51
|
Riedl SJ, Pasquale EB. Targeting the Eph System with Peptides and Peptide Conjugates. Curr Drug Targets 2015; 16:1031-47. [PMID: 26212263 PMCID: PMC4861043 DOI: 10.2174/1389450116666150727115934] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/02/2015] [Accepted: 07/20/2015] [Indexed: 01/06/2023]
Abstract
Eph receptor tyrosine kinases and ephrin ligands constitute an important cell communication system that controls development, tissue homeostasis and many pathological processes. Various Eph receptors/ephrins are present in essentially all cell types and their expression is often dysregulated by injury and disease. Thus, the 14 Eph receptors are attracting increasing attention as a major class of potential drug targets. In particular, agents that bind to the extracellular ephrin-binding pocket of these receptors show promise for medical applications. This pocket comprises a broad and shallow groove surrounded by several flexible loops, which makes peptides particularly suitable to target it with high affinity and selectivity. Accordingly, a number of peptides that bind to Eph receptors with micromolar affinity have been identified using phage display and other approaches. These peptides are generally antagonists that inhibit ephrin binding and Eph receptor/ ephrin signaling, but some are agonists mimicking ephrin-induced Eph receptor activation. Importantly, some of the peptides are exquisitely selective for single Eph receptors. Most identified peptides are linear, but recently the considerable advantages of cyclic scaffolds have been recognized, particularly in light of potential optimization towards drug leads. To date, peptide improvements have yielded derivatives with low nanomolar Eph receptor binding affinity, high resistance to plasma proteases and/or long in vivo half-life, exemplifying the merits of peptides for Eph receptor targeting. Besides their modulation of Eph receptor/ephrin function, peptides can also serve to deliver conjugated imaging and therapeutic agents or various types of nanoparticles to tumors and other diseased tissues presenting target Eph receptors.
Collapse
Affiliation(s)
| | - Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
52
|
Xue C, Chen Y, Huang Z, Ge Y, Wang H, Wang J. EphB4 expression in pterygium is associated with microvessel density. Int J Clin Exp Med 2014; 7:4008-4015. [PMID: 25550909 PMCID: PMC4276167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/08/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVE Angiogenesis is a key component of the pathogenesis process of pterygium, which is the growth of fibrovascular tissue on the cornea. It has been reported that EphB4, a receptor tyrosine kinase of the ephrin-Eph system, plays important role in vascular development during embryogenesis and tumor angiogenesis and is potentially involved in ocular angiogenesis. The aim of this study is to investigate the role of EphB4 in pterygia. METHODS Fifteen pterygium samples and their paired upper bulbar conjunctiva were evaluated for expression of EphB4 and CD31 by using immunohistochemical staining. The expression level of the mRNA of EphB4 gene in 7 pterygia and matched upper bulbar conjunctiva was evaluated by using a quantitative real-time RT-PCR analysis. Microvessel density (MVD) was assessed with antibody that targets CD31. RESULTS EphB4 protein was high expressed in the epithelium and stroma of pterygia compared to those in upper bulbar conjunctiva. Immunohistochemical staining showed that pterygia presented with statistically significant higher average count of microvessel compared to normal conjunctivae (28.24 ± 6.79 vs 11.09 ± 2.96 per high power field, P < 0.001). MVD values in stroma of the pterygium and normal conjunctiva presented a significant correlation with EphB4 staining (P < 0.001). Compared with autologous upper bulbar conjunctiva grafts, the expression of the EphB4 mRNA was increased in pterygia in 4 paired samples, including those 3 recurrent ones. CONCLUSION The expression of EphB4 in pterygium was significantly related with the increased MVD and may be involved in angiogenesis. EphB4 protein is a potential target for treatment of pterygium.
Collapse
Affiliation(s)
- Chunyan Xue
- Department of Ophthalmology, Jinling Hospital, Nanjing University School of MedicineNanjing 210002, P. R. China
| | - Yueqin Chen
- Department of Ophthalmology, Jinling Hospital, Nanjing University School of MedicineNanjing 210002, P. R. China
| | - Zhenping Huang
- Department of Ophthalmology, Jinling Hospital, Nanjing University School of MedicineNanjing 210002, P. R. China
| | - Yirui Ge
- Department of Ophthalmology, Jinling Hospital, Nanjing University School of MedicineNanjing 210002, P. R. China
| | - Haiyan Wang
- Department of Pathology, Jinling Hospital, Nanjing University School of MedicineNanjing 210002, P. R. China
| | - Jiandong Wang
- Department of Pathology, Jinling Hospital, Nanjing University School of MedicineNanjing 210002, P. R. China
| |
Collapse
|
53
|
Gucciardo E, Sugiyama N, Lehti K. Eph- and ephrin-dependent mechanisms in tumor and stem cell dynamics. Cell Mol Life Sci 2014; 71:3685-710. [PMID: 24794629 PMCID: PMC11113620 DOI: 10.1007/s00018-014-1633-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/31/2014] [Accepted: 04/17/2014] [Indexed: 01/17/2023]
Abstract
The erythropoietin-producing hepatocellular (Eph) receptors comprise the largest family of receptor tyrosine kinases (RTKs). Initially regarded as axon-guidance and tissue-patterning molecules, Eph receptors have now been attributed with various functions during development, tissue homeostasis, and disease pathogenesis. Their ligands, ephrins, are synthesized as membrane-associated molecules. At least two properties make this signaling system unique: (1) the signal can be simultaneously transduced in the receptor- and the ligand-expressing cell, (2) the signaling outcome through the same molecules can be opposite depending on cellular context. Moreover, shedding of Eph and ephrin ectodomains as well as ligand-dependent and -independent receptor crosstalk with other RTKs, proteases, and adhesion molecules broadens the repertoire of Eph/ephrin functions. These integrated pathways provide plasticity to cell-microenvironment communication in varying tissue contexts. The complex molecular networks and dynamic cellular outcomes connected to the Eph/ephrin signaling in tumor-host communication and stem cell niche are the main focus of this review.
Collapse
Affiliation(s)
- Erika Gucciardo
- Research Programs Unit, Genome-Scale Biology, Biomedicum Helsinki, University of Helsinki, P.O.B. 63, 00014 Helsinki, Finland
| | - Nami Sugiyama
- Research Programs Unit, Genome-Scale Biology, Biomedicum Helsinki, University of Helsinki, P.O.B. 63, 00014 Helsinki, Finland
- Department of Biosystems Science and Bioengineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Kaisa Lehti
- Research Programs Unit, Genome-Scale Biology, Biomedicum Helsinki, University of Helsinki, P.O.B. 63, 00014 Helsinki, Finland
| |
Collapse
|
54
|
Chen FY, Yan JJ, Yi HX, Hu FQ, Du YZ, Yuan H, You J, Zhao MD. TNYL peptide functional chitosan-g-stearate conjugate micelles for tumor specific targeting. Int J Nanomedicine 2014; 9:4597-608. [PMID: 25298734 PMCID: PMC4186491 DOI: 10.2147/ijn.s69572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nowadays, a real challenge in cancer therapy is to design drug delivery systems that can achieve high concentrations of drugs at the target site for improved therapeutic effect with reduced side effects. In this research, we designed and synthesized a homing peptide-(TNYLFSPNGPIA, TNYL) modified chitosan-g-stearate (CS) polymer micelle (named T-CS) for targeting delivery. The peptide displayed specific binding affinity to EphB4 which is a member of the Eph family of receptor tyrosine protein kinases. The amphiphilic polymer T-CS can gather into micelles by themselves in an aqueous environment with a low critical micelle concentration value (91.2 μg/L) and nano-scaled size (82.1±2.8 nm). The drug encapsulation efficiency reached 86.43% after loading the hydrophobic drug doxorubicin (DOX). The cytotoxicity of T-CS/DOX against SKOV3 cells was enhanced by approximately 2.3-fold when compared with CS/DOX. The quantitative and qualitative analysis for cellular uptake indicated that TNYL modification can markedly increase cellular internalization in the EphB4-overexpressing SKOV3 cell line, especially with a short incubation time. It is interesting that relatively higher uptake of the T-CS/DOX micelles by SKOV3 cells (positive-EphB4) than A549 cells (negative-EphB4) was observed when the two cells were co-incubated. Furthermore, in vivo distribution experiment using a bilateral-tumor model showed that there was more fluorescence accumulation in the SKOV3 tumor than in the A549 tumor over the whole experiment. These results suggest that TNYL-modified CS micelles may be promising drug carriers as targeting therapy for the EphB4-overexpressing tumor.
Collapse
Affiliation(s)
- Feng-Ying Chen
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jing-Jing Yan
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Han-Xi Yi
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Fu-Qiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Yong-Zhong Du
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Jian You
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Meng-Dan Zhao
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
55
|
Huang M, Xiong C, Lu W, Zhang R, Zhou M, Huang Q, Weinberg J, Li C. Dual-modality micro-positron emission tomography/computed tomography and near-infrared fluorescence imaging of EphB4 in orthotopic glioblastoma xenograft models. Mol Imaging Biol 2014; 16:74-84. [PMID: 23918654 DOI: 10.1007/s11307-013-0674-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
PURPOSE In glioblastoma, EphB4 receptors, a member of the largest family of receptor tyrosine kinases, are overexpressed in both tumor cells and angiogenic blood vessels. The purpose of this study was to examine whether the EphB4-binding peptide TNYL-RAW labeled with both (64)Cu and near-infrared fluorescence dye Cy5.5 could be used as a molecular imaging agent for dual-modality positron emission tomography/computed tomography [PET/CT] and optical imaging of human glioblastoma in orthotopic brain tumor models. MATERIALS AND METHODS TNYL-RAW was conjugated to Cy5.5 and the radiometal chelator 1,4,7,10-tetraazadodecane-N,N',N″,N‴-tetraacetic acid. The conjugate was then labeled with (64)Cu for in vitro binding and in vivo dual μPET/CT and optical imaging studies in nude mice implanted with EphB4-expressing U251 and EphB4-negative U87 human glioblastoma cells. Tumors and brains were removed at the end of the imaging sessions for immunohistochemical staining and fluorescence microscopic examinations. RESULTS μPET/CT and near-infrared optical imaging clearly showed specific uptake of the dual-labeled TNYL-RAW peptide in both U251 and U87 tumors in the brains of the nude mice after intravenous injection of the peptide. In U251 tumors, the Cy5.5-labeled peptide colocalized with both tumor blood vessels and tumor cells; in U87 tumors, the tracer colocalized only with tumor blood vessels, not with tumor cells. CONCLUSIONS Dual-labeled EphB4-specific peptide could be used as a noninvasive molecular imaging agent for PET/CT and optical imaging of glioblastoma owing to its ability to bind to both EphB4-expressing angiogenic blood vessels and EphB4-expressing tumor cells.
