1
|
Mhlongo F, Cordero-Maldonado ML, Crawford AD, Katerere D, Sandasi M, Hattingh AC, Koekemoer TC, van de Venter M, Viljoen AM. Evaluation of the wound healing properties of South African medicinal plants using zebrafish and in vitro bioassays. JOURNAL OF ETHNOPHARMACOLOGY 2022; 286:114867. [PMID: 34822956 DOI: 10.1016/j.jep.2021.114867] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/02/2021] [Accepted: 11/21/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In South Africa, medicinal plants have a history of traditional use, with many species used for treating wounds. The scientific basis of such uses remains largely unexplored. AIM OF THE STUDY To screen South African plants used ethnomedicinally for wound healing based on their pro-angiogenic and wound healing activity, using transgenic zebrafish larvae and cell culture assays. MATERIALS AND METHODS South African medicinal plants used for wound healing were chosen according to literature. Dried plant material was extracted using six solvents of varying polarities. Pro-angiogenesis was assessed in vivo by observing morphological changes in sub-intestinal vessels after crude extract treatment of transgenic zebrafish larvae with vasculature-specific expression of a green fluorescent protein. Subsequently, the in vitro anti-inflammatory, fibroblast proliferation and collagen production effects of the plant extracts that were active in the zebrafish angiogenesis assay were investigated using murine macrophage (RAW 264.7) and human fibroblast (MRHF) cell lines. RESULTS Fourteen plants were extracted using six different solvents to yield 84 extracts and the non-toxic (n=72) were initially screened for pro-angiogenic activity in the zebrafish assay. Of these plant species, extracts of Lobostemon fruticosus, Scabiosa columbaria and Cotyledon orbiculata exhibited good activity in a concentration-dependent manner. All active extracts showed negligible in vitro toxicity using the MTT assay. Lobostemon fruticosus and Scabiosa columbaria extracts showed noteworthy anti-inflammatory activity in RAW 264.7 macrophages. The acetone extract of Lobostemon fruticosus stimulated the most collagen production at 122% above control values using the MRHF cell line, while all four of the selected extracts significantly stimulated cellular proliferation in vitro in the MRHF cell line. CONCLUSIONS The screening of the selected plant species provided valuable preliminary information validating the use of some of the plants in traditional medicine used for wound healing in South Africa. This study is the first to discover through an evidence-based pharmacology approach the wound healing properties of such plant species using the zebrafish as an in vivo model.
Collapse
Affiliation(s)
- Fikile Mhlongo
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | | | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine, Université du Luxembourg, Belval, Luxembourg; Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - David Katerere
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - Maxleene Sandasi
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa; SAMRC Herbal Drugs Research Unit, Tshwane University of Technology, Pretoria, South Africa
| | - Anna C Hattingh
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| | - Trevor C Koekemoer
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| | - Maryna van de Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| | - Alvaro M Viljoen
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa; SAMRC Herbal Drugs Research Unit, Tshwane University of Technology, Pretoria, South Africa.
| |
Collapse
|
2
|
Advancing Diabetic Retinopathy Research: Analysis of the Neurovascular Unit in Zebrafish. Cells 2021; 10:cells10061313. [PMID: 34070439 PMCID: PMC8228394 DOI: 10.3390/cells10061313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/30/2022] Open
Abstract
Diabetic retinopathy is one of the most important microvascular complications associated with diabetes mellitus, and a leading cause of vision loss or blindness worldwide. Hyperglycaemic conditions disrupt microvascular integrity at the level of the neurovascular unit. In recent years, zebrafish (Danio rerio) have come into focus as a model organism for various metabolic diseases such as diabetes. In both mammals and vertebrates, the anatomy and the function of the retina and the neurovascular unit have been highly conserved. In this review, we focus on the advances that have been made through studying pathologies associated with retinopathy in zebrafish models of diabetes. We discuss the different cell types that form the neurovascular unit, their role in diabetic retinopathy and how to study them in zebrafish. We then present new insights gained through zebrafish studies. The advantages of using zebrafish for diabetic retinopathy are summarised, including the fact that the zebrafish has, so far, provided the only animal model in which hyperglycaemia-induced retinal angiogenesis can be observed. Based on currently available data, we propose potential investigations that could advance the field further.
Collapse
|
3
|
Anti-angiogenic effects of VEGF stimulation on endothelium deficient in phosphoinositide recycling. Nat Commun 2020; 11:1204. [PMID: 32139674 PMCID: PMC7058007 DOI: 10.1038/s41467-020-14956-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/08/2020] [Indexed: 01/09/2023] Open
Abstract
Anti-angiogenic therapies have generated significant interest for their potential to combat tumor growth. However, tumor overproduction of pro-angiogenic ligands can overcome these therapies, hampering success of this approach. To circumvent this problem, we target the resynthesis of phosphoinositides consumed during intracellular transduction of pro-angiogenic signals in endothelial cells (EC), thus harnessing the tumor’s own production of excess stimulatory ligands to deplete adjacent ECs of the capacity to respond to these signals. Using zebrafish and human endothelial cells in vitro, we show ECs deficient in CDP-diacylglycerol synthase 2 are uniquely sensitive to increased vascular endothelial growth factor (VEGF) stimulation due to a reduced capacity to re-synthesize phosphoinositides, including phosphatidylinositol-(4,5)-bisphosphate (PIP2), resulting in VEGF-exacerbated defects in angiogenesis and angiogenic signaling. Using murine tumor allograft models, we show that systemic or EC specific suppression of phosphoinositide recycling results in reduced tumor growth and tumor angiogenesis. Our results suggest inhibition of phosphoinositide recycling provides a useful anti-angiogenic approach. Tumors can overproduce pro-angiogenic ligands overcoming currently approved anti-angiogenic therapies and hindering their success. Here, the authors show that targeting phosphoinositide recycling during tumor angiogenesis harnesses the tumor’s own production of angiogenic ligands to deplete adjacent endothelial cells of the capacity to respond to these signals.
