1
|
Upreti A, Padula SL, Weaver JM, Wagner BD, Kneller AM, Petulla AL, Lachke SA, Robinson ML. A Transcriptomics Analysis of the Regulation of Lens Fiber Cell Differentiation in the Absence of FGFRs and PTEN. Cells 2024; 13:1222. [PMID: 39056803 PMCID: PMC11274593 DOI: 10.3390/cells13141222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/28/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Adding 50% vitreous humor to the media surrounding lens explants induces fiber cell differentiation and a significant immune/inflammatory response. While Fgfr loss blocks differentiation in lens epithelial explants, this blockage is partially reversed by deleting Pten. To investigate the functions of the Fgfrs and Pten during lens fiber cell differentiation, we utilized a lens epithelial explant system and conducted RNA sequencing on vitreous humor-exposed explants lacking Fgfrs, or Pten or both Fgfrs and Pten. We found that Fgfr loss impairs both vitreous-induced differentiation and inflammation while the additional loss of Pten restores these responses. Furthermore, transcriptomic analysis suggested that PDGFR-signaling in FGFR-deficient explants is required to mediate the rescue of vitreous-induced fiber differentiation in explants lacking both Fgfrs and Pten. The blockage of β-crystallin induction in explants lacking both Fgfrs and Pten in the presence of a PDGFR inhibitor supports this hypothesis. Our findings demonstrate that a wide array of genes associated with fiber cell differentiation are downstream of FGFR-signaling and that the vitreous-induced immune responses also depend on FGFR-signaling. Our data also demonstrate that many of the vitreous-induced gene-expression changes in Fgfr-deficient explants are rescued in explants lacking both Fgfrs and Pten.
Collapse
Affiliation(s)
- Anil Upreti
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Stephanie L. Padula
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Jacob M. Weaver
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Brad D. Wagner
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Allison M. Kneller
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Anthony L. Petulla
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| | - Salil A. Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19716, USA
| | - Michael L. Robinson
- Cell, Molecular and Structural Biology Program, Miami University, Oxford, OH 45056, USA; (A.U.); (S.L.P.); (J.M.W.)
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA; (B.D.W.); (A.M.K.); (A.L.P.)
| |
Collapse
|
2
|
Zhang L, Liu X, Li W, Liu K, Zhang J, Liu X, Wang J. Integrative transcriptomic profiling of ncRNAs and mRNAs in developing mouse lens. Front Genet 2024; 15:1405715. [PMID: 38933921 PMCID: PMC11199715 DOI: 10.3389/fgene.2024.1405715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
In recent years, burgeoning research has underscored the pivotal role of non-coding RNA in orchestrating the growth, development, and pathogenesis of various diseases across organisms. However, despite these advances, our understanding of the specific contributions of long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) to lens development remains notably limited. Clarifying the intricate gene regulatory networks is imperative for unraveling the molecular underpinnings of lens-related disorders. In this study, we aimed to address this gap by conducting a comprehensive analysis of the expression profiles of messenger RNAs (mRNAs), lncRNAs, and circRNAs at critical developmental time points of the mouse lens, encompassing both embryonic (E10.5, E12.5, and E16.5) and postnatal stages (P0.5, P10.5, and P60). Leveraging RNA-sequencing technology, we identified key transcripts pivotal to lens development. Our analysis revealed differentially expressed (DE) mRNAs, lncRNAs, and circRNAs across various developmental stages. Particularly noteworthy, there were 1831 co-differentially expressed (CO-DE) mRNAs, 150 CO-DE lncRNAs, and 13 CO-DE circRNAs identified during embryonic stages. Gene Ontology (GO) enrichment analysis unveiled associations primarily related to lens development, DNA conformational changes, and angiogenesis among DE mRNAs and lncRNAs. Furthermore, employing protein-protein interaction networks, mRNA-lncRNA co-expression networks, and circRNA-microRNA-mRNA networks, we predicted candidate key molecules implicated in lens development. Our findings underscore the pivotal roles of lncRNAs and circRNAs in this process, offering fresh insights into the pathogenesis of lens-related disorders and paving the way for future exploration in this field.
Collapse
Affiliation(s)
- Liyun Zhang
- Department of Ophthalmology, General Hospital of Central Theater Command, Wuhan, China
| | - Xin Liu
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Wei Li
- Department of Pediatric Respiratory Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaiqing Liu
- The Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Jing Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Xinhua Liu
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Jiantao Wang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Serpelloni M, Arricca M, Ravelli C, Grillo E, Mitola S, Salvadori A. Mechanobiology of the relocation of proteins in advecting cells: in vitro experiments, multi-physics modeling, and simulations. Biomech Model Mechanobiol 2023:10.1007/s10237-023-01717-2. [PMID: 37067608 PMCID: PMC10366044 DOI: 10.1007/s10237-023-01717-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/29/2023] [Indexed: 04/18/2023]
Abstract
Cell motility-a cellular behavior of paramount relevance in embryonic development, immunological response, metastasis, or angiogenesis-demands a mechanical deformation of the cell membrane and influences the surface motion of molecules and their biochemical interactions. In this work, we develop a fully coupled multi-physics model able to capture and predict the protein flow on endothelial advecting plasma membranes. The model has been validated against co-designed in vitro experiments. The complete picture of the receptor dynamics has been understood, and limiting factors have been identified together with the laws that regulate receptor polarization. This computational approach might be insightful in the prediction of endothelial cell behavior in different tumoral environments, circumventing the time-consuming and expensive empirical characterization of each tumor.