Collapse
Affiliation(s)
- Miao Huang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Unit 59, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
56
|
Eph receptors as therapeutic targets in glioblastoma. Br J Cancer 2014; 111:1255-61. [PMID: 25144626 PMCID: PMC4183860 DOI: 10.1038/bjc.2014.73] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/15/2014] [Accepted: 01/16/2014] [Indexed: 12/20/2022] Open
Abstract
The dismal outlook for patients with the most aggressive and common form of adult brain cancer, glioblastoma (GBM), motivates a search for new therapeutic strategies and targets for this aggressive disease. Here we review the findings to date on the role of Eph family receptor tyrosine kinases and their ephrin ligands in brain cancer. Expression of the Eph family of cell surface proteins is generally downregulated to very low levels in normal adult tissues making them particularly attractive for directed therapeutic targeting. Recent Eph targeting studies in pre-clinical models of GBM have been very encouraging and may provide an avenue to treat these highly refractory aggressive tumours.
Collapse
|
57
|
|
58
|
Scianna M, Bell C, Preziosi L. A review of mathematical models for the formation of vascular networks. J Theor Biol 2013; 333:174-209. [DOI: 10.1016/j.jtbi.2013.04.037] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 02/15/2013] [Accepted: 04/30/2013] [Indexed: 02/08/2023]
|
59
|
Liu S, Li D, Park R, Liu R, Xia Z, Guo J, Krasnoperov V, Gill PS, Li Z, Shan H, Conti PS. PET imaging of colorectal and breast cancer by targeting EphB4 receptor with 64Cu-labeled hAb47 and hAb131 antibodies. J Nucl Med 2013; 54:1094-100. [PMID: 23667241 DOI: 10.2967/jnumed.112.116822] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Accumulating evidence suggests that ephrin type B receptor 4 (EphB4) plays a key role in the progression of numerous cancer types. In this study, we developed a series of (64)Cu-labeled antibodies for PET imaging of tumor EphB4 expression. METHODS Anti-EphB4 antibodies (hAb47 and hAb131) were conjugated with the (64)Cu-chelator DOTA through lysine, cysteine, or oligosaccharide on the antibody. DOTA-human IgG (hIgG) was also prepared as a control, which did not bind to EphB4. The EphB4 binding activity of these probes was evaluated through the bead-based binding assay with EphB4-alkaline phosphatase. The resulting PET probes were further evaluated in both HT29 (colorectal cancer) and MDA-MB-231 (breast cancer) xenografts. RESULTS All 3 conjugation methods retained most of the EphB4 binding activity of the antibodies (83.85% ± 3.82%, 76.25% ± 5.90%, 98.93% ± 3.75%, and 82.09% ± 4.14% for DOTA-Lys-hAb47, DOTA-Cys-hAb47, DOTA-Sug-hAb47, and DOTA-Lys-hAb131, respectively). Although DOTA-Sug-hAb47 demonstrated the highest receptor binding activity based on a EphB4 binding assay, the corresponding PET probe was trapped in the liver quickly in vivo. In HT29 xenografts, both (64)Cu-DOTA-Lys-hAb47 and (64)Cu-DOTA-Cys-hAb47 demonstrated prominent tumor accumulation, which reached a maximum at 48 h after injection (18.13 ± 1.73 percentage injected dose [%ID]/g and 11.81 ± 2.05 %ID/g, respectively). In contrast, (64)Cu-DOTA-Lys-hIgG had a low tumor accumulation, thus demonstrating the target specificity of EphB4-antibody-based probes. Moreover, (64)Cu-DOTA-Lys-hAb131 (29.48 ± 2.60 %ID/g) demonstrated significantly higher HT29 tumor accumulation than (64)Cu-DOTA-Lys-hAb47. (64)Cu-DOTA-Lys-hAb131 was also found to specifically accumulate in the MDA-MB-231 tumor model (12.96 ± 2.31 %ID/g). CONCLUSION We have demonstrated that EphB4 can serve as a valid target for colorectal and breast cancer imaging. This approach would be valuable for evaluating disease course and therapeutic efficacy at the earliest stages of anti-EphB4 treatment. Moreover, these newly developed probes may have important applications in other cancer types overexpressing EphB4.
Collapse
Affiliation(s)
- Shuanglong Liu
- Molecular Imaging Center, Department of Radiology, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Su D, Li X, Gao D. Inhibition of choroidal neovascularization by anti-EphB4 monoclonal antibody. Exp Ther Med 2013; 5:1226-1230. [PMID: 23596494 PMCID: PMC3628074 DOI: 10.3892/etm.2013.962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 01/16/2013] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to determine the effect of the EphB4 monoclonal antibody on experimental choroidal neovascularization (CNV) progression. Experimental CNV was established by argon laser photocoagulation. In the experimental group, the EphB4 monoclonal antibody was injected into the vitreous space in the eye specimens on days 0, 3, 6 and 9 after CNV model establishment. In the control group, an equal amount of balanced salt solution was injected at the same time points. On day 10 after CNV model establishment, fluorescein isothiocyanate-dextran endocardial perfusion and choroidal stretched preparation were conducted, respectively, for the two groups. The CNV area in each light spot and the mean values were determined. Histopathological examination was conducted and the ratio of the maximum thickness of the CNV in each light spot to the surrounding normal choroidal thickness, as well as the mean ratio, were calculated. Choroidal stretched preparation confirmed that the CNV of the experimental group was smaller, whereas the CNV of the control group was wider and larger. Quantitative analysis revealed that CNV in the experimental group was significantly inhibited (t=11.84, P<0.01) and that CNV progression in the experimental group was significantly suppressed (t=7.45, P<0.01). Histopathological examination revealed that CNV in the experimental group was thinner and smaller. Vitreous injection of the EphB4 monoclonal antibody inhibits experimental CNV progression. However, its specific mechanism remains unclear. Endogenous EphrinB2/EphB4 regulates ocular neovascularization and may become a new target in treating CNV diseases.
Collapse
Affiliation(s)
- Dongfeng Su
- Department of Ophthalmology, 463rd Hospital of Chinese People's Liberation Army, Shenyang 110042; ; Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang 110004
| | | | | |
Collapse
|
61
|
Nakada M, Kita D, Teng L, Pyko IV, Watanabe T, Hayashi Y, Hamada JI. Receptor tyrosine kinases: principles and functions in glioma invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 986:143-70. [PMID: 22879068 DOI: 10.1007/978-94-007-4719-7_8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein tyrosine kinases are enzymes that are capable of adding a phosphate group to specific tyrosines on target proteins. A receptor tyrosine kinase (RTK) is a tyrosine kinase located at the cellular membrane and is activated by binding of a ligand via its extracellular domain. Protein phosphorylation by kinases is an important mechanism for communicating signals within a cell and regulating cellular activity; furthermore, this mechanism functions as an "on" or "off" switch in many cellular functions. Ninety unique tyrosine kinase genes, including 58 RTKs, were identified in the human genome; the products of these genes regulate cellular proliferation, survival, differentiation, function, and motility. Tyrosine kinases play a critical role in the development and progression of many types of cancer, in addition to their roles as key regulators of normal cellular processes. Recent studies have revealed that RTKs such as epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR), c-Met, Tie, Axl, discoidin domain receptor 1 (DDR1), and erythropoietin-producing human hepatocellular carcinoma (Eph) play a major role in glioma invasion. Herein, we summarize recent advances in understanding the role of RTKs in glioma pathobiology, especially the invasive phenotype, and present the perspective that RTKs are a potential target of glioma therapy.
Collapse
Affiliation(s)
- Mitsutoshi Nakada
- Department of Neurosurgery, Division of Neuroscience, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| | | | | | | | | | | | | |
Collapse
|
62
|
Li D, Liu S, Liu R, Park R, Hughes L, Krasnoperov V, Gill PS, Li Z, Shan H, Conti PS. Targeting the EphB4 receptor for cancer diagnosis and therapy monitoring. Mol Pharm 2012; 10:329-36. [PMID: 23211050 DOI: 10.1021/mp300461b] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Accumulating evidence suggests that EphB4 plays key roles in cancer progression in numerous cancer types. In fact, therapies focusing on EphB4 have become potentially important components of various cancer treatment strategies. However, tumor sensitivity to EphB4 suppression may not be uniform for different cancers. In this study, we developed near-infrared fluorescence (NIRF) probes for EphB4 targeted imaging, based on EphB4-specific humanized monoclonal antibody hAb47. NIRF dye Cy5.5 was introduced to hAb47 either through the reaction with amino groups (named hAb47-Cy5.5) or sulfhydryl groups (named hAb47-Cy5.5-Mal). The resulting probes were evaluated in both HT-29 xenograft and the mAb131 (anti-EphB4) treated models. Although these methods lead to modifications of both the heavy chain and light chain of the antibody, the majority of the EphB4 binding affinity was maintained (81.62 ± 2.08% for hAb47-Cy5.5 and 77.14 ± 2.46% for hAb47-Cy5.5-Mal, respectively). hAb47-Cy5.5 was then chosen for in vivo NIRF imaging of EphB4 expression. In HT29 colorectal tumor xenografts, hAb47-Cy5.5 demonstrated significantly higher tumor uptake compared with that of the hIgG-Cy5.5 control, which was further confirmed by immunofluorescent staining. Moreover, hAb47-Cy5.5 successfully imaged the decreased EphB4 expression (confirmed by Western blot) in EphB4-targeted immunotherapy using another EphB4-specific antibody, mAb131. Collectively, hAb47-Cy5.5 could be used as a specific NIRF contrast agent for noninvasive imaging of EphB4 expression, which may predict whether an individual tumor would likely respond to EphB4 targeted interventions, as well as monitor the therapeutic response.
