosis. Although sinusoidal remodeling is believed to contribute to fibrogenesis, the impact of sinusoidal angiogenesis on the resolution of liver fibrosis remains undefined. Myeloid cells, particularly macrophages, constantly infiltrate the fibrotic liver and can profoundly contribute to remodeling of liver sinusoids. We observe that the development of fibrosis is associated with decreased hepatic vascular endothelial growth factor (VEGF) expression as well as sinusoidal rarefication of the fibrotic scar. In contrast, the resolution of fibrosis is characterized by a rise in hepatic VEGF levels and revascularization of the fibrotic tissue. Genetic ablation of VEGF in myeloid cells or pharmacological inhibition of VEGF receptor 2 signaling prevents this angiogenic response and the resolution of liver fibrosis. We observe increased expression of matrix metalloproteases as well as decreased expression of tissue inhibitor of metalloproteases confined to sinusoidal endothelial cells in response to myeloid cell VEGF. Remarkably, reintroduction of myeloid cell-derived VEGF upon recovery restores collagenolytic acitivity and the resolution of fibrosis. CONCLUSION We identify myeloid cell-derived VEGF as a critical regulator of extracellular matrix degradation by liver endothelial cells, thereby unmasking an unanticipated link between angiogenesis and the resolution of fibrosis.
Collapse
Affiliation(s)
- Chahrazade Kantari-Mimoun
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France
| | - Magali Castells
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France
| | - Ralph Klose
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France
| | - Anna-Katharina Meinecke
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Duisburg, Germany
| | - Ursula J Lemberger
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Pierre-Emmanuel Rautou
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.,DHU Unity, Pôle des Maladies de l'Appareil Digestif, Service d'Hépatologie, Centre de Référence des Maladies Vasculaires du Foie, Hôpital Beaujon, AP-HP, Clichy, France
| | - Hélène Pinot-Roussel
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.,Service d'Anatomie et Pathologie, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Cécile Badoual
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.,Service d'Anatomie et Pathologie, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Katrin Schrödter
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Duisburg, Germany
| | - Christoph H Österreicher
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Joachim Fandrey
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Duisburg, Germany
| | - Christian Stockmann
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
25
|
Klegerman ME, Naji AK, Haworth KJ, Zou Y, Golunski E, Peng T, Britton GL, Huang SL, Holland CK, McPherson DD. Ultrasound-enhanced bevacizumab release from echogenic liposomes for inhibition of atheroma progression. J Liposome Res 2015; 26:47-56. [PMID: 25865025 DOI: 10.3109/08982104.2015.1029494] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
CONTEXT Bevacizumab (BEV) is a monoclonal antibody to vascular endothelial growth factor (VEGF) that ameliorates atheroma progression by inhibiting neovascularization. OBJECTIVE We aimed to determine whether BEV release from echogenic liposomes (BEV-ELIP) could be enhanced by color Doppler ultrasound (US) and whether the released BEV inhibits VEGF expression by endothelial cells in vitro. MATERIALS AND METHODS BEV-ELIP samples were subjected to 6 MHz color Doppler ultrasound (MI = 0.4) for 5 min. We assessed release of BEV with a direct ELISA and with fluoresceinated BEV (FITC-BEV) loaded into ELIP by the same method. Human umbilical vein endothelial cell (HUVEC) cultures were stimulated to express VEGF by 10 nM phorbol-12-myristate 13-acetate (PMA). Cell-associated VEGF levels were determined using a cell-based ELISA. RESULTS Overall, US caused an additional 100 µg of BEV to be released or exposed per BEV-ELIP aliquot within 60 min BEV-ELIP treated with US inhibited VEGF expression by 90% relative to non-treated controls and by 70% relative to BEV-ELIP without US. Also, US-treated BEV-ELIP inhibited HUVEC proliferation by 64% relative to untreated controls and by 45% relative to BEV-ELIP without US. DISCUSSION AND CONCLUSION We have demonstrated that BEV-ELIP retains its VEGF-binding activity in a liposomal formulation and that clinical Doppler US can significantly increase that activity, both by releasing free BEV and by enhancing the surface exposure of the immunoreactive antibody.
Collapse
Affiliation(s)
- Melvin E Klegerman
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| | - Ali K Naji
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| | - Kevin J Haworth
- b Department of Internal Medicine, Division of Cardiovascular Diseases , University of Cincinnati , Cincinnati , OH , USA , and.,c Biomedical Engineering Program , University of Cincinnati , Cincinnati , OH , USA
| | - Yuejiao Zou
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| | - Eva Golunski
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| | - Tao Peng
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| | - George L Britton
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| | - Shao-Ling Huang
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| | - Christy K Holland
- b Department of Internal Medicine, Division of Cardiovascular Diseases , University of Cincinnati , Cincinnati , OH , USA , and.,c Biomedical Engineering Program , University of Cincinnati , Cincinnati , OH , USA
| | - David D McPherson
- a Department of Internal Medicine, Division of Cardiovascular Medicine , University of Texas Health Science Center at Houston , Houston , TX , USA
| |
Collapse
|
26
|
Zhang H, Chen Y, Fan B, Wang W, Zhu W. Overexpression of VEGF183 promotes murine breast cancer cell proliferation in vitro and induces dilated intratumoral microvessels. Tumour Biol 2015; 36:3871-80. [PMID: 25577246 DOI: 10.1007/s13277-014-3029-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 12/30/2014] [Indexed: 02/04/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) was considered as a critical growth factor for tumor expansion. The roles of VEGF121, VEGF165, and VEGF189 in tumor growth have been intensely investigated; however, involvements of another extracellular matrix (ECM)-binding VEGF isoform, namely VEGF183 (six amino acids shorter than VEGF189 in exon 6a), in physiological or pathological processes are still unclear although the wide tissue distribution. To investigate the role of VEGF183 in carcinogenesis, we generated murine breast cancer cell (EMT-6) clones stably overexpressing VEGF183, VEGF121, VEGF165, and VEGF189 shortened as V183, V121, V165, and V189, respectively. Methylthiazolyldiphenyl-tetrazolium bromide (MTT) results showed that VEGF183, like all other VEGF-overexpressing isoforms except for VEGF121, could enhance the proliferation of mouse breast cancer EMT-6 cells. Immunochemistry results displayed that overexpressing VEGF183 and VEGF189 in EMT-6 cells induced larger proportional dilated microvessels. On the other hand, results from cell wound healing experiments demonstrated that all of the VEGF-overexpressing isoforms could increase the chemotaxis of EMT-6 cells in vitro. In conclusion, our results supported the idea that overexpression of VEGF183 promotes murine breast cancer cell proliferation in vitro and induces dilated intratumoral microvessels, and it plays a dissimilar role in comparison with that of VEGF189.