Collapse
|
4
|
Galvin O, Srivastava A, Carroll O, Kulkarni R, Dykes S, Vickers S, Dickinson K, Reynolds AL, Kilty C, Redmond G, Jones R, Cheetham S, Pandit A, Kennedy BN. A sustained release formulation of novel quininib-hyaluronan microneedles inhibits angiogenesis and retinal vascular permeability in vivo. J Control Release 2016; 233:198-207. [DOI: 10.1016/j.jconrel.2016.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 12/17/2022]
|
5
|
The tetrapeptide Arg-Leu-Tyr-Glu inhibits VEGF-induced angiogenesis. Biochem Biophys Res Commun 2015; 463:532-7. [DOI: 10.1016/j.bbrc.2015.05.073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 05/22/2015] [Indexed: 11/17/2022]
|
6
|
A Heparan Sulfate-Binding Cell Penetrating Peptide for Tumor Targeting and Migration Inhibition. BIOMED RESEARCH INTERNATIONAL 2015; 2015:237969. [PMID: 26064887 PMCID: PMC4433633 DOI: 10.1155/2015/237969] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/31/2014] [Accepted: 11/14/2014] [Indexed: 11/18/2022]
Abstract
As heparan sulfate proteoglycans (HSPGs) are known as co-receptors to interact with numerous growth factors and then modulate downstream biological activities, overexpression of HS/HSPG on cell surface acts as an increasingly reliable prognostic factor in tumor progression. Cell penetrating peptides (CPPs) are short-chain peptides developed as functionalized vectors for delivery approaches of impermeable agents. On cell surface negatively charged HS provides the initial attachment of basic CPPs by electrostatic interaction, leading to multiple cellular effects. Here a functional peptide (CPPecp) has been identified from critical HS binding region in hRNase3, a unique RNase family member with in vitro antitumor activity. In this study we analyze a set of HS-binding CPPs derived from natural proteins including CPPecp. In addition to cellular binding and internalization, CPPecp demonstrated multiple functions including strong binding activity to tumor cell surface with higher HS expression, significant inhibitory effects on cancer cell migration, and suppression of angiogenesis in vitro and in vivo. Moreover, different from conventional highly basic CPPs, CPPecp facilitated magnetic nanoparticle to selectively target tumor site in vivo. Therefore, CPPecp could engage its capacity to be developed as biomaterials for diagnostic imaging agent, therapeutic supplement, or functionalized vector for drug delivery.
Collapse
|
7
|
Iman V, Karimian H, Mohan S, Hobani YH, Noordin MI, Mustafa MR, Noor SM. In vitro and in vivo anti-angiogenic activity of girinimbine isolated from Murraya koenigii. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1281-92. [PMID: 25767375 PMCID: PMC4354401 DOI: 10.2147/dddt.s71557] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Girinimbine is a carbazole alkaloid isolated from the stem bark and root of Murraya koenigii. Here we report that girinimbine is an inhibitor of angiogenic activity both in vitro and in vivo. MTT results showed that girinimbine inhibited proliferation of human umbilical vein endothelial cells, while results from endothelial cell invasion, migration, tube formation, and wound healing assays demonstrated significant time- and dose-dependent inhibition by girinimbine. A proteome profiler array done on girinimbine-treated human umbilical vein endothelial cells showed that girinimbine had mediated regulation of pro-angiogenic and anti-angiogenic proteins. The anti-angiogenic potential of girinimbine was also evidenced in vivo in the zebrafish embryo model wherein girinimbine inhibited neo vessel formation in zebrafish embryos following 24 hours of exposure. Together, these results showed that girinimbine could effectively suppress angiogenesis, suggestive of its therapeutic potential as a novel angiogenesis inhibitor.
Collapse
Affiliation(s)
- Venoos Iman
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hamed Karimian
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Syam Mohan
- Medical Research Center, University of Jazan, Jazan, Saudi Arabia
| | | | | | - Mohd Rais Mustafa
- Department of Pharmacology, Centre for Natural Products and Drug Discovery (CENAR), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Sasore T, Kennedy B. Deciphering combinations of PI3K/AKT/mTOR pathway drugs augmenting anti-angiogenic efficacy in vivo. PLoS One 2014; 9:e105280. [PMID: 25144531 PMCID: PMC4140730 DOI: 10.1371/journal.pone.0105280] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 07/23/2014] [Indexed: 12/27/2022] Open
Abstract
Ocular neovascularization is a common pathology associated with human eye diseases e.g. age-related macular degeneration and proliferative diabetic retinopathy. Blindness represents one of the most feared disabilities and remains a major burden to health-care systems. Current approaches to treat ocular neovascularisation include laser photocoagulation, photodynamic therapy and anti-VEGF therapies: Ranibizumab (Lucentis) and Aflibercept (Eylea). However, high clinical costs, frequent intraocular injections, and increased risk of infections are challenges related with these standards of care. Thus, there is a clinical need to develop more effective drugs that overcome these challenges. Here, we focus on an alternative approach by quantifying the in vivo anti-angiogenic efficacy of combinations of phosphatidylinositol-3-kinase (PI3K) pathway inhibitors. The PI3K/AKT/mTOR pathway is a complex signalling pathway involved in crucial cellular functions such as cell proliferation, migration and angiogenesis. RT-PCR confirms the expression of PI3K target genes (pik3ca, pik3r1, mtor and akt1) in zebrafish trunks from 6 hours post fertilisation (hpf) and in eyes from 2 days post fertilisation (dpf). Using both the zebrafish intersegmental vessel and hyaloid vessel assays to measure the in vivo anti-angiogenic efficacy of PI3K/Akt/mTOR pathway inhibitors, we identified 5 µM combinations of i) NVP-BEZ235 (dual PI3K-mTOR inhibitor) + PI-103 (dual PI3K-mTOR inhibitor); or ii) LY-294002 (pan-PI3K inhibitor) + NVP-BEZ235; or iii) NVP-BEZ235 + rapamycin (mTOR inhibitor); or iv) LY-294002 + rapamycin as the most anti-angiogenic. Treatment of developing larvae from 2–5 dpf with 5 µM NVP-BEZ235 plus PI-103 resulted in an essentially intact ocular morphology and visual behaviour, whereas other combinations severely disrupted the developing retinal morphology and visual function. In human ARPE19 retinal pigment epithelium cells, however, no significant difference in cell number was observed following treatment with the inhibitor combinations. Collectively, these results highlight the potential of combinations of PI3K/AKT/mTOR pathway inhibitors to safely and effectively treat ocular neovascularization.