Collapse
Affiliation(s)
- Dan Li
- Department of Radiology, Molecular Imaging Center, University of Southern California, Los Angeles, California 90033, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
The effect of interstitial pressure on tumor growth: coupling with the blood and lymphatic vascular systems. J Theor Biol 2012; 320:131-51. [PMID: 23220211 DOI: 10.1016/j.jtbi.2012.11.031] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 10/31/2012] [Accepted: 11/28/2012] [Indexed: 12/17/2022]
Abstract
The flow of interstitial fluid and the associated interstitial fluid pressure (IFP) in solid tumors and surrounding host tissues have been identified as critical elements in cancer growth and vascularization. Both experimental and theoretical studies have shown that tumors may present elevated IFP, which can be a formidable physical barrier for delivery of cell nutrients and small molecules into the tumor. Elevated IFP may also exacerbate gradients of biochemical signals such as angiogenic factors released by tumors into the surrounding tissues. These studies have helped to understand both biochemical signaling and treatment prognosis. Building upon previous work, here we develop a vascular tumor growth model by coupling a continuous growth model with a discrete angiogenesis model. We include fluid/oxygen extravasation as well as a continuous lymphatic field, and study the micro-environmental fluid dynamics and their effect on tumor growth by accounting for blood flow, transcapillary fluid flux, interstitial fluid flow, and lymphatic drainage. We thus elucidate further the non-trivial relationship between the key elements contributing to the effects of interstitial pressure in solid tumors. In particular, we study the effect of IFP on oxygen extravasation and show that small blood/lymphatic vessel resistance and collapse may contribute to lower transcapillary fluid/oxygen flux, thus decreasing the rate of tumor growth. We also investigate the effect of tumor vascular pathologies, including elevated vascular and interstitial hydraulic conductivities inside the tumor as well as diminished osmotic pressure differences, on the fluid flow across the tumor capillary bed, the lymphatic drainage, and the IFP. Our results reveal that elevated interstitial hydraulic conductivity together with poor lymphatic function is the root cause of the development of plateau profiles of the IFP in the tumor, which have been observed in experiments, and contributes to a more uniform distribution of oxygen, solid tumor pressure and a broad-based collapse of the tumor lymphatics. We also find that the rate that IFF is fluxed into the lymphatics and host tissue is largely controlled by an elevated vascular hydraulic conductivity in the tumor. We discuss the implications of these results on microenvironmental transport barriers, and the tumor invasive and metastatic potential. Our results suggest the possibility of developing strategies of targeting tumor cells based on the cues in the interstitial fluid.
Collapse
|
64
|
Du J, Zhao W, Wang Y, Cai Y. Lentivirus vector-mediated knockdown of erythropoietin-producing hepatocellular carcinoma receptors B4 inhibits laser-induced choroidal neovascularization. J Ocul Pharmacol Ther 2012; 29:14-22. [PMID: 23035975 DOI: 10.1089/jop.2012.0077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE To evaluate the efficacy of erythropoietin-producing hepatocellular carcinoma receptors B4 (EphB4) knockdown on the development of laser-induced choroidal neovascularization (CNV) in vivo. METHODS We constructed recombinant lentiviral vectors (Lv) Lv-shRNA-EphB4 to specifically knock down the expression of EphB4. The mRNA and protein expression of EphB4 was investigated by real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blot. CNV was induced by laser photocoagulation in C57BL/6 mice. The mice were then randomly assigned to be intravitreally injected with phosphate-buffered saline (PBS), Lv-shRNA-EphB4 recombinant lentivirus, or an unrelated shRNA recombinant lentivirus (pFU LV-shRNA-NC). An uninjected group was used as the control. Fundus fluorescein angiography (FFA), histologic analysis, and choroidal flat mounts analysis were applied to evaluate the inhibition of CNV after an intravitreal injection. RESULTS Transfection of Lv-shRNA-EphB4 led to the knockdown of EphB4, and EphB4 mRNA was down-regulated by about 80%. FFA and histologic analysis revealed that the leakage areas and the mean thickness of CNV were much smaller in the Lv-shRNA-EphB4 group than in the PBS-treated, pFU Lv-shRNA-NC group and the non-injection group. Choroidal flat mounts showed significantly less leakage and smaller leakage areas in the Lv-shRNA-EphB4 group than those in other groups. CONCLUSION Knocking down the expression of EphB4 exerts an inhibitory effect on CNV in vivo. It may provide a potential strategy for the treatment of CNV.
Collapse
Affiliation(s)
- Jing Du
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University , Xi'an, People's Republic of China
| | | | | | | |
Collapse
|
65
|
Salvucci O, Tosato G. Essential roles of EphB receptors and EphrinB ligands in endothelial cell function and angiogenesis. Adv Cancer Res 2012; 114:21-57. [PMID: 22588055 DOI: 10.1016/b978-0-12-386503-8.00002-8] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Eph receptor tyrosine kinases and their Ephrin ligands represent an important signaling system with widespread roles in cell physiology and disease. Receptors and ligands in this family are anchored to the cell surface; thus Eph/Ephrin interactions mainly occur at sites of cell-to-cell contact. EphB4 and EphrinB2 are the Eph/Ephrin molecules that play essential roles in vascular development and postnatal angiogenesis. Analysis of expression patterns and function has linked EphB4/EphrinB2 to endothelial cell growth, survival, migration, assembly, and angiogenesis. Signaling from these molecules is complex, with the potential for being bidirectional, emanating both from the Eph receptors (forward signaling) and from the Ephrin ligands (reverse signaling). In this review, we describe recent advances on the roles of EphB/EphrinB protein family in endothelial cell function and outline potential approaches to inhibit pathological angiogenesis based on this understanding.
Collapse
Affiliation(s)
- Ombretta Salvucci
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | |
Collapse
|
66
|
Chen J. Regulation of tumor initiation and metastatic progression by Eph receptor tyrosine kinases. Adv Cancer Res 2012; 114:1-20. [PMID: 22588054 DOI: 10.1016/b978-0-12-386503-8.00001-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In recent years, a growing body of evidence has indicated that signaling molecules previously implicated in axon guidance are important regulators of multistep tumorigenesis and progression. Eph receptors and ephrins belong to this special class of molecules that play important roles in both axon guidance and cancer. Tremendous progress has been made in the past few years in both understanding the role of Eph receptors and ephrins in cancer and designing therapeutic strategies for cancer therapy. This review will focus on new advances in elucidating the contribution of Eph/ephrin molecules to key processes in tumor initiation and metastatic progression, including cancer cell proliferation, invasion and metastasis, and tumor angiogenesis.
Collapse
Affiliation(s)
- Jin Chen
- VA Medical Center, Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
67
|
You J, Zhang R, Xiong C, Zhong M, Melancon M, Gupta S, Nick AM, Sood AK, Li C. Effective photothermal chemotherapy using doxorubicin-loaded gold nanospheres that target EphB4 receptors in tumors. Cancer Res 2012; 72:4777-86. [PMID: 22865457 DOI: 10.1158/0008-5472.can-12-1003] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photothermal ablation (PTA) is an emerging technique that uses near-infrared (NIR) laser light-generated heat to destroy tumor cells. However, complete tumor eradication by PTA therapy alone is difficult because heterogeneous heat distribution can lead to sublethal thermal dose in some areas of the tumor. Successful PTA therapy requires selective delivery of photothermal conducting nanoparticles to mediate effective PTA of tumor cells, and the ability to combine PTA with other therapy modalities. Here, we synthesized multifunctional doxorubicin (DOX)-loaded hollow gold nanospheres (DOX@HAuNS) that target EphB4, a member of the Eph family of receptor tyrosine kinases overexpressed on the cell membrane of multiple tumors and angiogenic blood vessels. Increased uptake of targeted nanoparticles T-DOX@HAuNS was observed in three EphB4-positive tumors both in vitro and in vivo. In vivo release of DOX from DOX@HAuNS, triggered by NIR laser, was confirmed by dual-radiotracer technique. Treatment with T-DOX@HAuNS followed by NIR laser irradiation resulted in significantly decreased tumor growth when compared with treatments with nontargeted DOX@HAuNS plus laser or HAuNS plus laser. The tumors in 6 of the 8 mice treated with T-DOX@HAuNS plus laser regressed completely with only residual scar tissue by 22 days following injection, and none of the treatment groups experienced a loss in body weight. Together, our findings show that concerted chemo-photothermal therapy with a single nanodevice capable of mediating simultaneous PTA and local drug release may have promise as a new anticancer therapy.