Collapse
Affiliation(s)
- Huiyong Zhang
- College of Life Science and Biotechnology, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | | | | | | | | |
Collapse
|
27
|
Salton M, Voss TC, Misteli T. Identification by high-throughput imaging of the histone methyltransferase EHMT2 as an epigenetic regulator of VEGFA alternative splicing. Nucleic Acids Res 2014; 42:13662-73. [PMID: 25414343 PMCID: PMC4267647 DOI: 10.1093/nar/gku1226] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recent evidence points to a role of chromatin in regulation of alternative pre-mRNA splicing (AS). In order to identify novel chromatin regulators of AS, we screened an RNAi library of chromatin proteins using a cell-based high-throughput in vivo assay. We identified a set of chromatin proteins that regulate AS. Using simultaneous genome-wide expression and AS analysis, we demonstrate distinct and non-overlapping functions of these chromatin modifiers on transcription and AS. Detailed mechanistic characterization of one dual function chromatin modifier, the H3K9 methyltransferase EHMT2 (G9a), identified VEGFA as a major chromatin-mediated AS target. Silencing of EHMT2, or its heterodimer partner EHMT1, affects AS by promoting exclusion of VEGFA exon 6a, but does not alter total VEGFA mRNA levels. The epigenetic regulatory mechanism of AS by EHMT2 involves an adaptor system consisting of the chromatin modulator HP1γ, which binds methylated H3K9 and recruits splicing regulator SRSF1. The epigenetic regulation of VEGFA is physiologically relevant since EHMT2 is transcriptionally induced in response to hypoxia and triggers concomitant changes in AS of VEGFA. These results characterize a novel epigenetic regulatory mechanism of AS and they demonstrate separate roles of epigenetic modifiers in transcription and alternative splicing.
Collapse
Affiliation(s)
- Maayan Salton
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Ty C Voss
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Tom Misteli
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Zhang HY, Fan BL, Wu XS, Mu LM, Wang WF, Zhu WL. Overexpression of the chimeric plasmin-resistant VEGF165/VEGF183 (132-158) protein in murine breast cancer induces distinct vascular patterning adjacent to tumors and retarded tumor growth. Mol Med Rep 2014; 11:1483-9. [PMID: 25373557 DOI: 10.3892/mmr.2014.2866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 08/19/2014] [Indexed: 11/05/2022] Open
Abstract
A chimeric plasmin‑resistant vascular endothelial growth factor (VEGF)165/VEGF183 (132-158) protein, named as VEGF183 (according to the nomenclature of VEGF), designed by a previous study, was demonstrated to have an enhanced affinity for the extracellular matrix (ECM) amongst other bioactivities. However, it is now accepted that mutant VEGFs frequently demonstrate different angiogenic activities and produce different vascular patterning from the parental molecule. The present study hypothesized that VEGF183, due to its enhanced binding affinity to the ECM, would exhibit a different angiogenic activity and produce a different vascular patterning compared to those of VEGF165. Murine breast cancer EMT‑6 cells were manipulated to stably overexpress VEGF165 or VEGF183. These cells were then inoculated intradermally into BALB/c mice in order to monitor the formation of vascular patterning in skin proximal to tumors. In vivo angiogenesis experiments revealed that overexpression of VEGF183 in murine breast cancer cells resulted in irregular, disorganized and dense vascular patterning as well as induced a significant inhibition of tumor growth compared with that of VEGF165. In addition, allograft tumor immunochemical assays of VEGF183‑overexpressing tumors demonstrated significantly lower vascular densities than those of VEGF165‑overexpressing tumors; however, VEGF183 tumors had a significantly enlarged vascular caliber. Conversely, cell wound healing experiments revealed that VEGF183‑overexpressing EMT‑6 cells had significantly decreased migration rates compared with those of VEGF165‑overexpressing EMT‑6 cells. In conclusion, the results of the present study supported the hypothesis that the altered ECM affinity of VEGF induced structural alterations to vasculature. In addition, these results provided a novel insight into VEGF design and indirect evidence for the function of exon 8 in VEGF. [Corrected]
Collapse
Affiliation(s)
- Hui-Yong Zhang
- College of Life Science and Biotechnology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Bing-Lin Fan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xin-Sheng Wu
- Department of Vasculocardiology, Xinxiang 371 Central Hospital, Xinxiang, Henan 453003, P.R. China
| | - Ling-Min Mu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Wen-Feng Wang
- College of Life Science and Biotechnology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Wu-Ling Zhu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
29
|
Vascular endothelial growth factor: therapeutic possibilities and challenges for the treatment of ischemia. Cytokine 2014; 71:385-93. [PMID: 25240960 DOI: 10.1016/j.cyto.2014.08.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/02/2014] [Accepted: 08/19/2014] [Indexed: 12/16/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a notable chemokine that plays critical roles in angiogenesis and vasculogenesis. The contemporary body of literature contains a substantial amount of information regarding its chemical properties as well as its fundamental role in vascular development. Studies strongly indicate its potential use as a therapeutic agent, especially in the vascular restoration of injured and ischemic tissues. VEGF therapy could be most beneficial for diseases whose pathologies revolve around tissue inflammation and necrosis, such as myocardial infarction and stroke, as well as ischemic bowel diseases such as acute mesenteric ischemia and necrotizing enterocolitis. However, a delicate balance exists between the therapeutic benefits of VEGF and the hazards of tumor growth and neo-angiogenesis. Effective future research surrounding VEGF may allow for the development of effective therapies for ischemia which simultaneously limit its more deleterious side effects. This review will: (1) summarize the current understanding of the molecular aspects and function of VEGF, (2) review potential benefits of its use in medical therapy, (3) denote its role in tumorigenesis and inflammation when overexpressed, and (4) elucidate the qualities which make it a viable compound of study for diagnostic and therapeutic applications.