Collapse
Affiliation(s)
- Temitope Sasore
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Breandán Kennedy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
9
|
Park H, Shim JS, Kim BS, Jung HJ, Huh TL, Kwon HJ. Purpurin inhibits adipocyte-derived leucine aminopeptidase and angiogenesis in a zebrafish model. Biochem Biophys Res Commun 2014; 450:561-7. [PMID: 24928393 DOI: 10.1016/j.bbrc.2014.06.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 06/03/2014] [Indexed: 12/30/2022]
Abstract
Adipocyte-derived leucine aminopeptidase (A-LAP) is a novel member of the M1 family of zinc metallopeptidases, which has been reported to play a crucial role in angiogenesis. In the present study, we conducted a target-based screening of natural products and synthetic chemical libraries using the purified enzyme to search novel inhibitors of A-LAP. Amongst several hits isolated, a natural product purpurin was identified as one of the most potent inhibitors of A-LAP from the screening. In vitro enzymatic analyses demonstrated that purpurin inhibited A-LAP activity in a non-competitive manner with a Ki value of 20 M. In addition, purpurin showed a strong selectivity toward A-LAP versus another member of M1 family of zinc metallopeptidase, aminopeptidase N (APN). In angiogenesis assays, purpurin inhibited the vascular endothelial growth factor (VEGF)-induced invasion and tube formation of human umbilical vein endothelial cells (HUVEC). Moreover, purpurin inhibited in vivo angiogenesis in zebrafish embryo without toxicity. These data demonstrate that purpurin is a novel specific inhibitor of A-LAP and could be developed as a new anti-angiogenic agent.
Collapse
Affiliation(s)
- Hyomi Park
- Chemical Genomics National Research Laboratory, Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Joong Sup Shim
- Faculty of Health Sciences, University of Macau, Av. Universidade, Taipa, Macau Special Administrative Region, China
| | - Beom Seok Kim
- Chemical Genomics National Research Laboratory, Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Hye Jin Jung
- Department of Pharmaceutical Engineering, University of Sun Moon, Asansi, Chungnam 336-708, Republic of Korea
| | - Tae-Lin Huh
- Department of Genetic Engineering, Kyungpook National University, Puk-Gu, Sankyuk-Dong, 702-701 Daegu, Republic of Korea
| | - Ho Jeong Kwon
- Chemical Genomics National Research Laboratory, Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea.
| |
Collapse
|
10
|
Integrated chip-based physiometer for automated fish embryo toxicity biotests in pharmaceutical screening and ecotoxicology. Cytometry A 2014; 85:537-47. [DOI: 10.1002/cyto.a.22464] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 12/29/2022]
|
11
|
Akagi J, Hall CJ, Crosier KE, Cooper JM, Crosier PS, Wlodkowic D. OpenSource lab-on-a-chip physiometer for accelerated zebrafish embryo biotests. ACTA ACUST UNITED AC 2014; 67:9.44.1-9.44.16. [PMID: 24510773 DOI: 10.1002/0471142956.cy0944s67] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Zebrafish (Danio rerio) embryo assays have recently come into the spotlight as convenient experimental models in both biomedicine and ecotoxicology. As a small aquatic model organism, zebrafish embryo assays allow for rapid physiological, embryo-, and genotoxic tests of drugs and environmental toxins that can be simply dissolved in water. This protocol describes prototyping and application of an innovative, miniaturized, and polymeric chip-based device capable of immobilizing a large number of living fish embryos for real-time and/or time-lapse microscopic examination. The device provides a physical address designation to each embryo during analysis, continuous perfusion of medium, and post-analysis specimen recovery. Miniaturized embryo array is a new concept of immobilization and real-time drug perfusion of multiple individual and developing zebrafish embryos inside the mesofluidic device. The OpenSource device presented in this protocol is particularly suitable to perform accelerated fish embryo biotests in ecotoxicology and phenotype-based pharmaceutical screening.
Collapse
Affiliation(s)
- Jin Akagi
- The OpenTech Factory, School of Applied Sciences, RMIT University, Melbourne, Australia
| | - Chris J Hall
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn E Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Jonathan M Cooper
- School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Donald Wlodkowic
- The OpenTech Factory, School of Applied Sciences, RMIT University, Melbourne, Australia
| |
Collapse
|
12
|
Characterization of zebrafish von Willebrand factor reveals conservation of domain structure, multimerization, and intracellular storage. Adv Hematol 2012; 2012:214209. [PMID: 23049555 PMCID: PMC3462383 DOI: 10.1155/2012/214209] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/18/2012] [Accepted: 07/26/2012] [Indexed: 11/24/2022] Open
Abstract
von Willebrand disease (VWD) is the most common inherited human bleeding disorder and is caused by quantitative or qualitative defects in von Willebrand factor (VWF). VWF is a secreted glycoprotein that circulates as large multimers. While reduced VWF is associated with bleeding, elevations in overall level or multimer size are implicated in thrombosis. The zebrafish is a powerful genetic model in which the hemostatic system is well conserved with mammals. The ability of this organism to generate thousands of offspring and its optical transparency make it unique and complementary to mammalian models of hemostasis. Previously, partial clones of zebrafish vwf have been identified, and some functional conservation has been demonstrated. In this paper we clone the complete zebrafish vwf cDNA and show that there is conservation of domain structure. Recombinant zebrafish Vwf forms large multimers and pseudo-Weibel-Palade bodies (WPBs) in cell culture. Larval expression is in the pharyngeal arches, yolk sac, and intestinal epithelium. These results provide a foundation for continued study of zebrafish Vwf that may further our understanding of the mechanisms of VWD.