Collapse
Affiliation(s)
- Jian You
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
McDougall SR, Watson MG, Devlin AH, Mitchell CA, Chaplain MAJ. A hybrid discrete-continuum mathematical model of pattern prediction in the developing retinal vasculature. Bull Math Biol 2012; 74:2272-314. [PMID: 22829182 DOI: 10.1007/s11538-012-9754-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 07/03/2012] [Indexed: 01/26/2023]
Abstract
Pathological angiogenesis has been extensively explored by the mathematical modelling community over the past few decades, specifically in the contexts of tumour-induced vascularisation and wound healing. However, there have been relatively few attempts to model angiogenesis associated with normal development, despite the availability of animal models with experimentally accessible and highly ordered vascular topologies: for example, growth and development of the vascular plexus layers in the murine retina. The current study aims to address this issue through the development of a hybrid discrete-continuum mathematical model of the developing retinal vasculature in neonatal mice that is closely coupled with an ongoing experimental programme. The model of the functional vasculature is informed by a range of morphological and molecular data obtained over a period of several days, from 6 days prior to birth to approximately 8 days after birth. The spatio-temporal formation of the superficial retinal vascular plexus (RVP) in wild-type mice occurs in a well-defined sequence. Prior to birth, astrocytes migrate from the optic nerve over the surface of the inner retina in response to a chemotactic gradient of PDGF-A, formed at an earlier stage by migrating retinal ganglion cells (RGCs). Astrocytes express a variety of chemotactic and haptotactic proteins, including VEGF and fibronectin (respectively), which subsequently induce endothelial cell sprouting and modulate growth of the RVP. The developing RVP is not an inert structure; however, the vascular bed adapts and remodels in response to a wide variety of metabolic and biomolecular stimuli. The main focus of this investigation is to understand how these interacting cellular, molecular, and metabolic cues regulate RVP growth and formation. In an earlier one-dimensional continuum model of astrocyte and endothelial migration, we showed that the measured frontal velocities of the two cell types could be accurately reproduced by means of a system of five coupled partial differential equations (Aubert et al. in Bull. Math. Biol. 73:2430-2451, 2011). However, this approach was unable to generate spatial information and structural detail for the entire retinal surface. Building upon this earlier work, a more realistic two-dimensional hybrid PDE-discrete model is derived here that tracks the migration of individual astrocytes and endothelial tip cells towards the outer retinal boundary. Blood perfusion is included throughout plexus development and the emergent retinal architectures adapt and remodel in response to various biological factors. The resulting in silico RVP structures are compared with whole-mounted retinal vasculatures at various stages of development, and the agreement is found to be excellent. Having successfully benchmarked the model against wild-type data, the effect of transgenic over-expression of various genes is predicted, based on the ocular-specific expression of VEGF-A during murine development. These results can be used to help inform future experimental investigations of signalling pathways in ocular conditions characterised by aberrant angiogenesis.
Collapse
Affiliation(s)
- S R McDougall
- Institute of Petroleum Engineering, Heriot-Watt University, Edinburgh, Scotland, UK
| | | | | | | | | |
Collapse
|
69
|
Tu Y, He S, Fu J, Li G, Xu R, Lu H, Deng J. Expression of EphrinB2 and EphB4 in glioma tissues correlated to the progression of glioma and the prognosis of glioblastoma patients. Clin Transl Oncol 2012; 14:214-20. [PMID: 22374425 DOI: 10.1007/s12094-012-0786-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
OBJECTIVE The ligand EphrinB2 and the corresponding receptor EphB4 are up-regulated and involved in tumour growth in various human cancers. However, little is known about how this receptor-ligand complex contributes to the progression of glioma. This prompted us to study the association between the expressions of EphrinB2 and EphB4, clinicopathological variables, and glioma patient outcome. METHODS Immunohistochemical staining was performed to detect the expression patterns of EphrinB2 and EphB4 in the biopsies from 96 patients with primary gliomas. Kaplan-Meier survival and Cox regression analyses were performed to evaluate the prognosis of patients. RESULTS Immunohistochemical analysis revealed that the expression of EphrinB2 was significantly correlated with that of EphB4 (r=0.86, p=0.002). EphrinB2 and EphB4 were significantly associated with the Karnofsky performance scale (KPS) score and World Health Organization grades of patients with gliomas, respectively. Especially, the positive expression rates of EphrinB2 and EphB4 were significantly higher in patients with higher grade (both p=0.001) and lower KPS score (p=0.002 and 0.003, respectively). Multivariate Cox regression analysis revealed that EphrinB2 and EphB4 expressions were both independent prognostic factors for progress-free survival of glioblastoma patients (both p=0.02). CONCLUSION Our data indicated for the first time that EphrinB2 and EphB4 expressions increase according to the histopathological grade and KPS score of glioma, and their expression levels are related to the progression-free survival of glioblastoma patients.
Collapse
Affiliation(s)
- Yanyang Tu
- Department of Emergency, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | | | | | | | | | | | | |
Collapse
|
70
|
Zheng MF, Ji Y, Wu XB, Ye SG, Chen JY. EphB4 gene polymorphism and protein expression in non-small-cell lung cancer. Mol Med Rep 2012; 6:405-8. [PMID: 22684742 DOI: 10.3892/mmr.2012.936] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/14/2012] [Indexed: 11/05/2022] Open
Abstract
The objective of this study was to identify new diagnostic, prognostic or therapeutic molecules for non-small- cell lung cancer (NSCLC). We investigated the expression of EphB4, a tyrosine kinase receptor which has been shown to act as a tumor promoter in other cancers. Using immunohistochemistry, we visualized EphB4 expression in 28 samples of NSCLC and 12 samples of adjacent normal tissues. Additionally, we assessed a single-nucleotide polymorphism in EphB4 to determine its effect on protein expression. The correlation of both genotype and protein expression with disease severity was determined. EphB4 was expressed in 53.6% of patients with lung cancer, a significant increase compared to control lung samples (0.0%, P<0.05). Furthermore, EphB4 expression was correlated with differentiation, lymph node metastasis and TNM stage of tumors (P<0.05). Additionally, the polymorphism in EphB4 at rs314310 appeared to correspond to protein expression and disease susceptibility. While the frequencies of CC, CA and AA genotypes were not different between lung cancer patients and healthy controls, the frequencies of C and A alleles were significantly different between these groups (P<0.05). Further analysis showed that the positive rate of EphB4 expression in patients with the AA genotype was significantly higher compared to that in patients with other genotypes (P<0.05). Overexpression of EphB4 plays a role in the occurrence and development of NSCLC, and the polymorphism at rs314310 may predispose individuals to this disease.
Collapse
Affiliation(s)
- Ming-Feng Zheng
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, PR China
| | | | | | | | | |
Collapse
|
71
|
Nievergall E, Lackmann M, Janes PW. Eph-dependent cell-cell adhesion and segregation in development and cancer. Cell Mol Life Sci 2012; 69:1813-42. [PMID: 22204021 PMCID: PMC11114713 DOI: 10.1007/s00018-011-0900-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/06/2011] [Accepted: 11/28/2011] [Indexed: 01/23/2023]
Abstract
Numerous studies attest to essential roles for Eph receptors and their ephrin ligands in controlling cell positioning and tissue patterning during normal and oncogenic development. These studies suggest multiple, sometimes contradictory, functions of Eph-ephrin signalling, which under different conditions can promote either spreading and cell-cell adhesion or cytoskeletal collapse, cell rounding, de-adhesion and cell-cell segregation. A principle determinant of the balance between these two opposing responses is the degree of receptor/ligand clustering and activation. This equilibrium is likely altered in cancers and modulated by somatic mutations of key Eph family members that have emerged as candidate cancer markers in recent profiling studies. In addition, cross-talk amongst Ephs and with other signalling pathways significantly modulates cell-cell adhesion, both between and within Eph- and ephrin-expressing cell populations. This review summarises our current understanding of how Eph receptors control cell adhesion and morphology, and presents examples demonstrating the importance of these events in normal development and cancer.
Collapse
Affiliation(s)
- Eva Nievergall
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, VIC 3800 Australia
- Present Address: Haematology Department, SA Pathology, Frome Road, Adelaide, SA 5000 Australia
| | - Martin Lackmann
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, VIC 3800 Australia
| | - Peter W. Janes
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, VIC 3800 Australia
| |
Collapse
|
72
|
Kawano H, Katayama Y, Minagawa K, Shimoyama M, Henkemeyer M, Matsui T. A novel feedback mechanism by Ephrin-B1/B2 in T-cell activation involves a concentration-dependent switch from costimulation to inhibition. Eur J Immunol 2012; 42:1562-72. [PMID: 22622783 DOI: 10.1002/eji.201142175] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 01/25/2012] [Accepted: 02/16/2012] [Indexed: 11/10/2022]
Abstract
Bidirectional signals via Eph receptors/ephrins have been recognized as major forms of contact-dependent cell communications such as cell attraction and repulsion. T cells express EphBs, and their ligands, the ephrin-Bs, have been known as costimulatory molecules for T-cell proliferation. Recently, another remarkable feature of ephrin-As has emerged in the form of a concentration-dependent transition from promotion to inhibition in axon growth. Here we examined whether this modification plays a role in ephrin-B costimulation in murine primary T cells. Low doses of ephrin-B1 and ephrin-B2 costimulated T-cell proliferation induced by anti-CD3, but high concentrations strongly inhibited it. In contrast, ephrin-B3 showed a steadily increasing stimulatory effect. This modulation was virtually preserved in T cells from mice simultaneously lacking four genes, EphB1, EphB2, EphB3, and EphB6. High concentrations of ephrin-B1/B2, but not ephrin-B3, inhibited the anti-CD3-induced phosphorylation of Lck and its downstream signals such as Erk and Akt. Additionally, high doses of any ephrin-Bs could phosphorylate EphB4. However, only ephrin-B1/B2 but not ephrin-B3 recruited SHP1, a phosphatase to suppress the phosphorylation of Lck. These data suggest that EphB4 signaling could engage in negative feedback to TCR signals. T-cell activation may be finely adjusted by the combination and concentration of ephrin-Bs expressed in the immunological microenvironment.