Collapse
|
30
|
Marsters P, Alhamdan R, Campbell BK. Cell density-mediated pericellular hypoxia and the local dynamic regulation of VEGF-a splice variants in ovine ovarian granulosa cells. Biol Reprod 2014; 91:35. [PMID: 24966396 DOI: 10.1095/biolreprod.113.113068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The primary aims of this study were to utilize a specialized culture system to further elucidate the functional significance of pericellular hypoxia within the granulosa cell (GC) compartment of growing follicles, and to clarify its effects on the production of vascular endothelial growth factor (VEGF)-A isoforms and steroid hormones. Multilaminar clusters formed rapidly in ovine GCs seeded at high density (HD), and Hypoxyprobe-1 protein adducts appeared markedly more abundant and HIF-1 activation significantly (P < 0.001) greater than in cells seeded at low density (LD). Four proangiogenic VEGF mRNA transcript variants were identified in cultured GCs. Most abundant were VEGF120 and VEGF164, but VEGF182 and VEGF188 were also detected. Total VEGF mRNA was shown to be up-regulated transiently in the HD cells (P < 0.001) and VEGF164 mRNA appeared to contribute most to this. The hypoxia mimetic cobalt chloride also induced marked increases in HIF-1 activation (P < 0.01) and total VEGF mRNA (P < 0.01) production. HD cells increased levels of HIF-1alpha (P < 0.001) and VEGF receptor type 1 (P < 0.05), but not VEGF receptor type 2 mRNA, compared to LD cells or cells grown under chemically induced hypoxia. Both 17beta-estradiol (E2) and progesterone (P4) were markedly lower (P < 0.001) in the HD, cells but though cobalt chloride treatment accompanied significantly reduced P4 production (P < 0.05), E2 levels remained similar to those in untreated cells. These outcomes suggest that pericellular hypoxia may be an important mediator of VEGF production in the GCs of growing follicles, but that local regulation is complex and may involve multiple mechanisms such as mediation by steroid hormones and differential variant mRNA production.
Collapse
Affiliation(s)
- Peter Marsters
- Division of Human Development, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Rana Alhamdan
- Division of Human Development, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Bruce K Campbell
- Division of Human Development, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| |
Collapse
|
31
|
Esquibies AE, Karihaloo A, Quaggin SE, Bazzy-Asaad A, Cantley LG. Heparin binding VEGF isoforms attenuate hyperoxic embryonic lung growth retardation via a FLK1-neuropilin-1-PKC dependent pathway. Respir Res 2014; 15:32. [PMID: 24641672 PMCID: PMC4004166 DOI: 10.1186/1465-9921-15-32] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 03/14/2014] [Indexed: 12/14/2022] Open
Abstract
Background Previous work in our laboratory demonstrated that hyperoxia suppressed the expression of vascular endothelial growth factor (VEGF) by the embryonic lung, leading to increased epithelial cell apoptosis and failure of explant airway growth and branching that was rescued by the addition of Vegf165. The aims of this study were to determine protective pathways by which VEGF isoforms attenuate hyperoxic lung growth retardation and to identify the target cell for VEGF action. Methods Timed pregnant CD-1 or fetal liver kinase (FLK1)-eGFP lung explants cultured in 3% or 50% oxygen were treated ± Vegf121, VEGF164/Vegf165 or VEGF188 in the presence or absence of anti-rat neuropilin-1 (NRP1) antibody or GO6983 (protein kinase C (PKC) pan-inhibitor) and lung growth and branching quantified. Immunofluorescence studies were performed to determine apoptosis index and location of FLK1 phosphorylation and western blot studies of lung explants were performed to define the signaling pathways that mediate the protective effects of VEGF. Results Heparin-binding VEGF isoforms (VEGF164/Vegf165 and VEGF188) but not Vegf121 selectively reduced epithelial apoptosis and partially rescued lung bud branching and growth. These protective effects required NRP1-dependent FLK1 activation in endothelial cells. Analysis of downstream signaling pathways demonstrated that the VEGF-mediated anti-apoptotic effects were dependent on PKC activation. Conclusions Vegf165 activates FLK1-NRP1 signaling in endothelial cells, leading to a PKC-dependent paracrine signal that in turn inhibits epithelial cell apoptosis.
Collapse
Affiliation(s)
- Americo E Esquibies
- Department of Pediatrics Section of Respiratory Medicine, Yale University School of Medicine, 333 Cedar Street, P,O, Box 208064, New Haven, CT 06520, USA.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Pericytes are interstitial mesenchymal cells found in many major organs. In the kidney, microvascular pericytes are defined anatomically as extensively branched, collagen-producing cells in close contact with endothelial cells. Although many molecular markers have been proposed, none of them can identify the pericytes with satisfactory specificity or sensitivity. The roles of microvascular pericytes in kidneys were poorly understood in the past. Recently, by using genetic lineage tracing to label collagen-producing cells or mesenchymal cells, the elusive characteristics of the pericytes have been illuminated. The purpose of this article is to review recent advances in the understanding of microvascular pericytes in the kidneys. In healthy kidney, the pericytes are found to take part in the maintenance of microvascular stability. Detachment of the pericytes from the microvasculature and loss of the close contact with endothelial cells have been observed during renal insult. Renal microvascular pericytes have been shown to be the major source of scar-forming myofibroblasts in fibrogenic kidney disease. Targeting the crosstalk between pericytes and neighboring endothelial cells or tubular epithelial cells may inhibit the pericyte-myofibroblast transition, prevent peritubular capillary rarefaction, and attenuate renal fibrosis. In addition, renal pericytes deserve attention for their potential to produce erythropoietin in healthy kidneys as pericytes stand in the front line, sensing the change of oxygenation and hemoglobin concentration. Further delineation of the mechanisms underlying the reduced erythropoietin production occurring during pericyte-myofibroblast transition may be promising for the development of new treatment strategies for anemia in chronic kidney disease.
Collapse
Affiliation(s)
- Szu-Yu Pan
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan ; Department of Internal Medicine, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, Taiwan
| | - Yu-Ting Chang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shuei-Liong Lin
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan ; Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
33
|
Zhang B, Xiao W, Qiu H, Zhang F, Moniz HA, Jaworski A, Condac E, Gutierrez-Sanchez G, Heiss C, Clugston RD, Azadi P, Greer JJ, Bergmann C, Moremen KW, Li D, Linhardt RJ, Esko JD, Wang L. Heparan sulfate deficiency disrupts developmental angiogenesis and causes congenital diaphragmatic hernia. J Clin Invest 2013; 124:209-21. [PMID: 24355925 DOI: 10.1172/jci71090] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 10/11/2013] [Indexed: 12/12/2022] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a common birth malformation with a heterogeneous etiology. In this study, we report that ablation of the heparan sulfate biosynthetic enzyme NDST1 in murine endothelium (Ndst1ECKO mice) disrupted vascular development in the diaphragm, which led to hypoxia as well as subsequent diaphragm hypoplasia and CDH. Intriguingly, the phenotypes displayed in Ndst1ECKO mice resembled the developmental defects observed in slit homolog 3 (Slit3) knockout mice. Furthermore, introduction of a heterozygous mutation in roundabout homolog 4 (Robo4), the gene encoding the cognate receptor of SLIT3, aggravated the defect in vascular development in the diaphragm and CDH. NDST1 deficiency diminished SLIT3, but not ROBO4, binding to endothelial heparan sulfate and attenuated EC migration and in vivo neovascularization normally elicited by SLIT3-ROBO4 signaling. Together, these data suggest that heparan sulfate presentation of SLIT3 to ROBO4 facilitates initiation of this signaling cascade. Thus, our results demonstrate that loss of NDST1 causes defective diaphragm vascular development and CDH and that heparan sulfate facilitates angiogenic SLIT3-ROBO4 signaling during vascular development.