Collapse
|
13
|
Izumi N, Helker C, Ehling M, Behrens A, Herzog W, Adams RH. Fbxw7 controls angiogenesis by regulating endothelial Notch activity. PLoS One 2012; 7:e41116. [PMID: 22848434 PMCID: PMC3407154 DOI: 10.1371/journal.pone.0041116] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 06/18/2012] [Indexed: 11/18/2022] Open
Abstract
Notch signaling controls fundamental aspects of angiogenic blood vessel growth including the selection of sprouting tip cells, endothelial proliferation and arterial differentiation. The E3 ubiquitin ligase Fbxw7 is part of the SCF protein complex responsible for the polyubiquitination and thereby proteasomal degradation of substrates such as Notch, c-Myc and c-Jun. Here, we show that Fbxw7 is a critical regulator of angiogenesis in the mouse retina and the zebrafish embryonic trunk, which we attribute to its role in the degradation of active Notch. Growth of retinal blood vessel was impaired and the Notch ligand Dll4, which is also a Notch target, upregulated in inducible and endothelial cell-specific Fbxw7(iECKO) mutant mice. The stability of the cleaved and active Notch intracellular domain was increased after siRNA knockdown of the E3 ligase in cultured human endothelial cells. Injection of fbxw7 morpholinos interfered with the sprouting of zebrafish intersegmental vessels (ISVs). Arguing strongly that Notch and not other Fbxw7 substrates are primarily responsible for these phenotypes, the genetic inactivation of Notch pathway components reversed the impaired ISV growth in the zebrafish embryo as well as sprouting and proliferation in the mouse retina. Our findings establish that Fbxw7 is a potent positive regulator of angiogenesis that limits the activity of Notch in the endothelium of the growing vasculature.
Collapse
Affiliation(s)
- Nanae Izumi
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Muenster, Germany
| | - Christian Helker
- University of Muenster, Faculty of Biology, and Max-Planck-Institute for Molecular Biomedicine, Angiogenesis Laboratory, Muenster, Germany
| | - Manuel Ehling
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Muenster, Germany
| | - Axel Behrens
- Mammalian Genetics Laboratory, CRUK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom
| | - Wiebke Herzog
- University of Muenster, Faculty of Biology, and Max-Planck-Institute for Molecular Biomedicine, Angiogenesis Laboratory, Muenster, Germany
| | - Ralf H. Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Muenster, Germany
| |
Collapse
|
14
|
Lai SL, Cheah SC, Wong PF, Noor SM, Mustafa MR. In vitro and in vivo anti-angiogenic activities of Panduratin A. PLoS One 2012; 7:e38103. [PMID: 22666456 PMCID: PMC3364190 DOI: 10.1371/journal.pone.0038103] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Targeting angiogenesis has emerged as an attractive and promising strategy in anti-cancer therapeutic development. The present study investigates the anti-angiogenic potential of Panduratin A (PA), a natural chalcone isolated from Boesenbergia rotunda by using both in vitro and in vivo assays. METHODOLOGY/PRINCIPAL FINDINGS PA exerted selective cytotoxicity on human umbilical vein endothelial cells (HUVECs) with IC(50) value of 6.91 ± 0.85 µM when compared to human normal fibroblast and normal liver epithelial cells. Assessment of the growth kinetics by cell impedance-based Real-Time Cell Analyzer showed that PA induced both cytotoxic and cytostatic effects on HUVECs, depending on the concentration used. Results also showed that PA suppressed VEGF-induced survival and proliferation of HUVECs. Furthermore, endothelial cell migration, invasion, and morphogenesis or tube formation demonstrated significant time- and dose-dependent inhibition by PA. PA also suppressed matrix metalloproteinase-2 (MMP-2) secretion and attenuated its activation to intermediate and active MMP-2. In addition, PA suppressed F-actin stress fiber formation to prevent migration of the endothelial cells. More importantly, anti-angiogenic potential of PA was also evidenced in two in vivo models. PA inhibited neo-vessels formation in murine Matrigel plugs, and angiogenesis in zebrafish embryos. CONCLUSIONS/SIGNIFICANCE Taken together, our study demonstrated the distinctive anti-angiogenic properties of PA, both in vitro and in vivo. This report thus reveals another biological activity of PA in addition to its reported anti-inflammatory and anti-cancer activities, suggestive of PA's potential for development as an anti-angiogenic agent for cancer therapy.