Collapse
Affiliation(s)
- Hiroki Kawano
- Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | | | | | | | | | | |
Collapse
|
73
|
Pericytes on the tumor vasculature: jekyll or hyde? CANCER MICROENVIRONMENT 2012; 6:1-17. [PMID: 22467426 DOI: 10.1007/s12307-012-0102-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 03/08/2012] [Indexed: 12/15/2022]
Abstract
The induction of tumor vasculature, known as the 'angiogenic switch', is a rate-limiting step in tumor progression. Normal blood vessels are composed of two distinct cell types: endothelial cells which form the channel through which blood flows, and mural cells, the pericytes and smooth muscle cells which serve to support and stabilize the endothelium. Most functional studies have focused on the responses of endothelial cells to pro-angiogenic stimuli; however, there is mounting evidence that the supporting mural cells, particularly pericytes, may play key regulatory roles in both promoting vessel growth as well as terminating vessel growth to generate a mature, quiescent vasculature. Tumor vessels are characterized by numerous structural and functional abnormalities, including altered association between endothelial cells and pericytes. These dysfunctional, unstable vessels contribute to hypoxia, interstitial fluid pressure, and enhanced susceptibility to metastatic invasion. Increasing evidence points to the pericyte as a critical regulator of endothelial activation and subsequent vessel development, stability, and function. Here we discuss both the stimulatory and inhibitory effects of pericytes on the vasculature and the possible utilization of vessel normalization as a therapeutic strategy to combat cancer.
Collapse
|
74
|
Wnuk M, Hlushchuk R, Janot M, Tuffin G, Martiny-Baron G, Holzer P, Imbach-Weese P, Djonov V, Huynh-Do U. Podocyte EphB4 signaling helps recovery from glomerular injury. Kidney Int 2012; 81:1212-25. [PMID: 22398409 DOI: 10.1038/ki.2012.17] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Eph receptor tyrosine kinases and their ligands (ephrins) have a pivotal role in the homeostasis of many adult organs and are widely expressed in the kidney. Glomerular diseases beginning with mesangiolysis can recover, with podocytes having a critical role in this healing process. We studied here the role of Eph signaling in glomerular disease recovery following mesangiolytic Thy1.1 nephritis in rats. EphB4 and ephrinBs were expressed in healthy glomerular podocytes and were upregulated during Thy1.1 nephritis, with EphB4 strongly phosphorylated around day 9. Treatment with NPV-BHG712, an inhibitor of EphB4 phosphorylation, did not cause glomerular changes in control animals. Nephritic animals treated with vehicle did not have morphological evidence of podocyte injury or loss; however, application of this inhibitor to nephritic rats induced glomerular microaneurysms, podocyte damage, and loss. Prolonged NPV-BHG712 treatment resulted in increased albuminuria and dysregulated mesangial recovery. Additionally, NPV-BHG712 inhibited capillary repair by intussusceptive angiogenesis (an alternative to sprouting angiogenesis), indicating a previously unrecognized role of podocytes in regulating intussusceptive vessel splitting. Thus, our results identify EphB4 signaling as a pathway allowing podocytes to survive transient capillary collapse during glomerular disease.
Collapse
Affiliation(s)
- Monika Wnuk
- Department of Nephrology and Hypertension, Inselspital, University of Bern Medical School, Bern, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Schlosser S, Dennler C, Schweizer R, Eberli D, Stein JV, Enzmann V, Giovanoli P, Erni D, Plock JA. Paracrine effects of mesenchymal stem cells enhance vascular regeneration in ischemic murine skin. Microvasc Res 2012; 83:267-75. [PMID: 22391452 DOI: 10.1016/j.mvr.2012.02.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 02/08/2012] [Accepted: 02/19/2012] [Indexed: 02/07/2023]
Abstract
New theories on the regeneration of ischemic vasculature have emerged indicating a pivotal role of adult stem cells. The aim of this study was to investigate homing and hemodynamic effects of circulating bone marrow-derived mesenchymal stem cells (MSCs) in a critically ischemic murine skin flap model. Bone marrow-derived mesenchymal stem cells (Lin(-)CD105(+)) were harvested from GFP(+)-donor mice and transferred to wildtype C57BL/6 mice. Animals receiving GFP(+)-fibroblasts served as a control group. Laser scanning confocal microscopy and intravital fluorescence microscopy were used for morphological analysis, monitoring and quantitative assessment of the stem cell homing and microhemodynamics over two weeks. Immunohistochemical staining was performed for GFP, eNOS, iNOS, VEGF. Tissue viability was analyzed by TUNEL-assay. We were able to visualize perivascular homing of MSCs in vivo. After 4 days, MSCs aligned along the vascular wall without undergoing endothelial or smooth muscle cell differentiation during the observation period. The gradual increase in arterial vascular resistance observed in the control group was abolished after MSC administration (P<0.01). At capillary level, a strong angiogenic response was found from day 7 onwards. Functional capillary density was raised in the MSC group to 197% compared to 132% in the control group (P<0.01). Paracrine expression of VEGF and iNOS, but not eNOS could be shown in the MSC group but not in the controls. In conclusion, we demonstrated that circulating bone marrow-derived MSCs home to perivascular sites in critically ischemic tissue, exhibits paracrine function and augment microhemodynamics. These effects were mediated through arteriogenesis and angiogenesis, which contributed to vascular regeneration.
Collapse
Affiliation(s)
- Stefan Schlosser
- Department of Clinical Research, University of Bern, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Dou GR, Wang L, Wang YS, Han H. Notch signaling in ocular vasculature development and diseases. Mol Med 2012; 18:47-55. [PMID: 21989947 PMCID: PMC3269647 DOI: 10.2119/molmed.2011.00256] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 10/04/2011] [Indexed: 01/10/2023] Open
Abstract
Ocular angiogenesis, characterized by the formation of new blood vessels in the avascular area in eyes, is a highly coordinated process involved in retinal vasculature formation and several ocular diseases such as age-related macular degeneration, proliferative diabetic retinopathy and retinopathy of prematurity. This process is orchestrated by complicated cellular interactions and vascular growth factors, during which endothelial cells acquire heterogeneous phenotypes and distinct cellular destinations. To date, while the vascular endothelial growth factor has been identified as the most critical angiogenic agent with a remarkable therapeutic value, the Notch signaling pathway appears to be a similarly important regulator in several angiogenic steps. Recent progress has highlighted the involvement, mechanisms and therapeutic potential of Notch signaling in retinal vasculature development and pathological angiogenesis-related eye disorders, which may cause irreversible blindness.
Collapse
Affiliation(s)
- Guo-Rui Dou
- Department of Ophthalmology, Xijing Hospital, Xi’an, China
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| | - Lin Wang
- Department of Hepatic Surgery, Xijing Hospital, Xi’an, China
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| | - Yu-Sheng Wang
- Department of Ophthalmology, Xijing Hospital, Xi’an, China
| | - Hua Han
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
77
|
Jumping the barrier: VE-cadherin, VEGF and other angiogenic modifiers in cancer. Biol Cell 2012; 103:593-605. [PMID: 22054419 DOI: 10.1042/bc20110069] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The endothelial barrier controls the passage of fluids, nutrients and cells through the vascular wall. This physiological function is closely related to developmental and adult angiogenesis, blood pressure control, as well as immune responses. Moreover, cancer progression is frequently characterized by disorganized and leaky blood vessels. In this context, vascular permeability drives tumour-induced angiogenesis, blood flow disturbances, inflammatory cell infiltration and tumour cell extravasation. Although various molecules have been implicated, the vascular endothelial adhesion molecule, VE-cadherin (vascular endothelial cadherin), has emerged as a critical player involved in maintaining endothelial barrier integrity and homoeostasis. Indeed, VE-cadherin coordinates the endothelial cell-cell junctions through its adhesive and signalling properties. Of note, many angiogenic and inflammatory mediators released into the tumour microenvironment influence VE-cadherin behaviour. Therefore restoring VE-cadherin function could be one very promising target for vascular normalization in cancer therapies. In this review, we will mainly focus on recent discoveries concerning the molecular mechanisms involved in modulating VE-cadherin plasticity in cancer.
Collapse
|
78
|
Trindade A, Djokovic D, Gigante J, Badenes M, Pedrosa AR, Fernandes AC, Lopes-da-Costa L, Krasnoperov V, Liu R, Gill PS, Duarte A. Low-dosage inhibition of Dll4 signaling promotes wound healing by inducing functional neo-angiogenesis. PLoS One 2012; 7:e29863. [PMID: 22279550 PMCID: PMC3261161 DOI: 10.1371/journal.pone.0029863] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 12/05/2011] [Indexed: 01/09/2023] Open
Abstract
Recent findings regarding Dll4 function in physiological and pathological conditions indicate that this Notch ligand may constitute an important therapeutic target. Dll4 appears to be a major anti-angiogenic agent, occupying a central role in various angiogenic pathways. The first trials of anti-Dll4 therapy in mice demonstrated a paradoxical effect, as it reduced tumor perfusion and growth despite leading to an increase in vascular density. This is seen as the result of insufficient maturation of the newly formed vasculature causing a circulatory defect and increased tumor hypoxia. As Dll4 function is known to be closely dependent on expression levels, we envisioned that the therapeutic anti-Dll4 dosage could be modulated to result in the increase of adequately functional blood vessels. This would be useful in conditions where vascular function is a limiting factor for recovery, like wound healing and tissue hypoxia, especially in diabetic patients. Our experimental results in mice confirmed this possibility, revealing that low dosage inhibition of Dll4/Notch signaling causes improved vascular function and accelerated wound healing.