Collapse
|
34
|
Akerman S, Fisher M, Daniel RA, Lefley D, Reyes-Aldasoro CC, Lunt SJ, Harris S, Bjorndahl M, Williams LJ, Evans H, Barber PR, Prise VE, Vojnovic B, Kanthou C, Tozer GM. Influence of soluble or matrix-bound isoforms of vascular endothelial growth factor-A on tumor response to vascular-targeted strategies. Int J Cancer 2013; 133:2563-76. [PMID: 23712501 DOI: 10.1002/ijc.28281] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 03/31/2013] [Accepted: 04/05/2013] [Indexed: 12/12/2022]
Abstract
Antiangiogenic therapy based on blocking the actions of vascular endothelial growth factor-A (VEGF) can lead to "normalization" of blood vessels in both animal and human tumors. Differential expression of VEGF isoforms affects tumor vascular maturity, which could influence the normalization process and response to subsequent treatment. Fibrosarcoma cells expressing only VEGF120 or VEGF188 isoforms were implanted either subcutaneously (s.c.) or in dorsal skin-fold "window" chambers in SCID mice. VEGF120 was associated with vascular fragility and hemorrhage. Tumor-bearing mice were treated with repeat doses of SU5416, an indolinone receptor tyrosine kinase inhibitor with activity against VEGFR-2 and proven preclinical ability to induce tumor vascular normalization. SU5416 reduced vascularization in s.c. implants of both VEGF120 and VEGF188 tumors. However, in the window chamber, SU5416 treatment increased red cell velocity in VEGF120 (representing vascular normalization) but not VEGF188 tumors. SU5416 treatment had no effect on growth or necrosis levels in either tumor type but tended to counteract the increase in interstitial fluid pressure seen with growth of VEGF120 tumors. SU5416 pretreatment resulted in the normally fragile blood vessels in VEGF120-expressing tumors becoming resistant to the vascular damaging effects of the tubulin-binding vascular disrupting agent (VDA), combretastatin A4 3-O-phosphate (CA4P). Thus, vascular normalization induced by antiangiogenic treatment can reduce the efficacy of subsequent VDA treatment. Expression of VEGF120 made tumors particularly susceptible to vascular normalization by SU5416, which in turn made them resistant to CA4P. Therefore, VEGF isoform expression may be useful for predicting response to both antiangiogenic and vascular-disrupting therapy.
Collapse
Affiliation(s)
- Simon Akerman
- Tumor Microcirculation Group, CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, Department of Oncology, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Vempati P, Popel AS, Mac Gabhann F. Extracellular regulation of VEGF: isoforms, proteolysis, and vascular patterning. Cytokine Growth Factor Rev 2013; 25:1-19. [PMID: 24332926 DOI: 10.1016/j.cytogfr.2013.11.002] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/14/2013] [Accepted: 11/19/2013] [Indexed: 12/15/2022]
Abstract
The regulation of vascular endothelial growth factor A (VEGF) is critical to neovascularization in numerous tissues under physiological and pathological conditions. VEGF has multiple isoforms, created by alternative splicing or proteolytic cleavage, and characterized by different receptor-binding and matrix-binding properties. These isoforms are known to give rise to a spectrum of angiogenesis patterns marked by differences in branching, which has functional implications for tissues. In this review, we detail the extensive extracellular regulation of VEGF and the ability of VEGF to dictate the vascular phenotype. We explore the role of VEGF-releasing proteases and soluble carrier molecules on VEGF activity. While proteases such as MMP9 can 'release' matrix-bound VEGF and promote angiogenesis, for example as a key step in carcinogenesis, proteases can also suppress VEGF's angiogenic effects. We explore what dictates pro- or anti-angiogenic behavior. We also seek to understand the phenomenon of VEGF gradient formation. Strong VEGF gradients are thought to be due to decreased rates of diffusion from reversible matrix binding, however theoretical studies show that this scenario cannot give rise to lasting VEGF gradients in vivo. We propose that gradients are formed through degradation of sequestered VEGF. Finally, we review how different aspects of the VEGF signal, such as its concentration, gradient, matrix-binding, and NRP1-binding can differentially affect angiogenesis. We explore how this allows VEGF to regulate the formation of vascular networks across a spectrum of high to low branching densities, and from normal to pathological angiogenesis. A better understanding of the control of angiogenesis is necessary to improve upon limitations of current angiogenic therapies.
Collapse
Affiliation(s)
- Prakash Vempati
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Feilim Mac Gabhann
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
36
|
Arcondéguy T, Lacazette E, Millevoi S, Prats H, Touriol C. VEGF-A mRNA processing, stability and translation: a paradigm for intricate regulation of gene expression at the post-transcriptional level. Nucleic Acids Res 2013; 41:7997-8010. [PMID: 23851566 PMCID: PMC3783158 DOI: 10.1093/nar/gkt539] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Vascular Endothelial Growth Factor A (VEGF-A) is a potent secreted mitogen crucial for physiological and pathological angiogenesis. Post-transcriptional regulation of VEGF-A occurs at multiple levels. Firstly, alternative splicing gives rise to different transcript variants encoding diverse isoforms that exhibit distinct biological properties with regard to receptor binding and extra-cellular localization. Secondly, VEGF-A mRNA stability is regulated by effectors such as hypoxia or growth factors through the binding of stabilizing and destabilizing proteins at AU-rich elements located in the 3′-untranslated region. Thirdly, translation of VEGF-A mRNA is a controlled process involving alternative initiation codons, internal ribosome entry sites (IRESs), an upstream open reading frame (uORF), miRNA targeting and a riboswitch in the 3′ untranslated region. These different levels of regulation cooperate for the crucial fine-tuning of the expression of VEGF-A variants. This review will be focused on our current knowledge of the complex post-transcriptional regulatory switches that modulate the cellular VEGF-A level, a paradigmatic model of post-transcriptional regulation.