Collapse
Affiliation(s)
- Siew-Li Lai
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohd Rais Mustafa
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
15
|
Akagi J, Khoshmanesh K, Evans B, Hall CJ, Crosier KE, Cooper JM, Crosier PS, Wlodkowic D. Miniaturized embryo array for automated trapping, immobilization and microperfusion of zebrafish embryos. PLoS One 2012; 7:e36630. [PMID: 22606275 PMCID: PMC3351474 DOI: 10.1371/journal.pone.0036630] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 04/04/2012] [Indexed: 11/28/2022] Open
Abstract
Zebrafish (Danio rerio) has recently emerged as a powerful experimental model in drug discovery and environmental toxicology. Drug discovery screens performed on zebrafish embryos mirror with a high level of accuracy the tests usually performed on mammalian animal models, and fish embryo toxicity assay (FET) is one of the most promising alternative approaches to acute ecotoxicity testing with adult fish. Notwithstanding this, automated in-situ analysis of zebrafish embryos is still deeply in its infancy. This is mostly due to the inherent limitations of conventional techniques and the fact that metazoan organisms are not easily susceptible to laboratory automation. In this work, we describe the development of an innovative miniaturized chip-based device for the in-situ analysis of zebrafish embryos. We present evidence that automatic, hydrodynamic positioning, trapping and long-term immobilization of single embryos inside the microfluidic chips can be combined with time-lapse imaging to provide real-time developmental analysis. Our platform, fabricated using biocompatible polymer molding technology, enables rapid trapping of embryos in low shear stress zones, uniform drug microperfusion and high-resolution imaging without the need of manual embryo handling at various developmental stages. The device provides a highly controllable fluidic microenvironment and post-analysis eleuthero-embryo stage recovery. Throughout the incubation, the position of individual embryos is registered. Importantly, we also for first time show that microfluidic embryo array technology can be effectively used for the analysis of anti-angiogenic compounds using transgenic zebrafish line (fli1a:EGFP). The work provides a new rationale for rapid and automated manipulation and analysis of developing zebrafish embryos at a large scale.
Collapse
Affiliation(s)
- Jin Akagi
- The BioMEMS Research Group, School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Khashayar Khoshmanesh
- The BioMEMS Research Group, School of Chemical Sciences, University of Auckland, Auckland, New Zealand
- School of Electrical and Computer Engineering, RMIT University, Melbourne, Australia
| | - Barbara Evans
- The BioMEMS Research Group, School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Chris J. Hall
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn E. Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | | | - Philip S. Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Donald Wlodkowic
- The BioMEMS Research Group, School of Chemical Sciences, University of Auckland, Auckland, New Zealand
- School of Applied Sciences, RMIT University, Melbourne, Australia
| |
Collapse
|
16
|
Identification of phosphorylase kinase as a novel therapeutic target through high-throughput screening for anti-angiogenesis compounds in zebrafish. Oncogene 2011; 31:4333-42. [PMID: 22179836 DOI: 10.1038/onc.2011.594] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Angiogenesis is essential for development and tumor progression. With the aim of identifying new compound inhibitors of the angiogenesis process, we used an established enhanced green fluorescent protein-transgenic zebrafish line to develop an automated assay that enables high-throughput screening of compound libraries in a whole-organism setting. Using this system, we have identified novel kinase inhibitor compounds that show anti-angiogenic properties in both zebrafish in-vivo system and in human endothelial cell in-vitro angiogenesis models. Furthermore, we have determined the kinase target of these compounds and have identified and validated a previously uncharacterized involvement of phosphorylase kinase subunit G1 (PhKG1) in angiogenesis in vivo. In addition, we have found that PhKG1 is upregulated in human tumor samples and that aberrations in gene copy number of PhK subunits are a common feature of human tumors. Our results provide a novel insight into the angiogenesis process, as well as identify new potential targets for anti-angiogenic therapies.
Collapse
|
17
|
Beliaeva NF, Kashirtseva VN, Medvedeva NV, Khudoklinova II, Ipatova OM, Archakov AI. [Zebrafish as a model organism for biomedical studies]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2011; 56:120-31. [PMID: 21328916 DOI: 10.18097/pbmc20105601120] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Zebrafish (Danio rerio) are now firmly established as a powerful research model for many areas of biology and medicine. Here, we review some achievements of zebrafish-based assays for modeling human diseases and for drug discovery and development. For drug discovery, zebrafish are especially valuable in the earlier stages of research as they provide a model organism to demonstrate a new treatment's efficacy and toxicity before more costly mammalian models are used. This review provides examples of compounds known to be toxic to humans that have been demonstrated to functional similarly in zebrafish. Major advantages of zebrafish embryons are that they are readily permeable to small molecules added to their incubation medium and the transparent chorion enables the easy observation of development. Assay of acute toxicity (LC50 estimation) in embryos can also include the screening for developmental disorders as an indicator of teratogenic effects. We used zebrafish for toxicity testing of new drugs on the base of phospholipid nanoparticles. The organization of the genome and the pathways controlling signal transduction appear to be highly conserved between zebrafish and humans that allow using zebrafish for modeling of human diseases some examples of which are illustrated in this paper.
Collapse
|
18
|
Crawford AD, Liekens S, Kamuhabwa AR, Maes J, Munck S, Busson R, Rozenski J, Esguerra CV, de Witte PAM. Zebrafish bioassay-guided natural product discovery: isolation of angiogenesis inhibitors from East African medicinal plants. PLoS One 2011; 6:e14694. [PMID: 21379387 PMCID: PMC3040759 DOI: 10.1371/journal.pone.0014694] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 09/12/2010] [Indexed: 01/10/2023] Open
Abstract
Natural products represent a significant reservoir of unexplored chemical diversity for early-stage drug discovery. The identification of lead compounds of natural origin would benefit from therapeutically relevant bioassays capable of facilitating the isolation of bioactive molecules from multi-constituent extracts. Towards this end, we developed an in vivo bioassay-guided isolation approach for natural product discovery that combines bioactivity screening in zebrafish embryos with rapid fractionation by analytical thin-layer chromatography (TLC) and initial structural elucidation by high-resolution electrospray mass spectrometry (HRESIMS). Bioactivity screening of East African medicinal plant extracts using fli-1:EGFP transgenic zebrafish embryos identified Oxygonum sinuatum and Plectranthus barbatus as inhibiting vascular development. Zebrafish bioassay-guided fractionation identified the active components of these plants as emodin, an inhibitor of the protein kinase CK2, and coleon A lactone, a rare abietane diterpenoid with no previously described bioactivity. Both emodin and coleon A lactone inhibited mammalian endothelial cell proliferation, migration, and tube formation in vitro, as well as angiogenesis in the chick chorioallantoic membrane (CAM) assay. These results suggest that the combination of zebrafish bioassays with analytical chromatography methods is an effective strategy for the rapid identification of bioactive natural products.