Collapse
Affiliation(s)
- Alexandre Trindade
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Dusan Djokovic
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Joana Gigante
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
| | - Marina Badenes
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
| | - Ana-Rita Pedrosa
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
| | - Ana-Carina Fernandes
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
| | - Luís Lopes-da-Costa
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
| | | | - Ren Liu
- Department of Pathology, University of Southern California, Los Angeles, California, United States of America
| | - Parkash S. Gill
- Department of Pathology, University of Southern California, Los Angeles, California, United States of America
- Department of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail: (PSG); (AD)
| | - António Duarte
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Lisbon Technical University, Lisbon, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- * E-mail: (PSG); (AD)
| |
Collapse
|
79
|
Noberini R, Mitra S, Salvucci O, Valencia F, Duggineni S, Prigozhina N, Wei K, Tosato G, Huang Z, Pasquale EB. PEGylation potentiates the effectiveness of an antagonistic peptide that targets the EphB4 receptor with nanomolar affinity. PLoS One 2011; 6:e28611. [PMID: 22194865 PMCID: PMC3237458 DOI: 10.1371/journal.pone.0028611] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 11/11/2011] [Indexed: 01/12/2023] Open
Abstract
The EphB4 receptor tyrosine kinase together with its preferred ligand, ephrin-B2, regulates a variety of physiological and pathological processes, including tumor progression, pathological forms of angiogenesis, cardiomyocyte differentiation and bone remodeling. We previously reported the identification of TNYL-RAW, a 15 amino acid-long peptide that binds to the ephrin-binding pocked of EphB4 with low nanomolar affinity and inhibits ephrin-B2 binding. Although ephrin-B2 interacts promiscuously with all the EphB receptors, the TNYL-RAW peptide is remarkably selective and only binds to EphB4. Therefore, this peptide is a useful tool for studying the biological functions of EphB4 and for imaging EphB4-expressing tumors. Furthermore, TNYL-RAW could be useful for treating pathologies involving EphB4-ephrin-B2 interaction. However, the peptide has a very short half-life in cell culture and in the mouse blood circulation due to proteolytic degradation and clearance by the kidneys and reticuloendothelial system. To overcome these limitations, we have modified TNYL-RAW by fusion with the Fc portion of human IgG1, complexation with streptavidin or covalent coupling to a 40 KDa branched polyethylene glycol (PEG) polymer. These modified forms of TNYL-RAW all have greatly increased stability in cell culture, while retaining high binding affinity for EphB4. Furthermore, PEGylation most effectively increases peptide half-life in vivo. Consistent with increased stability, submicromolar concentrations of PEGylated TNYL-RAW effectively impair EphB4 activation by ephrin-B2 in cultured B16 melanoma cells as well as capillary-like tube formation and capillary sprouting in co-cultures of endothelial and epicardial mesothelial cells. Therefore, PEGylated TNYL-RAW may be useful for inhibiting pathological forms of angiogenesis through a novel mechanism involving disruption of EphB4-ephrin-B2 interactions between endothelial cells and supporting perivascular mesenchymal cells. Furthermore, the PEGylated peptide is suitable for other cell culture and in vivo applications requiring prolonged EphB4 receptor targeting.
Collapse
Affiliation(s)
- Roberta Noberini
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Sayantan Mitra
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Ombretta Salvucci
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fatima Valencia
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Srinivas Duggineni
- Department of Pharmacology, State University of New York Upstate Cancer Research Institute, State University of New York, Syracuse, New York, United States of America
| | - Natalie Prigozhina
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- Biology Department, University of San Diego, San Diego, California, United States of America
| | - Ke Wei
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ziwei Huang
- Department of Pharmacology, State University of New York Upstate Cancer Research Institute, State University of New York, Syracuse, New York, United States of America
| | - Elena B. Pasquale
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- Department of Pathology, University of California San Diego, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
80
|
Endothelial colony-forming cells show a mature transcriptional response to shear stress. In Vitro Cell Dev Biol Anim 2011; 48:21-9. [PMID: 22101679 DOI: 10.1007/s11626-011-9470-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 11/02/2011] [Indexed: 12/23/2022]
Abstract
Endothelial progenitor cells (EPC) play a central role in endothelial maintenance and repair. Endothelial colony-forming cells (ECFC) form a subpopulation of EPC. ECFC are readily attainable, can be easily isolated, possess a high proliferation potential, and are therefore a promising source of endothelial cells (EC) for future cardiovascular therapeutic applications. The extent to which these cells respond to shear stress as adult vascular EC remains to be elucidated. Here, we study the transcriptional response of ECFC induced by shear stress and compare it with the response of mature arterial and venous cells. ECFC, as well as human umbilical vein EC (HUVEC) and human umbilical artery EC (HUAEC), were subjected to low (0.5 Pa) and high (2.5 Pa) shear stress. The endothelial differentiation phenotype and transcriptional responses were analyzed using immunocytochemistry and quantitative polymerase chain reaction (Q-PCR). Performing absolute quantification of copy numbers by Q-PCR allows comparing the responses of cell types relative to each other. Our data show that isolated ECFC resemble mature EC in cobblestone morphology and endothelial marker expression. Absolute Q-PCR quantification revealed that although being truly endothelial, ECFC do not fully resemble HUVEC or HUAEC in the expression of specific differentiation markers. When subjected to shear stress, ECFC show a mature response to fluid flow, comparable to that of HUVEC and HUAEC. The capacity of endothelial progenitors to respond to fluid flow in a similar manner to HUVEC and HUAEC highlights the universal response of EC to fluid shear stress, independently of their endothelial differentiation status. This property supports the use of these cells as an EC source for tissue engineering applications.
Collapse
|
81
|
Diercke K, Kohl A, Lux CJ, Erber R. Strain-dependent up-regulation of ephrin-B2 protein in periodontal ligament fibroblasts contributes to osteogenesis during tooth movement. J Biol Chem 2011; 286:37651-64. [PMID: 21880727 DOI: 10.1074/jbc.m110.166900] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
During orthodontic tooth movement, the application of adequate orthodontic forces allows teeth to be moved through the alveolar bone. These forces are transmitted through the periodontal ligaments (PDL) to the supporting alveolar bone and lead to deposition or resorption of bone, depending on whether the tissues are exposed to a tensile or compressive mechanical strain. Fibroblasts within the PDL (PDLF) are considered to be mechanoresponsive. The transduction mechanisms from mechanical loading of the PDLF to the initiation of bone remodeling are not clearly understood. Recently, members of the ephrin/Eph family have been shown to be involved in the regulation of bone homeostasis. For the first time, we demonstrate that PDLF exposed to tensile strain induce the expression of ephrin-B2 via a FAK-, Ras-, ERK1/2-, and SP1-dependent pathway. Osteoblasts of the alveolar bone stimulated with ephrin-B2 increased their osteoblastogenic gene expression and showed functional signs of osteoblastic differentiation. In a physiological setting, ephrin-B2-EphB4 signaling between PDLF and osteoblasts of the alveolar bone might contribute to osteogenesis at tension sites during orthodontic tooth movement.
Collapse
Affiliation(s)
- Katja Diercke
- Department of Orthodontics and Dentofacial Orthopaedics, Dental School, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
82
|
Nakada M, Hayashi Y, Hamada JI. Role of Eph/ephrin tyrosine kinase in malignant glioma. Neuro Oncol 2011; 13:1163-70. [PMID: 21856686 DOI: 10.1093/neuonc/nor102] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Accumulating evidence has revealed that the tyrosine kinases play a major role in glioma proliferation and invasion. The largest family of tyrosine kinases, the Eph family, and its ligands, the ephrins, are frequently overexpressed in glioma, suggesting important roles for their bidirectional signals in glioma pathobiology. Ephs bind to cell surface-associated ephrin ligands on neighboring cells and have many biological functions during embryonic development of the central nervous system, including axon mapping, cell migration, and angiogenesis. Recent findings suggest that Eph/ephrin signaling affects glioma cell growth, migration, and invasion in vitro and in vivo. However, their roles in glioma seem complex, because both tumor growth promoter and suppressor potentials have been ascribed to Ephs and ephrins. Here, we review recent advances in research on the role of Eph/ephrin signaling in glioma and suggest that the Eph/ephrin system could be a potential target of glioma therapy.
Collapse
Affiliation(s)
- Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| | | | | |
Collapse
|
83
|
Li JL, Sainson RCA, Oon CE, Turley H, Leek R, Sheldon H, Bridges E, Shi W, Snell C, Bowden ET, Wu H, Chowdhury PS, Russell AJ, Montgomery CP, Poulsom R, Harris AL. DLL4-Notch signaling mediates tumor resistance to anti-VEGF therapy in vivo. Cancer Res 2011; 71:6073-83. [PMID: 21803743 DOI: 10.1158/0008-5472.can-11-1704] [Citation(s) in RCA: 188] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Resistance to VEGF inhibitors is emerging as a major clinical problem. Notch signaling has been implicated in tumor angiogenesis. Therefore, to investigate mechanisms of resistance to angiogenesis inhibitors, we transduced human glioblastoma cells with retroviruses encoding Notch delta-like ligand 4 (DLL4), grew them as tumor xenografts and then treated the murine hosts with the VEGF-A inhibitor bevacizumab. We found that DLL4-mediated tumor resistance to bevacizumab in vivo. The large vessels induced by DLL4-Notch signaling increased tumor blood supply and were insensitive to bevacizumab. However, blockade of Notch signaling by dibenzazepine, a γ-secretase inhibitor, disrupted the large vessels and abolished the tumor resistance. Multiple molecular mechanisms of resistance were shown, including decreased levels of hypoxia-induced VEGF and increased levels of the VEGF receptor VEGFR1 in the tumor stroma, decreased levels of VEGFR2 in large blood vessels, and reduced levels of VEGFR3 overall. DLL4-expressing tumors were also resistant to a VEGFR targeting multikinase inhibitor. We also observed activation of other pathways of tumor resistance driven by DLL4-Notch signaling, including the FGF2-FGFR and EphB4-EprinB2 pathways, the inhibition of which reversed tumor resistance partially. Taken together, our findings show the importance of classifying mechanisms involved in angiogenesis in tumors, and how combination therapy to block DLL4-Notch signaling may enhance the efficacy of VEGF inhibitors, particularly in DLL4-upregulated tumors, and thus provide a rational base for the development of novel strategies to overcome antiangiogenic resistance in the clinic.