Collapse
Affiliation(s)
- M Serpelloni
- The Mechanobiology research center, UNIBS, 25123, Brescia, Italy
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, 25123, Brescia, Italy
| | - M Arricca
- The Mechanobiology research center, UNIBS, 25123, Brescia, Italy
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, 25123, Brescia, Italy
| | - C Ravelli
- The Mechanobiology research center, UNIBS, 25123, Brescia, Italy
- Department of Molecular and Translational Medicine, Università degli Studi di Brescia, 25123, Brescia, Italy
| | - E Grillo
- The Mechanobiology research center, UNIBS, 25123, Brescia, Italy
- Department of Molecular and Translational Medicine, Università degli Studi di Brescia, 25123, Brescia, Italy
| | - S Mitola
- The Mechanobiology research center, UNIBS, 25123, Brescia, Italy
- Department of Molecular and Translational Medicine, Università degli Studi di Brescia, 25123, Brescia, Italy
| | - A Salvadori
- The Mechanobiology research center, UNIBS, 25123, Brescia, Italy.
- Department of Mechanical and Industrial Engineering, Università degli Studi di Brescia, 25123, Brescia, Italy.
| |
Collapse
|
4
|
Khokhar S, Surve A, Verma S, Azad S, Chandra P, Dhull C, Vohra R. Cataract in retinopathy of prematurity - A review. Indian J Ophthalmol 2022; 70:369-377. [PMID: 35086199 PMCID: PMC9023920 DOI: 10.4103/ijo.ijo_125_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Preterm babies with retinopathy of prematurity (ROP) can become blind if they do not receive appropriate timely intervention. The presence of cataract in these individuals in addition to visual deprivation amblyopia, also delays proper screening, adequate treatment, and makes follow-up assessment difficult. Anatomical differences in these infants and amblyopia management, especially in unilateral cataract, are other important concerns, and hence, management of these cases with cataract and ROP is challenging. In this review, studies where ROP cases were associated with cataract, were evaluated with a focus on preterm individuals less than 6 months age. Preterm babies are at increased risk of developing cataract because of systemic factors. In addition, those with ROP may have cataract associated with retinal detachment or treatment received. The type of cataract, risk factors, and pathophysiology associated with each cause varies. This review highlights these different aspects of cataract in ROP including causes, pathophysiology, types of cataracts, and management. The management of these cases is critical in terms of the timing of cataract surgery and the challenges associated with surgery and posterior segment management for ROP. Anatomical differences, preoperative retina status, pupillary dilatation, neovascularization of iris in aggressive posterior ROP, fundus examination, amblyopia, and follow-up are various important aspects in the management of the same. The preoperative workup, intraoperative challenges, postoperative care, and rehabilitation in these individuals are discussed.
Collapse
Affiliation(s)
- Sudarshan Khokhar
- Cataract and Refractive Service; Dr. Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Abhidnya Surve
- Cataract and Refractive Service; Vitreoretinal, Uvea and ROP Service; Dr. Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Saurabh Verma
- Cataract and Refractive Service; Vitreoretinal, Uvea and ROP Service; Dr. Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Shorya Azad
- Vitreoretinal, Uvea and ROP Service; Dr. Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Parijat Chandra
- Vitreoretinal, Uvea and ROP Service; Dr. Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Chirakshi Dhull
- Cataract and Refractive Service; Vitreoretinal, Uvea and ROP Service; Dr. Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Rajpal Vohra
- Vitreoretinal, Uvea and ROP Service; Dr. Rajendra Prasad Center for Ophthalmic Sciences, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| |
Collapse
|
5
|
McGraw MD, Kim SY, White CW, Veress LA. Acute cytotoxicity and increased vascular endothelial growth factor after in vitro nitrogen mustard vapor exposure. Ann N Y Acad Sci 2020; 1479:223-233. [PMID: 32408394 DOI: 10.1111/nyas.14367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022]
Abstract
Nitrogen mustard (NM) is a highly toxic alkylating agent. Inhalation exposure can cause acute and chronic lung injury. This study's aims were to develop an in vitro coculture model of mustard-induced airway injury and to identify growth factors contributing to airway pathology. Primary human bronchial epithelial cells cultured with pulmonary endothelial cells were exposed to NM (25, 50, 100, 250, or 500 μM) or PBS (control) for 1 hour. Lactate dehydrogenase (LDH) and transepithelial electrical resistance (TEER) were measured before and 24 h after NM exposure. Fixed cultures were stained for hematoxylin and eosin or live/dead staining. Culture media were analyzed for 11 growth factors. A 1-h vapor exposure to greater than or equal to 50 μM NM increased supernatant LDH, decreased TEER, and caused airway epithelial cell detachment. Endothelial cell death occurred at 500 μM NM. Vascular endothelial growth factor A (VEGF-A) and placental growth factor (PlGF) expression increased in 500 μM NM-exposed cultures compared with PBS-exposed control cultures. NM vapor exposure causes differential cytotoxicity to airway epithelial and endothelial injury in culture. Increased VEGF-A and PlGF expression occurred acutely in airway cocultures. Future studies are required to validate the role of VEGF signaling in mustard-induced airway pathology.