Collapse
Affiliation(s)
- Tania Arcondéguy
- Inserm UMR1037, Centre de Recherches en Cancérologie de Toulouse, CHU Rangueil, BP84225, 31432 Toulouse Cedex 4, France and Université Toulouse III Paul-Sabatier, 118 Route de Narbonne, 31400 Toulouse, France
| | | | | | | | | |
Collapse
|
37
|
Genetic and pharmacologic inhibition of complement impairs endothelial cell function and ablates ovarian cancer neovascularization. Neoplasia 2013; 14:994-1004. [PMID: 23226093 DOI: 10.1593/neo.121262] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/21/2012] [Accepted: 09/27/2012] [Indexed: 12/16/2022] Open
Abstract
Complement activation plays a critical role in controlling inflammatory responses. To assess the role of complement during ovarian cancer progression, we crossed two strains of mice with genetic complement deficiencies with transgenic mice that develop epithelial ovarian cancer (TgMISIIR-TAg). TgMISIIR-TAg mice fully or partially deficient for complement factor 3 (C3) (Tg(+)C3(KO) and Tg(+)C3(HET), respectively) or fully deficient for complement factor C5a receptor (C5aR) (Tg(+)C5aR(KO)) develop either no ovarian tumors or tumors that were small and poorly vascularized compared to wild-type littermates (Tg(+)C3(WT), Tg(+)C5aR(WT)). The percentage of tumor infiltrating immune cells in Tg(+)C3(HET) tumors compared to Tg(+)C3(WT) controls was either similar (macrophages, B cells, myeloid-derived suppressor cells), elevated (effector T cells), or decreased (regulatory T cells). Regardless of these ratios, cytokine production by immune cells taken from Tg(+)C3(HET) tumors was reduced on stimulation compared to Tg(+)C3(WT) controls. Interestingly, CD31(+) endothelial cell (EC) function in angiogenesis was significantly impaired in both C3(KO) and C5aR(KO) mice. Further, using the C5aR antagonist PMX53, tube formation of ECs was shown to be C5a-dependent, possibly through interactions with the VEGF(165) but not VEGF(121) isoform. Finally, the mouse VEGF(164) transcript was underexpressed in C3(KO) livers compare to C3(WT) livers. Thus, we conclude that complement inhibition blocks tumor outgrowth by altering EC function and VEGF(165) expression.
Collapse
|
38
|
Finley SD, Popel AS. Effect of tumor microenvironment on tumor VEGF during anti-VEGF treatment: systems biology predictions. J Natl Cancer Inst 2013; 105:802-11. [PMID: 23670728 DOI: 10.1093/jnci/djt093] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is known to be a potent promoter of angiogenesis under both physiological and pathological conditions. Given its role in regulating tumor vascularization, VEGF has been targeted in various cancer treatments, and anti-VEGF therapy has been used clinically for treatment of several types of cancer. Systems biology approaches, particularly computational models, provide insight into the complexity of tumor angiogenesis. These models complement experimental studies and aid in the development of effective therapies targeting angiogenesis. METHODS We developed an experiment-based, molecular-detailed compartment model of VEGF kinetics and transport to investigate the distribution of two major VEGF isoforms (VEGF121 and VEGF165) in the body. The model is applied to predict the dynamics of tumor VEGF and, importantly, to gain insight into how tumor VEGF responds to an intravenous injection of an anti-VEGF agent. RESULTS The model predicts that free VEGF in the tumor interstitium is seven to 13 times higher than plasma VEGF and is predominantly in the form of VEGF121 (>70%), predictions that are validated by experimental data. The model also predicts that tumor VEGF can increase or decrease with anti-VEGF treatment depending on tumor microenvironment, pointing to the importance of personalized medicine. CONCLUSIONS This computational study suggests that the rate of VEGF secretion by tumor cells may serve as a biomarker to predict the patient population that is likely to respond to anti-VEGF treatment. Thus, the model predictions have important clinical relevance and may aid clinicians and clinical researchers seeking interpretation of pharmacokinetic and pharmacodynamic observations and optimization of anti-VEGF therapies.
Collapse
Affiliation(s)
- Stacey D Finley
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
39
|
Kuo IC, Sambuelli RH, Bono J, Smith RJ, Reviglio VE. Progression of choroidal metastasis of ovarian serous cystoadenocarcinoma after intravitreal bevacizumab treatment. Rare Tumors 2013; 5:e5. [PMID: 23772304 PMCID: PMC3682457 DOI: 10.4081/rt.2013.e5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 12/24/2012] [Accepted: 01/14/2013] [Indexed: 12/24/2022] Open
Abstract
A 57-year-old woman presented to her ophthalmologist because of rapid deterioration in vision. Dilated funduscopic examination of the right eye showed an elevated, yellow-orange choroidal mass temporal to the fovea; a complete retinal detachment was present in the left eye. The patient was referred to an oncologist. Computerized tomography of the brain, thorax, abdomen, and pelvis were obtained. They revealed an 11-mm mass in the right parietal lobe, a 30-mm mass in the left temporal lobe, 23-mm mass in the right kidney, and multiple nodules in both lungs. Supported by published experience with intravitreal bevacizumab for choroidal metastasis, the patient was injected into the vitreous through the pars plana of the left eye. The tumor mass did not show signs of regression and the visual acuity was unchanged. The patient suffered from end-state complications tumor metastasis and expired one month after the invitreal injection.