Collapse
Affiliation(s)
- Ji-Liang Li
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Zhang R, Xiong C, Huang M, Zhou M, Huang Q, Wen X, Liang D, Li C. Peptide-conjugated polymeric micellar nanoparticles for Dual SPECT and optical imaging of EphB4 receptors in prostate cancer xenografts. Biomaterials 2011; 32:5872-9. [PMID: 21612822 DOI: 10.1016/j.biomaterials.2011.04.070] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 04/24/2011] [Indexed: 12/14/2022]
Abstract
EphB4, a member of the largest family of receptor tyrosine kinases, is overexpressed in numerous tumors. In this study, we developed a new class of multimodal nanoplatform for dual single photon emission computed tomography (SPECT) and near-infrared fluorescence imaging of EphB4. EphB4-binding peptide TNYL-FSPNGPIARAW (TNYL-RAW) was conjugated to polyethylene glycol-coated, core-crosslinked polymeric micelles (CCPM) dually labeled with near-infrared fluorescence fluorophores (Cy7) and a radioisotope (indium 111). In vitro, TNYL-RAW-CCPM selectively bound to EphB4-positive PC-3M prostate cancer cells, but not to EphB4-negative A549 lung cancer cells. In vivo, PC-3M tumors were clearly visualized by both SPECT and near-infrared fluorescence tomography after intravenous administration of (111)In-labeled TNYL-RAW-CCPM. In contrast, there was little signal in A549 tumors of mice injected with (111)In-labeled TNYL-RAW-CCPM or in PC-3M tumors of mice injected with (111)In-labeled CCPM. The high accumulation of (111)In-labeled TNYL-RAW-CCPM in PC-3M tumor could be significantly reduced after co-injection with an excess amount of TNYL-RAW peptide. Immunohistochemical analysis showed that fluorescence signal from the nanoparticles correlated with their radioactivity count, and co-localized with the EphB4 expressing region. (111)In-labeled TNYL-RAW-CCPM allowed visualization of cancer cells overexpressing EphB4 by both nuclear and optical techniques. The complementary information acquired with multiple imaging techniques should be advantageous in early detection of cancer.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Experimental Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Muto A, Yi T, Harrison KD, Dávalos A, Fancher TT, Ziegler KR, Feigel A, Kondo Y, Nishibe T, Sessa WC, Dardik A. Eph-B4 prevents venous adaptive remodeling in the adult arterial environment. J Exp Med 2011; 208:561-75. [PMID: 21339325 PMCID: PMC3058579 DOI: 10.1084/jem.20101854] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 01/21/2011] [Indexed: 01/08/2023] Open
Abstract
Eph-B4 determines mammalian venous differentiation in the embryo but is thought to be a quiescent marker of adult veins. We have previously shown that surgical transposition of a vein into the arterial environment is characterized by loss of venous identity, as indicated by the loss of Eph-B4, and intimal thickening. We used a mouse model of vein graft implantation to test the hypothesis that Eph-B4 is a critical determinant of venous wall thickness during postsurgical adaptation to the arterial environment. We show that stimulation of Eph-B4 signaling, either via ligand stimulation or expression of a constitutively active Eph-B4, inhibits venous wall thickening and preserves venous identity; conversely, reduction of Eph-B4 signaling is associated with increased venous wall thickness. Stimulated Eph-B4 associates with caveolin-1 (Cav-1); loss of Cav-1 or Eph-B4 kinase function abolishes inhibition of vein graft thickening. These results show that Eph-B4 is active in adult veins and regulates venous remodeling. Eph-B4-Cav-1-mediated vessel remodeling may be a venous-specific adaptive mechanism. Controlled stimulation of embryonic signaling pathways such as Eph-B4 may be a novel strategy to manipulate venous wall remodeling in adults.
Collapse
Affiliation(s)
- Akihito Muto
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Tai Yi
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Kenneth D. Harrison
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Alberto Dávalos
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Tiffany T. Fancher
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Kenneth R. Ziegler
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Amanda Feigel
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Yuka Kondo
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Toshiya Nishibe
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - William C. Sessa
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| | - Alan Dardik
- Department of Surgery and Department of Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
86
|
Kamouchi M, Ago T, Kitazono T. Brain pericytes: emerging concepts and functional roles in brain homeostasis. Cell Mol Neurobiol 2011; 31:175-93. [PMID: 21061157 PMCID: PMC11498428 DOI: 10.1007/s10571-010-9605-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 09/22/2010] [Indexed: 11/30/2022]
Abstract
Brain pericytes are an important constituent of neurovascular unit. They encircle endothelial cells and contribute to the maturation and stabilization of the capillaries in the brain. Recent studies have revealed that brain pericytes play pivotal roles in a variety of brain functions, such as regulation of capillary flow, angiogenesis, blood brain barrier, immune responses, and hemostasis. In addition, brain pericytes are pluripotent and can differentiate into different lineages similar to mesenchymal stem cells. The brain pericytes are revisited as a key player to maintain brain function and repair brain damage.
Collapse
Affiliation(s)
- Masahiro Kamouchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | |
Collapse
|
87
|
Xiong C, Huang M, Zhang R, Song S, Lu W, Flores L, Gelovani J, Li C. In Vivo Small-Animal PET/CT of EphB4 Receptors Using 64Cu-Labeled Peptide. J Nucl Med 2011; 52:241-8. [DOI: 10.2967/jnumed.110.081943] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
88
|
Duffy GP, D'Arcy S, Ahsan T, Nerem RM, O'Brien T, Barry F. Mesenchymal stem cells overexpressing ephrin-b2 rapidly adopt an early endothelial phenotype with simultaneous reduction of osteogenic potential. Tissue Eng Part A 2010; 16:2755-68. [PMID: 20491587 DOI: 10.1089/ten.tea.2009.0623] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Restoration of the vascular supply to ischemic tissues is of high clinical relevance, and proangiogenic therapies aim to reduce morbidity and mortality rates associated with the onset of cardiovascular disease. Stem cell therapy has been proposed as a potentially useful proangiogenic therapy. Mesenchymal stem cells (MSCs) have been shown to be proangiogenic and produce a number of cytokines involved in vessel development and maturation. Preclinical studies have reported increased angiogenesis after MSC delivery to the heart, and similar outcomes have been reported in recent clinical trials. Stem-cell-mediated neovascularization has been augmented by genetic modification with overexpression of angiogenic cytokines, including vascular endothelial growth factor (VEGF) and platelet-derived growth factor, showing promising results. In this study we aimed to enhance the proangiogenic capability of MSCs. MSCs were genetically modified to overexpress a versatile molecule, Ephrin-B2, involved in tissue morphogenesis and vascular development to enhance inherent neovascularization potential. Using nucleofection, Ephrin-B2 was transiently overexpressed on the cell surface of MSCs to recapitulate embryonic signaling and promote neovascularization. Ephrin-B2-expressing MSCs adopted an early endothelial phenotype under endothelial cell culture conditions increasing expression of von Willebrand factor and VEGF-Receptor 2. The cells had an increased ability to form vessel-like structures, produce VEGF, and incorporate into newly formed endothelial cell structures. These data indicate that MSCs expressing Ephrin-B2 represent a novel proangiogenic cell source to promote neovascularization in ischemic tissues.
Collapse
Affiliation(s)
- Garry P Duffy
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Ireland
| | | | | | | | | | | |
Collapse
|
89
|
Inhibitors of the tyrosine kinase EphB4. Part 3: Identification of non-benzodioxole-based kinase inhibitors. Bioorg Med Chem Lett 2010; 20:6242-5. [DOI: 10.1016/j.bmcl.2010.08.100] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 08/18/2010] [Accepted: 08/19/2010] [Indexed: 12/24/2022]
|
90
|
Welter M, Rieger H. Physical determinants of vascular network remodeling during tumor growth. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2010; 33:149-163. [PMID: 20607341 DOI: 10.1140/epje/i2010-10611-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Indexed: 05/29/2023]
Abstract
The process in which a growing tumor transforms a hierarchically organized arterio-venous blood vessel network into a tumor specific vasculature is analyzed with a theoretical model. The physical determinants of this remodeling involve the morphological and hydrodynamic properties of the initial network, generation of new vessels (sprouting angiogenesis), vessel dilation (circumferential growth), vessel regression, tumor cell proliferation and death, and the interdependence of these processes via spatio-temporal changes of blood flow parameters, oxygen/nutrient supply and growth factor concentration fields. The emerging tumor vasculature is non-hierarchical, compartmentalized into well-characterized zones, displays a complex geometry with necrotic zones and "hot spots" of increased vascular density and blood flow of varying size, and transports drug injections efficiently. Implications for current theoretical views on tumor-induced angiogenesis are discussed.
Collapse
Affiliation(s)
- M Welter
- Theoretical Physics, Saarland University, 66041, Saarbrücken, Germany
| | | |
Collapse
|
91
|
Andres AC, Djonov V. The mammary gland vasculature revisited. J Mammary Gland Biol Neoplasia 2010; 15:319-28. [PMID: 20706777 DOI: 10.1007/s10911-010-9186-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 08/04/2010] [Indexed: 12/11/2022] Open
Abstract
Concomitant with the extensive growth and differentiation of the mammary epithelium during pregnancy and lactation, and epithelial involution after weaning, the vasculature of the mammary gland undergoes repeated cycles of expansion and regression. Vascular expansion is effected by sprouting angiogenesis, intussusception and conceivably also vasculogenesis. The capacity of the epithelial cells to stimulate vascular growth and differentiation is dependent on the constellation of systemic and local hormones and growth factors as well as the changing demands for oxygenation and nutrient supply. This results in the release of angiogenic factors which stimulate endothelial cell growth and regulate vascular architecture. In contrast to the angiogenic phase of the mammary gland cycle, little is known about the control of vascular regression although this would possibly offer new insights into therapeutic possibilities against breast cancer. In this review we summarize knowledge regarding the mechanisms regulating the vasculature of the mammary gland and delineate the importance of the vasculature in the attainment of organ function. In addition, we discuss the angiogenic mechanisms observed during mammary carcinogenesis and their consequences for breast cancer therapy.