Collapse
Affiliation(s)
| | - Sandra Liekens
- Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
- Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Appolinary R. Kamuhabwa
- Department of Pharmacognosy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Jan Maes
- Department of Pharmaceutical Sciences, University of Leuven, Leuven, Belgium
| | - Sebastian Munck
- Department of Human Genetics, Flanders Interuniversity Institute of Biotechnology, University of Leuven, Leuven, Belgium
| | - Roger Busson
- Department of Pharmaceutical Sciences, University of Leuven, Leuven, Belgium
- Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Jef Rozenski
- Department of Pharmaceutical Sciences, University of Leuven, Leuven, Belgium
- Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Camila V. Esguerra
- Department of Pharmaceutical Sciences, University of Leuven, Leuven, Belgium
| | | |
Collapse
|
19
|
Belyaeva NF, Kashirtseva VN, Medvedeva NV, Khudoklinova YY, Ipatova OM, Archakov AI. Zebrafish as a model system for biomedical studies. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2009. [DOI: 10.1134/s1990750809040039] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Harfouche R, Hentschel DM, Piecewicz S, Basu S, Print C, Eavarone D, Kiziltepe T, Sasisekharan R, Sengupta S. Glycome and transcriptome regulation of vasculogenesis. Circulation 2009; 120:1883-1892. [PMID: 19858418 PMCID: PMC4001715 DOI: 10.1161/circulationaha.108.837724] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Therapeutic vasculogenesis is an emerging concept that can potentially be harnessed for the management of ischemic pathologies. The present study elucidates the potential coregulation of vasculogenesis by the heparan sulfate glycosaminoglycan-rich cell-surface glycome and the transcriptome. METHODS AND RESULTS Differentiation of embryonic stem cells into endothelial cells in an in vitro embryoid body is paralleled by an amplification of heparan sulfate glycosaminoglycan sulfation, which correlates with the levels of the enzyme N-deacetylase/N-sulfotransferase 1 (NDST1). Small hairpin RNA-mediated knockdown of NDST1 or modification of heparan sulfate glycosaminoglycans in embryonic stem cells with heparinases or sodium chlorate inhibited differentiation of embryonic stem cells into endothelial cells. This was translated to an in vivo zebrafish embryo model, in which the genetic knockdown of NDST1 resulted in impaired vascularization characterized by a concentration-dependent decrease in intersegmental vessel lumen and a large tail-vessel configuration, which could be rescued by use of exogenous sulfated heparan sulfate glycosaminoglycans. To explore the cross talk between the glycome and the transcriptome during vasculogenesis, we identified by microarray and then validated wild-type and NDST1 knockdown-associated gene-expression patterns in zebrafish embryos. Temporal analysis at 3 developmental stages critical for vasculogenesis revealed a cascade of pathways that may mediate glycocalyx regulation of vasculogenesis. These pathways were intimately connected to cell signaling, cell survival, and cell fate determination. Specifically, we demonstrated that forkhead box O3A/5 proteins and insulin-like growth factor were key downstream signals in this process. CONCLUSIONS The present study for the first time implicates interplay between the glycome and the transcriptome during vasculogenesis, revealing the possibility of harnessing specific cellular glyco-microenvironments for therapeutic vascularization.
Collapse
Affiliation(s)
- Rania Harfouche
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - Dirk M Hentschel
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - Stephanie Piecewicz
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - Sudipta Basu
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - Cristin Print
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - David Eavarone
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - Tanyel Kiziltepe
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - Ram Sasisekharan
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| | - Shiladitya Sengupta
- Department of Medicine (R.H., D.M.H., S.P., S.B., S.S.), Brigham and Women's Hospital, Boston, Mass; Department of Molecular Medicine and Pathology (C.P.), University of Auckland, Auckland, New Zealand; Department of Biological Engineering (D.E., T.K., R.S.), Massachusetts Institute of Technology, Cambridge, Mass; and Harvard-MIT Division of Health Sciences and Technology (R.H., S.P., S.B., D.E., T.K., R.S., S.S.), Cambridge, Mass
| |
Collapse
|
21
|
Stoletov K, Fang L, Choi SH, Hartvigsen K, Hansen LF, Hall C, Pattison J, Juliano J, Miller ER, Almazan F, Crosier P, Witztum JL, Klemke RL, Miller YI. Vascular lipid accumulation, lipoprotein oxidation, and macrophage lipid uptake in hypercholesterolemic zebrafish. Circ Res 2009; 104:952-60. [PMID: 19265037 PMCID: PMC2834250 DOI: 10.1161/circresaha.108.189803] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lipid accumulation in arteries induces vascular inflammation and atherosclerosis, the major cause of heart attack and stroke in humans. Extreme hyperlipidemia induced in mice and rabbits enables modeling many aspects of human atherosclerosis, but microscopic examination of plaques is possible only postmortem. Here we report that feeding adult zebrafish (Danio rerio) a high-cholesterol diet (HCD) resulted in hypercholesterolemia, remarkable lipoprotein oxidation, and fatty streak formation in the arteries. Feeding an HCD supplemented with a fluorescent cholesteryl ester to optically transparent fli1:EGFP zebrafish larvae in which endothelial cells express green fluorescent protein (GFP), and using confocal microscopy enabled monitoring vascular lipid accumulation and the endothelial cell layer disorganization and thickening in a live animal. The HCD feeding also increased leakage of a fluorescent dextran from the blood vessels. Administering ezetimibe significantly diminished the HCD-induced endothelial cell layer thickening and improved its barrier function. Feeding HCD to lyz:DsRed2 larvae in which macrophages and granulocytes express DsRed resulted in the accumulation of fluorescent myeloid cells in the vascular wall. Using a fluorogenic substrate for phospholipase A(2) (PLA(2)), we observed an increased vascular PLA(2) activity in live HCD-fed larvae compared to control larvae. Furthermore, by transplanting genetically modified murine cells into HCD-fed larvae, we demonstrated that toll-like receptor-4 was required for efficient in vivo lipid uptake by macrophages. These results suggest that the novel zebrafish model is suitable for studying temporal characteristics of certain inflammatory processes of early atherogenesis and the in vivo function of vascular cells.