Collapse
Affiliation(s)
- Irene C. Kuo
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruben H. Sambuelli
- Pathology Department, Cornea and Anterior Segment Research, Catholic University of Cordoba, School of Medicine, Cordoba, Argentina
| | - Javier Bono
- Ophthalmology Service, Cordoba Hospital, Cordoba, Argentina
| | - Ricardo J. Smith
- Instituto de la Visión Cerro de las Rosas, Sanatorio Allende, Córdoba, Argentina
| | - Victor E. Reviglio
- Pathology Department, Cornea and Anterior Segment Research, Catholic University of Cordoba, School of Medicine, Cordoba, Argentina
- Ophthalmology Service, Cordoba Hospital, Cordoba, Argentina
- Instituto de la Visión Cerro de las Rosas, Sanatorio Allende, Córdoba, Argentina
| |
Collapse
|
40
|
Siddiq I, Park E, Liu E, Spratt SK, Surosky R, Lee G, Ando D, Giedlin M, Hare GMT, Fehlings MG, Baker AJ. Treatment of traumatic brain injury using zinc-finger protein gene therapy targeting VEGF-A. J Neurotrauma 2012; 29:2647-59. [PMID: 23016562 DOI: 10.1089/neu.2012.2444] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) plays a role in angiogenesis and has been shown to be neuroprotective following central nervous system trauma. In the present study we evaluated the pro-angiogenic and neuroprotective effects of an engineered zinc-finger protein transcription factor transactivator targeting the vascular endothelial growth factor A (VEGF-ZFP). We used two virus delivery systems, adeno-virus and adeno-associated virus, to examine the effects of early and delayed VEGF-A upregulation after brain trauma, respectively. Male Sprague-Dawley rats were subject to a unilateral fluid percussion injury (FPI) of moderate severity (2.2-2.5 atm) followed by intracerebral microinjection of either adenovirus vector (Adv) or an adeno-associated vector (AAV) carrying the VEGF-ZFP construct. Adv-VEGF-ZFP-treated animals had significantly fewer TUNEL positive cells in the injured penumbra of the cortex (p<0.001) and hippocampus (p=0.001) relative to untreated rats at 72 h post-injury. Adv-VEGF-ZFP treatment significantly improved fEPSP values (p=0.007) in the CA1 region relative to injury alone. Treatment with AAV2-VEGF-ZFP resulted in improved post-injury microvascular diameter and improved functional recovery on the balance beam and rotarod task at 30 days post-injury. Collectively, the results provide supportive evidence for the concept of acute and delayed treatment following TBI using VEGF-ZFP to induce angiogenesis, reduce cell death, and enhance functional recovery.
Collapse
Affiliation(s)
- Ishita Siddiq
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chang FC, Chou YH, Chen YT, Lin SL. Novel insights into pericyte-myofibroblast transition and therapeutic targets in renal fibrosis. J Formos Med Assoc 2012; 111:589-98. [PMID: 23217594 DOI: 10.1016/j.jfma.2012.09.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 09/03/2012] [Accepted: 09/10/2012] [Indexed: 12/31/2022] Open
Abstract
Renal fibrosis is a disease affecting millions worldwide and is a harbinger of progressive renal failure. Understanding the mechanisms of renal fibrosis is important for discovering new therapies that are required to prevent loss of renal function. Recently, we identified pericytes that line the kidney microvasculature as the precursor cells of the scar-producing myofibroblasts during kidney injury. Kidney pericytes are extensively branched cells embedded within the capillary basement membrane and stabilize the capillary network through tissue inhibitor of metalloproteinase 3 and angiogenic growth factors. Pericytes detach from endothelial cells and migrate into the interstitial space where they undergo a transition into myofibroblasts after injury. Activation of endothelium, pericyte-myofibroblast transition, and recruitment of inflammatory macrophages lead to capillary rarefaction and fibrosis. Targeting endothelium-pericyte crosstalk by inhibiting vascular endothelial cell growth factor receptors and platelet-derived growth factor receptors in response to injury have been identified as new therapeutic interventions. Furthermore, targeting macrophage activation has also been proven as a novel and safe therapeutic approach for pericyte-myofibroblast transition. However, we are still far from understanding the interaction between pericytes and other cellular elements in normal physiology and during kidney fibrosis. Further studies will be required to translate into more specific therapeutic approaches.
Collapse
Affiliation(s)
- Fan-Chi Chang
- Renal Division, Department of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | |
Collapse
|
42
|
Seguin F, Carvalho MA, Bastos DC, Agostini M, Zecchin KG, Alvarez-Flores MP, Chudzinski-Tavassi AM, Coletta RD, Graner E. The fatty acid synthase inhibitor orlistat reduces experimental metastases and angiogenesis in B16-F10 melanomas. Br J Cancer 2012; 107:977-87. [PMID: 22892389 PMCID: PMC3464771 DOI: 10.1038/bjc.2012.355] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/12/2012] [Accepted: 07/17/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Fatty acid synthase (FASN) is overexpressed and associated with poor prognosis in several human cancers. Here, we investigate the effect of FASN inhibitors on the metastatic spread and angiogenesis in experimental melanomas and cultured melanoma cells. METHODS The lung colonisation assay and cutaneous melanomas were performed by the inoculation of mouse melanoma B16-F10 cells in C57BL6 mice. Blood vessel endothelial cells (RAEC and HUVEC) were applied to determine cell proliferation, apoptosis, and the formation of capillary-like structures. Vascular endothelial growth factor A (VEGFA) expression was evaluated by quantitative RT-PCR and ELISA in B16-F10, human melanoma (SK-MEL-25), and human oral squamous carcinoma (SCC-9) cells. Conditioned media from these cancer cell lines were used to study the effects of FASN inhibitors on endothelial cells. RESULTS B16-F10 melanoma-induced metastases and angiogenesis were significantly reduced in orlistat-treated mice. Fatty acid synthase inhibitors reduced the viability, proliferation, and the formation of capillary-like structures by RAEC cells, as well as the tumour cell-mediated formation of HUVEC capillary-like structures. Cerulenin and orlistat stimulated the production of total VEGFA in B16-F10, SK-MEL-25, and SCC-9 cells. Both drugs also enhanced VEGFA(121), (165), (189,) and (165b) in SK-MEL-25 and SCC-9 cells. CONCLUSION FASN inhibitors reduce metastasis and tumour-induced angiogenesis in experimental melanomas, and differentially modulate VEGFA expression in B16-F10 cells.