Collapse
Affiliation(s)
- Anne-Catherine Andres
- Department of Clinical Research, University of Bern, Tiefenaustrasse 120c, Bern, Switzerland.
| | | |
Collapse
|
92
|
Raza A, Franklin MJ, Dudek AZ. Pericytes and vessel maturation during tumor angiogenesis and metastasis. Am J Hematol 2010; 85:593-8. [PMID: 20540157 DOI: 10.1002/ajh.21745] [Citation(s) in RCA: 264] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Despite promising results in preclinical and clinical studies, the therapeutic efficacy of antiangiogenic therapies has been restricted by a narrow focus on inhibiting the growth of endothelial cells. Other cell types in the tumor stroma are also critical to the progression of cancer, including mural cells. Mural cells are vascular support cells that range in phenotype from pericytes to vascular smooth muscle cells. Although the role of pericytes and pericyte-like cells in the pathophysiology of cancer is still unclear, evidence indicates that aberrations in pericyte-endothelial cell signaling networks could contribute to tumor angiogenesis and metastasis. The purpose of this review is to evaluate critically recent evidence on the role of pericytes in tumor biology and discuss potential therapeutic targets for anticancer intervention.
Collapse
Affiliation(s)
- Ahmad Raza
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
93
|
Davies MH, Stempel AJ, Hubert KE, Powers MR. Altered vascular expression of EphrinB2 and EphB4 in a model of oxygen-induced retinopathy. Dev Dyn 2010; 239:1695-707. [PMID: 20503366 PMCID: PMC3095579 DOI: 10.1002/dvdy.22306] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
EphrinB2 ligands and EphB4 receptors are expressed on endothelial cells (EC) of arteries and veins, respectively, and are essential for vascular development. To understand how these molecules regulate retinal neovascularization (NV), we evaluated their expression in a model of oxygen-induced retinopathy (OIR). EphrinB2 and EphB4 were expressed on arterial and venous trunks, respectively, and on a subset of deep capillary vessels. EphB4 expression was reduced following hyperoxia, while ephrinB2 expression remained unaltered. In addition, a subset of EphB4-positive veins regressed in a caspase-3-dependent manner during hyperoxia. Arteriovenous malformations were also observed with loss of arterial-venous boundaries. Finally, both ephrinB2 and EphB4 were expressed on a subset of neovascular tufts following hyperoxia. These data confirm the contribution of ECs from both venous and arterial origins to the development of retinal NV.
Collapse
Affiliation(s)
- Michael H. Davies
- Department of Pediatrics, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
| | - Andrew J. Stempel
- Department of Pediatrics, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
| | - Kristin E. Hubert
- Department of Pediatrics, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
| | - Michael R. Powers
- Department of Pediatrics, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
94
|
Adams RH, Eichmann A. Axon guidance molecules in vascular patterning. Cold Spring Harb Perspect Biol 2010; 2:a001875. [PMID: 20452960 DOI: 10.1101/cshperspect.a001875] [Citation(s) in RCA: 299] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Endothelial cells (ECs) form extensive, highly branched and hierarchically organized tubular networks in vertebrates to ensure the proper distribution of molecular and cellular cargo in the vertebrate body. The growth of this vascular system during development, tissue repair or in disease conditions involves the sprouting, migration and proliferation of endothelial cells in a process termed angiogenesis. Surprisingly, specialized ECs, so-called tip cells, which lead and guide endothelial sprouts, share many feature with another guidance structure, the axonal growth cone. Tip cells are motile, invasive and extend numerous filopodial protrusions sensing growth factors, extracellular matrix and other attractive or repulsive cues in their tissue environment. Axonal growth cones and endothelial tip cells also respond to signals belonging to the same molecular families, such as Slits and Roundabouts, Netrins and UNC5 receptors, Semaphorins, Plexins and Neuropilins, and Eph receptors and ephrin ligands. Here we summarize fundamental principles of angiogenic growth, the selection and function of tip cells and the underlying regulation by guidance cues, the Notch pathway and vascular endothelial growth factor signaling.
Collapse
Affiliation(s)
- Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| | | |
Collapse
|
95
|
Eph receptors and ephrin ligands: important players in angiogenesis and tumor angiogenesis. JOURNAL OF ONCOLOGY 2010; 2010:135285. [PMID: 20224755 PMCID: PMC2836134 DOI: 10.1155/2010/135285] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 01/05/2010] [Indexed: 12/24/2022]
Abstract
Eph receptors and their ephrin ligands were identified in the late 1980's. Subsequently, they were linked to different physiological and pathophysiological processes like embryonic development, angiogenesis, and tumorigenesis. In this regard, recent work focused on the distribution and effects of Eph receptors and ephrins on tumor cells and tumor microenvironment. The purpose of this review is to outline the role of these molecules in physiological angiogenesis and pathophysiological tumor angiogenesis. Furthermore, novel therapeutical approaches are discussed as Eph receptors and ephrins represent attractive targets for antiangiogenic therapy.
Collapse
|
96
|
Abstract
The Eph receptor tyrosine kinases and their ephrin ligands have intriguing expression patterns in cancer cells and tumour blood vessels, which suggest important roles for their bidirectional signals in many aspects of cancer development and progression. Eph gene mutations probably also contribute to cancer pathogenesis. Eph receptors and ephrins have been shown to affect the growth, migration and invasion of cancer cells in culture as well as tumour growth, invasiveness, angiogenesis and metastasis in vivo. However, Eph signalling activities in cancer seem to be complex, and are characterized by puzzling dichotomies. Nevertheless, the Eph receptors are promising new therapeutic targets in cancer.
Collapse
Affiliation(s)
- Elena B Pasquale
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
97
|
Abstract
Guidance molecules were first described in the nervous system to control axon outgrowth direction. They are also widely expressed outside the nervous system where they control cell migration, tissue development and establishment of the vascular network. In addition, they are involved in cancer development, tumor angiogenesis and metastasis. This review is primarily focused on their functions in lung cancer and their involvement in lung development is also presented. Five guidance molecule families and their corresponding receptors are described, including the semaphorins/neuropilins/plexins, ephrins and Eph receptors, netrin/DCC/UNC5, Slit/Robo and Notch/Delta. In addition, the possibility to target these molecules as a therapeutic approach in cancer is discussed.
Collapse
Affiliation(s)
- Patrick Nasarre
- Medical University of South Carolina, Division of Hematology/Oncology, Charleston, SC, USA
| | | | | | | |
Collapse
|
98
|
Szczerba D, Kurz H, Szekely G. A computational model of intussusceptive microvascular growth and remodeling. J Theor Biol 2009; 261:570-83. [DOI: 10.1016/j.jtbi.2009.09.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 12/27/2022]
|
99
|
Paul R. Flow-correlated dilution of a regular network leads to a percolating network during tumor-induced angiogenesis. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2009; 30:101-114. [PMID: 19777279 DOI: 10.1140/epje/i2009-10513-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 06/24/2009] [Accepted: 09/02/2009] [Indexed: 05/28/2023]
Abstract
We study a simplified stochastic model for the vascularization of a growing tumor, incorporating the formation of new blood vessels at the tumor periphery as well as their regression in the tumor center. The resulting morphology of the tumor vasculature differs drastically from the original one. We demonstrate that the probabilistic vessel collapse has to be correlated with the blood shear force in order to yield percolating network structures. The resulting tumor vasculature displays fractal properties. Fractal dimension, Micro-Vascular Density (MVD), blood flow and shear force have been computed for a wide range of parameters.
Collapse
Affiliation(s)
- R Paul
- Department of Neurobiology, University of California, Davis, CA 95616, USA.
| |
Collapse
|
100
|
Weinsheimer S, Kim H, Pawlikowska L, Chen Y, Lawton MT, Sidney S, Kwok PY, McCulloch CE, Young WL. EPHB4 gene polymorphisms and risk of intracranial hemorrhage in patients with brain arteriovenous malformations. ACTA ACUST UNITED AC 2009; 2:476-82. [PMID: 20031623 DOI: 10.1161/circgenetics.109.883595] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Brain arteriovenous malformations (BAVMs) are a tangle of abnormal vessels directly shunting blood from the arterial to venous circulation and an important cause of intracranial hemorrhage (ICH). EphB4 is involved in arterial-venous determination during embryogenesis; altered signaling could lead to vascular instability resulting in ICH. We investigated the association of single-nucleotide polymorphisms (SNPs) and haplotypes in EPHB4 with risk of ICH at clinical presentation in patients with BAVM. METHODS AND RESULTS Eight haplotype-tagging SNPs spanning approximately 29 kb were tested for association with ICH presentation in 146 white patients with BAVM (phase I: 56 ICH, 90 non-ICH) using allelic, haplotypic, and principal components analysis. Associated SNPs were then genotyped in 102 additional cases (phase II: 37 ICH, 65 non-ICH), and data were combined for multivariable logistic regression. Minor alleles of 2 SNPs were associated with reduced risk of ICH presentation (rs314313_C, P=0.005; rs314308_T, P=0.0004). Overall, haplotypes were also significantly associated with ICH presentation (chi(2)=17.24, 6 df, P=0.008); 2 haplotypes containing the rs314308 T allele (GCCTGGGT, P=0.003; GTCTGGGC, P=0.036) were associated with reduced risk. In principal components analysis, 2 components explained 91% of the variance and complemented haplotype results by implicating 4 SNPs at the 5' end, including rs314308 and rs314313. These 2 SNPs were replicated in the phase II cohort, and combined data resulted in greater significance (rs314313, P=0.0007; rs314308, P=0.00008). SNP association with ICH presentation persisted after adjusting for age, sex, BAVM size, and deep venous drainage. CONCLUSIONS EPHB4 polymorphisms are associated with risk of ICH presentation in patients with BAVM, warranting further study.
Collapse
Affiliation(s)
- Shantel Weinsheimer
- Center for Cerebrovascular Research, University of California, San Francisco, Calif 94110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|