Collapse
Affiliation(s)
- Konstantin Stoletov
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Longhou Fang
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Karsten Hartvigsen
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Lotte F. Hansen
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Chris Hall
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Jennifer Pattison
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Joseph Juliano
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Elizabeth R. Miller
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Felicidad Almazan
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Phil Crosier
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Joseph L. Witztum
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Richard L. Klemke
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
22
|
Raghunath M, Sy Wong Y, Farooq M, Ge R. Pharmacologically induced angiogenesis in transgenic zebrafish. Biochem Biophys Res Commun 2009; 378:766-71. [DOI: 10.1016/j.bbrc.2008.11.127] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 11/21/2008] [Indexed: 01/08/2023]
|
23
|
Moorman SJ, Shorr AZ. The primary cilium as a gravitational force transducer and a regulator of transcriptional noise. Dev Dyn 2008; 237:1955-9. [PMID: 18366139 DOI: 10.1002/dvdy.21493] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Circumstantial evidence has suggested that the primary cilium might function as a gravity sensor. Direct evidence of its gravity-sensing function has recently been provided by studies of rohon beard neurons. These neurons showed changes in the variability of gene expression levels that are linked to the cyclic changes in the Earth's gravitational field due to the Sun and Moon. These cyclic changes also cause the tides. Rohon beard neurons, after the primary cilia have been selectively destroyed, no longer show changes in gene expression variability linked to the cyclic changes in Earth's gravitational field. After the neurons regrow their primary cilia, the link between variability in gene expression levels and the Earth's changing gravitational field returns. This suggests two new functions for the primary cilia, detecting the cyclical changes in the Earth's gravitational field and transducing those changes into changes in the variability (stochastic nature) of gene expression.
Collapse
Affiliation(s)
- Stephen J Moorman
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA.
| | | |
Collapse
|
24
|
Eyries M, Siegfried G, Ciumas M, Montagne K, Agrapart M, Lebrin F, Soubrier F. Hypoxia-induced apelin expression regulates endothelial cell proliferation and regenerative angiogenesis. Circ Res 2008; 103:432-40. [PMID: 18617693 DOI: 10.1161/circresaha.108.179333] [Citation(s) in RCA: 199] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Apelin has been identified as the endogenous ligand of the human orphan G protein-coupled receptor APJ. This peptide exerts a variety of cardiovascular effects and particularly acts as an activator of angiogenesis. Importantly, hypoxia has been reported to regulate apelin expression but the molecular mechanism underlying hypoxia-induced apelin expression and the relationship with the physiological response of the apelin/APJ system are still not established. Here, we demonstrate that apelin expression is induced by hypoxia in cultured endothelial and vascular smooth muscle cells as well as in lung from mice exposed to acute hypoxia. Transient transfection experiments show that hypoxia-inducible transcriptional activation of apelin requires an intact hypoxia-responsive element (+813/+826) located within the first intron of the human apelin gene. Chromatin immunoprecipitation assay reveals that hypoxia-inducible factor-1alpha binds to the endogenous hypoxia-responsive element site of the apelin gene. Moreover, overexpression of hypoxia-inducible factor-1alpha increases the transcriptional activity of a reporter construct containing this hypoxia-responsive element, whereas small interfering RNA-mediated hypoxia-inducible factor-1alpha knockdown abolishes hypoxia-induced apelin expression. Finally, microinterfering RNA-mediated apelin or APJ receptor knockdown inhibits both hypoxia-induced endothelial cell proliferation in vitro and hypoxia-induced vessel regeneration in the caudal fin regeneration of Fli-1 transgenic zebrafish. The hypoxia-induced apelin expression may, thus, provide a new mechanism involved in adaptive physiological and pathophysiological response of vascular cells to low oxygen level.
Collapse
Affiliation(s)
- Mélanie Eyries
- INSERM UMRS 525, Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | | | |
Collapse
|
25
|
Baldessari D, Mione M. How to create the vascular tree? (Latest) help from the zebrafish. Pharmacol Ther 2008; 118:206-30. [PMID: 18439684 DOI: 10.1016/j.pharmthera.2008.02.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 02/19/2008] [Indexed: 12/22/2022]
Abstract
The cardiovascular system provides oxygen, nutrients and hormones to organs, it directs traffic of metabolites and it maintains tissue homeostasis. It is one of the first organs assembled during vertebrate development and it is essential to life from early stages to adult. For these reasons, the process of vessel formation has being studied for more than a century, but it is only in the late eighties that there has been an explosion of research in the field with the employment of various in vitro and in vivo model systems. The zebrafish (Danio rerio) offers several advantages for in vivo studies; it played a fundamental role in new discoveries and helped to refine our knowledge of the vascular system. This review recapitulates the zebrafish data on vasculogenesis and angiogenesis, including the specification of the haemangioblasts from the mesoderm, their migration to form the vascular cord followed by axial vessels specification, the primary and secondary sprouting of intersomitic vessels, the formation of the lumen, the arterial versus venous specification and patterning. To emphasize the strengths of the zebrafish system in the vascular field, we summarize main tools, such as gene expression and mutagenesis screens, knock down technologies, transgenic lines and imaging, which played a major role in the development of the field and allowed significant discoveries, for instance the recent visualization of the lymphatic system in zebrafish. This information contributes to the prospective of drug discovery to cure human diseases linked to angiogenesis, not last tumours.