Collapse
Affiliation(s)
- F Seguin
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (UNICAMP), Avenida Limeira 901, CP 52, Areão, Piracicaba, CEP 13414-018, SP, Brazil
| | - M A Carvalho
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (UNICAMP), Avenida Limeira 901, CP 52, Areão, Piracicaba, CEP 13414-018, SP, Brazil
| | - D C Bastos
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (UNICAMP), Avenida Limeira 901, CP 52, Areão, Piracicaba, CEP 13414-018, SP, Brazil
| | - M Agostini
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (UNICAMP), Avenida Limeira 901, CP 52, Areão, Piracicaba, CEP 13414-018, SP, Brazil
| | - K G Zecchin
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (UNICAMP), Avenida Limeira 901, CP 52, Areão, Piracicaba, CEP 13414-018, SP, Brazil
| | - M P Alvarez-Flores
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Avenida Vital Brasil 1500, Butantã, São Paulo, CEP 05503-900, SP, Brazil
| | - A M Chudzinski-Tavassi
- Laboratory of Biochemistry and Biophysics, Butantan Institute, Avenida Vital Brasil 1500, Butantã, São Paulo, CEP 05503-900, SP, Brazil
| | - R D Coletta
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (UNICAMP), Avenida Limeira 901, CP 52, Areão, Piracicaba, CEP 13414-018, SP, Brazil
| | - E Graner
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (UNICAMP), Avenida Limeira 901, CP 52, Areão, Piracicaba, CEP 13414-018, SP, Brazil
| |
Collapse
|
43
|
Biomimetic hydrogels for controlled biomolecule delivery to augment bone regeneration. Adv Drug Deliv Rev 2012; 64:1078-89. [PMID: 22465487 DOI: 10.1016/j.addr.2012.03.010] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Revised: 02/12/2012] [Accepted: 03/05/2012] [Indexed: 11/21/2022]
Abstract
The regeneration of large bone defects caused by trauma or disease remains a significant clinical problem. Although osteoinductive growth factors such as bone morphogenetic proteins have entered clinics, transplantation of autologous bone remains the gold standard to treat bone defects. The effective treatment of bone defects by protein therapeutics in humans requires quantities that exceed the physiological doses by several orders of magnitude. This not only results in very high treatment costs but also bears considerable risks for adverse side effects. These issues have motivated the development of biomaterials technologies allowing to better control biomolecule delivery from the solid phase. Here we review recent approaches to immobilize biomolecules by affinity binding or by covalent grafting to biomaterial matrices. We focus on biomaterials concepts that are inspired by extracellular matrix (ECM) biology and in particular the dynamic interaction of growth factors with the ECM. We highlight the value of synthetic, ECM-mimicking matrices for future technologies to study bone biology and develop the next generation of 'smart' implants.
Collapse
|
44
|
Finley SD, Popel AS. Predicting the effects of anti-angiogenic agents targeting specific VEGF isoforms. AAPS JOURNAL 2012; 14:500-9. [PMID: 22547351 DOI: 10.1208/s12248-012-9363-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 04/13/2012] [Indexed: 01/04/2023]
Abstract
Vascular endothelial growth factor (VEGF) is a key mediator of angiogenesis, whose effect on cancer growth and development is well characterized. Alternative splicing of VEGF leads to several different isoforms, which are differentially expressed in various tumor types and have distinct functions in tumor blood vessel formation. Many cancer therapies aim to inhibit angiogenesis by targeting VEGF and preventing intracellular signaling leading to tumor vascularization; however, the effects of targeting specific VEGF isoforms have received little attention in the clinical setting. In this work, we investigate the effects of selectively targeting a single VEGF isoform, as compared with inhibiting all isoforms. We utilize a molecular-detailed whole-body compartment model of VEGF transport and kinetics in the presence of breast tumor. The model includes two major VEGF isoforms, VEGF(121) and VEGF(165), receptors VEGFR1 and VEGFR2, and co-receptors Neuropilin-1 and Neuropilin-2. We utilize the model to predict the concentrations of free VEGF, the number of VEGF/VEGFR2 complexes (considered to be pro-angiogenic), and the receptor occupancy profiles following inhibition of VEGF using isoform-specific anti-VEGF agents. We predict that targeting VEGF(121) leads to a 54% and 84% reduction in free VEGF in tumors that secrete both VEGF isoforms or tumors that overexpress VEGF(121), respectively. Additionally, 21% of the VEGFR2 molecules in the blood are ligated following inhibition of VEGF(121), compared with 88% when both isoforms are targeted. Targeting VEGF(121) reduces tumor free VEGF and is an effective treatment strategy. Our results provide a basis for clinical investigation of isoform-specific anti-VEGF agents.
Collapse
Affiliation(s)
- Stacey D Finley
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
45
|
Adluri RS, Thirunavukkarasu M, Zhan L, Dunna NR, Akita Y, Selvaraju V, Otani H, Sanchez JA, Ho YS, Maulik N. Glutaredoxin-1 overexpression enhances neovascularization and diminishes ventricular remodeling in chronic myocardial infarction. PLoS One 2012; 7:e34790. [PMID: 22523530 PMCID: PMC3327713 DOI: 10.1371/journal.pone.0034790] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 03/08/2012] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress plays a critical role in the pathophysiology of cardiac failure, including the modulation of neovascularization following myocardial infarction (MI). Redox molecules thioredoxin (Trx) and glutaredoxin (Grx) superfamilies actively maintain intracellular thiol-redox homeostasis by scavenging reactive oxygen species. Among these two superfamilies, the pro-angiogenic function of Trx-1 has been reported in chronic MI model whereas similar role of Grx-1 remains uncertain. The present study attempts to establish the role of Grx-1 in neovascularization and ventricular remodeling following MI. Wild-type (WT) and Grx-1 transgenic (Grx-1(Tg/+)) mice were randomized into wild-type sham (WTS), Grx-1(Tg/+) Sham (Grx-1(Tg/+)S), WTMI, Grx-1(Tg/+)MI. MI was induced by permanent occlusion of the LAD coronary artery. Sham groups underwent identical time-matched surgical procedures without LAD ligation. Significant increase in arteriolar density was observed 7 days (d) after surgical intervention in the Grx-1(Tg/+)MI group as compared to the WTMI animals. Further, improvement in myocardial functional parameters 30 d after MI was observed including decreased LVIDs, LVIDd, increased ejection fraction and, fractional shortening was also observed in the Grx-1(Tg/+)MI group as compared to the WTMI animals. Moreover, attenuation of oxidative stress and apoptotic cardiomyocytes was observed in the Grx-1(Tg/+)MI group as compared to the WTMI animals. Increased expression of p-Akt, VEGF, Ang-1, Bcl-2, survivin and DNA binding activity of NF-κB were observed in the Grx-1(Tg/+)MI group when compared to WTMI animals as revealed by Western blot analysis and Gel-shift analysis, respectively. These results are the first to demonstrate that Grx-1 induces angiogenesis and diminishes ventricular remodeling apparently through neovascularization mediated by Akt, VEGF, Ang-1 and NF-κB as well as Bcl-2 and survivin-mediated anti-apoptotic pathway in the infarcted myocardium.
Collapse
Affiliation(s)
- Ram Sudheer Adluri
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
| | - Mahesh Thirunavukkarasu
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
| | - Lijun Zhan
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
| | - Nageswara Rao Dunna
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
| | - Yuzo Akita
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
| | - Vaithinathan Selvaraju
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
| | - Hajime Otani
- Second Department of Internal Medicine, Kansai Medical University, Moriguchi, Japan
| | - Juan A. Sanchez
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Nilanjana Maulik
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, Health Center, University of Connecticut, Farmington, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
46
|
Development of a multiplexed PCR-coupled liquid bead array assay for vascular endothelial growth factor (VEGF) splice variants. Clin Biochem 2012; 45:475-82. [PMID: 22330939 DOI: 10.1016/j.clinbiochem.2012.01.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 01/24/2012] [Accepted: 01/26/2012] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To develop a multiplex PCR-coupled liquid bead array assay for the expression of VEGF splice variants. DESIGN AND METHODS The assay was based on the combination of multiplex PCR with liquid bead array technology, and optimized and evaluated in terms of analytical sensitivity, specificity, and reproducibility using the MCF-7 cell line. Clinical performance was evaluated in 16 pairs of fresh frozen cancerous and corresponding noncancerous adjacent tissues from NSCLC patients. RESULTS The assay is highly sensitive, reproducible and can detect specifically VEGF splice variants in clinical samples. When applied in 32 clinical samples it gave comparable results to RT-qPCR (concordance of 81%, 75%, 88% and 81% for PBGD, VEGF(121), VEGF(165), and VEGF(189) respectively). CONCLUSIONS The developed assay can simultaneously detect three VEGF splice variants with high specificity and sensitivity and can be further used to evaluate the role of each individual VEGF splice variant in molecular therapies targeted against VEGF.
Collapse
|
47
|
Balzer EM, Konstantopoulos K. Intercellular adhesion: mechanisms for growth and metastasis of epithelial cancers. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 4:171-81. [PMID: 21913338 DOI: 10.1002/wsbm.160] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cell-cell adhesion molecules (CAMs) comprise a broad class of linker proteins that are crucial for the development of multicellular organisms, and for the continued maintenance of organ and tissue structure. Because of its pivotal function in tissue homeostasis, the deregulation of intercellular adhesion is linked to the onset of most solid tumors. The breakdown of homeostatic cell adhesions in highly ordered epithelial sheets is directly implicated in carcinogenesis, while continued changes in the adhesion profile of the primary tumor mass facilitate growth and expansion into adjacent tissue. Intercellular adhesion molecules are also involved in each subsequent phase of metastasis, including transendothelial migration, transit through the bloodstream or lymphatics, and renewed proliferation in secondary sites. This review addresses various roles of cadherin- and selectin-mediated intercellular adhesion in tumor initiation and malignant transformation, and discusses the mechanisms for the arrest and adhesion of circulating tumor cells to the vessel endothelium. Considering the contributions of these CAMs to cancer progression in the context of a systematic biological framework may prove valuable in identifying new ways to diagnose and treat cancer.
Collapse
Affiliation(s)
- Eric M Balzer
- The Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
48
|
D'Onofrio PM, Thayapararajah M, Lysko MD, Magharious M, Spratt SK, Lee G, Ando D, Surosky R, Fehlings MG, Koeberle PD. Gene Therapy for Traumatic Central Nervous System Injury and Stroke Using an Engineered Zinc Finger Protein that Upregulates VEGF-A. J Neurotrauma 2011; 28:1863-79. [DOI: 10.1089/neu.2011.1896] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
| | | | - Meghan D. Lysko
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Mark Magharious
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Graduate Department of Rehabilitation Science, University of Toronto, Toronto, Ontario, Canada
| | - S. Kaye Spratt
- Department of Therapeutic Development, Sangamo Biosciences, Port Richmond, California
| | - Gary Lee
- Department of Therapeutic Development, Sangamo Biosciences, Port Richmond, California
| | - Dale Ando
- Department of Therapeutic Development, Sangamo Biosciences, Port Richmond, California
| | - Richard Surosky
- Department of Therapeutic Development, Sangamo Biosciences, Port Richmond, California
| | | | - Paulo D. Koeberle
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Graduate Department of Rehabilitation Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
49
|
Huang G, Zhou Z, Wang H, Kleinerman ES. CAPER-α alternative splicing regulates the expression of vascular endothelial growth factor₁₆₅ in Ewing sarcoma cells. Cancer 2011; 118:2106-16. [PMID: 22009261 DOI: 10.1002/cncr.26488] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/29/2011] [Accepted: 07/14/2011] [Indexed: 01/13/2023]
Abstract
BACKGROUND TC-71 Ewing sarcoma cells overexpress vascular endothelial growth factor (VEGF) with a shift from the 189 to the 165 isoform. METHODS The effect of CAPER-α on the expression of the VEGF isoforms, tumor growth, and vessel density was analyzed after transfection of TC-71 cells with CAPER-α cDNA or siRNA. RESULTS CAPER-α correlated inversely with the VEGF(165) /VEGF(189) mRNA ratio. Up-regulation of CAPER-α resulted in decreased tumor growth, tumor vessel density, and chemotactic activity of the cell's supernatant. CAPER-α expression was regulated by EWS/FLI-1 through a protein-protein interaction. CONCLUSIONS Increased VEGF(165) expression is secondary to the down-regulation of CAPER-α by EWS/FLI-1. CAPER-α mediates alternative splicing and controls the shift from VEGF(189) to VEGF(165) .
Collapse
Affiliation(s)
- Gangxiong Huang
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | |
Collapse
|
50
|
Feasibility and safety of silicone rubber contrast-enhanced microcomputed tomography in evaluating the angioarchitecture of prostatectomy specimens. Transl Oncol 2011; 4:173-7. [PMID: 21633672 DOI: 10.1593/tlo.10304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 03/24/2011] [Accepted: 03/24/2011] [Indexed: 01/04/2023] Open
Abstract
This ethics committee-approved pilot study was carried out with informed consent. A protocol was developed to assess the feasibility of in vitro Microfil injection of prostate cancer specimens followed by analysis with micro-computed tomography (microCT) to characterize the functional vascularity of prostatic tissue and evaluate its safety with respect to the preservation of a specimen for pathologic examination. The visible prostatic arteries of two surgically resected prostates frompatients with known prostate cancer (PCa) were injected with MicrofilMV-122 contrast medium immediately after removal. The specimens were scanned using microCT and were qualitatively examined using three-dimensional analysis software (MicroView; GE Healthcare Biosciences). The Microfil perfusion in the two samples was sufficient to view the functional vascularity arising from a major prostatic artery, up to a resolution of 17.626 µm without any indication of adverse effects due to Microfil injection. Malignant prostatic regions showed a greater vascular density on histology but decreased vascular perfusion compared with benign prostatic regions. The use of microCT on Microfil-injected prostates seems to be a feasible and specimen-preserving method for visualizing the three-dimensional vessel patterns present in resected human prostates.
Collapse
|