Collapse
Affiliation(s)
- Danila Baldessari
- IFOM-IEO Campus (FIRC Institute of Molecular Oncology Foundation-European Institute of Oncology), Via Adamello 16, 20139 Milan, Italy.
| | | |
Collapse
|
26
|
Esengil H, Chen JK. Gene regulation technologies in zebrafish. MOLECULAR BIOSYSTEMS 2008; 4:300-8. [DOI: 10.1039/b718447f] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
27
|
Schmidt M, Paes K, De Mazière A, Smyczek T, Yang S, Gray A, French D, Kasman I, Klumperman J, Rice DS, Ye W. EGFL7 regulates the collective migration of endothelial cells by restricting their spatial distribution. Development 2007; 134:2913-23. [PMID: 17626061 DOI: 10.1242/dev.002576] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
During sprouting angiogenesis, groups of endothelial cells (ECs) migrate together in units called sprouts. In this study, we demonstrate that the vascular-specific secreted factor EGFL7 regulates the proper spatial organization of ECs within each sprout and influences their collective movement. In the homozygous Egfl7-knockout mice, vascular development is delayed in many organs despite normal EC proliferation, and 50% of the knockout embryos die in utero. ECs in the mutant vasculatures form abnormal aggregates and the vascular basement membrane marker collagen IV is mislocalized, suggesting that ECs fail to recognize the proper spatial position of their neighbors. Although the migratory ability of individual ECs in isolation is not affected by the loss of EGFL7, the aberrant spatial organization of ECs in the mutant tissues decreases their collective movement. Using in vitro and in vivo analyses, we showed that EGFL7 is a component of the interstitial extracellular matrix deposited on the basal sides of sprouts, a location suitable for conveying positional information to neighboring ECs. Taken together, we propose that EGFL7 defines the optimal path of EC movement by assuring the correct positioning of each EC in a nascent sprout.
Collapse
Affiliation(s)
- Maike Schmidt
- Tumor Biology and Angiogenesis Department, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wu X, Zhong H, Song J, Damoiseaux R, Yang Z, Lin S. Mycophenolic Acid Is a Potent Inhibitor of Angiogenesis. Arterioscler Thromb Vasc Biol 2006; 26:2414-6. [PMID: 16990565 DOI: 10.1161/01.atv.0000238361.07225.fc] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Hu G, Tang J, Zhang B, Lin Y, Hanai JI, Galloway J, Bedell V, Bahary N, Han Z, Ramchandran R, Thisse B, Thisse C, Zon LI, Sukhatme VP. A novel endothelial-specific heat shock protein HspA12B is required in both zebrafish development and endothelial functions in vitro. J Cell Sci 2006; 119:4117-26. [PMID: 16968741 DOI: 10.1242/jcs.03179] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A zebrafish transcript dubbed GA2692 was initially identified via a whole-mount in situ hybridization screen for vessel specific transcripts. Its mRNA expression during embryonic development was detected in ventral hematopoietic and vasculogenic mesoderm and later throughout the vasculature up to 48 hours post fertilization. Morpholino-mediated knockdown of GA2692 in embryos resulted in multiple defects in vasculature, particularly, at sites undergoing active capillary sprouting: the intersegmental vessels, sub-intestinal vessels and the capillary sprouts of the pectoral fin vessel. During the course of these studies, a homology search indicated that GA2692 is the zebrafish orthologue of mammalian HspA12B, a distant member of the heat shock protein 70 (Hsp70) family. By a combination of northern blot and real-time PCR analysis, we showed that HspA12B is highly expressed in human endothelial cells in vitro. Knockdown of HspA12B by small interfering RNAs (siRNAs) in human umbilical vein endothelial cells blocked wound healing, migration and tube formation, whereas overexpression of HspA12B enhanced migration and accelerated wound healing - data that are consistent with the in vivo fish phenotype obtained in the morpholino-knockdown studies. Phosphorylation of Akt was consistently reduced by siRNAs against HspA12B. Overexpression of a constitutively active form of Akt rescued the inhibitory effects of knockdown of HspA12B on migration of human umbilical vein endothelial cells. Collectively, our data suggests that HspA12B is a highly endothelial-cell-specific distant member of the Hsp70 family and plays a significant role in endothelial cells during development and angiogenesis in vitro, partially attributable to modulation of Akt phosphorylation.
Collapse
Affiliation(s)
- Guang Hu
- Renal Division, Center for Study of the Tumor Microenvironment and Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The purpose of this review is not to provide an extensive overview of well-established mechanisms of angiogenesis and lymphangiogenesis but rather to highlight several recent key studies that constituted a significant conceptual or medical advancement to the field during the past year or so. The authors apologize for their inability, because of space restrictions, to reference all other relevant work of the past or previous years. RECENT FINDINGS In 1993, fewer than 400 studies on angiogenesis were published. During the past year alone, more than 4000 angiogenesis studies were reported, making angiogenesis one of the most rapidly growing fields. Moreover, the first studies on lymphangiogenesis were published only a couple of years ago. A milestone in the field in the past year has been the first successful report that the angiogenesis inhibitor bevacizumab (Avastin), an antibody against vascular endothelial growth factor, prolonged the survival of colorectal and renal cancer patients in phase 3 clinical trials. This remarkable achievement provides great promise and hope for the future development of therapeutic strategies to inhibit or stimulate angiogenesis. SUMMARY The intensive search for antiangiogenic and proangiogenic mechanisms during the past decade is starting to translate into clinical promise. Further discovery of novel pathways and concepts in angiogenesis may lead to the optimization and refinement of current strategies to improve the clinical benefit and therapeutic safety for a vast number of patients with angiogenesis-related disease.
Collapse
Affiliation(s)
- Aernout Luttun
- Center for Transgene Technology and Gene Therapy, Flanders Interuniversity Institute for Biotechnology, Leuven, Belgium
| | | |
Collapse
|