Collapse
Affiliation(s)
- Matthew D McGraw
- Department of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - So-Young Kim
- Department of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York
| | - Carl W White
- Department of Pediatrics, Pulmonology Section, Pediatric Airway Research Center, University of Colorado Denver, Aurora, Colorado
| | - Livia A Veress
- Department of Pediatrics, Pulmonology Section, Pediatric Airway Research Center, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
6
|
Gaspar D, Peixoto R, De Pieri A, Striegl B, Zeugolis DI, Raghunath M. Local pharmacological induction of angiogenesis: Drugs for cells and cells as drugs. Adv Drug Deliv Rev 2019; 146:126-154. [PMID: 31226398 DOI: 10.1016/j.addr.2019.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/12/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
The past decades have seen significant advances in pro-angiogenic strategies based on delivery of molecules and cells for conditions such as coronary artery disease, critical limb ischemia and stroke. Currently, three major strategies are evolving. Firstly, various pharmacological agents (growth factors, interleukins, small molecules, DNA/RNA) are locally applied at the ischemic region. Secondly, preparations of living cells with considerable bandwidth of tissue origin, differentiation state and preconditioning are delivered locally, rarely systemically. Thirdly, based on the notion, that cellular effects can be attributed mostly to factors secreted in situ, the cellular secretome (conditioned media, exosomes) has come into the spotlight. We review these three strategies to achieve (neo)angiogenesis in ischemic tissue with focus on the angiogenic mechanisms they tackle, such as transcription cascades, specific signalling steps and cellular gases. We also include cancer-therapy relevant lymphangiogenesis, and shall seek to explain why there are often conflicting data between in vitro and in vivo. The lion's share of data encompassing all three approaches comes from experimental animal work and we shall highlight common technical obstacles in the delivery of therapeutic molecules, cells, and secretome. This plethora of preclinical data contrasts with a dearth of clinical studies. A lack of adequate delivery vehicles and standardised assessment of clinical outcomes might play a role here, as well as regulatory, IP, and manufacturing constraints of candidate compounds; in addition, completed clinical trials have yet to reveal a successful and efficacious strategy. As the biology of angiogenesis is understood well enough for clinical purposes, it will be a matter of time to achieve success for well-stratified patients, and most probably with a combination of compounds.
Collapse
Affiliation(s)
- Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Rita Peixoto
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Britta Striegl
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Michael Raghunath
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland.
| |
Collapse
|
7
|
Tam OH, Pennisi D, Wilkinson L, Little MH, Wazin F, Wan VL, Lovicu FJ. Crim1 is required for maintenance of the ocular lens epithelium. Exp Eye Res 2018; 170:58-66. [PMID: 29458060 DOI: 10.1016/j.exer.2018.02.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 12/28/2022]
Abstract
The development and growth of the vertebrate ocular lens is dependent on the regulated proliferation of an anterior monolayer of epithelial cells, and their subsequent differentiation into elongate fiber cells. The growth factor rich ocular media that bathes the lens mediates these cellular processes, and their respective intracellular signaling pathways are in turn regulated to ensure that the proper lens architecture is maintained. Recent studies have proposed that Cysteine Rich Motor Neuron 1 (Crim1), a transmembrane protein involved in organogenesis of many tissues, might influence cell adhesion, polarity and proliferation in the lens by regulating integrin-signaling. Here, we characterise the lens and eyes of the Crim1KST264 mutant mice, and show that the loss of Crim1 function in the ocular tissues results in inappropriate differentiation of the lens epithelium into fiber cells. Furthermore, restoration of Crim1 levels in just the lens tissue of Crim1KST264 mice is sufficient to ameliorate most of the dysgenesis observed in the mutant animals. Based on our findings, we propose that tight regulation of Crim1 activity is required for maintenance of the lens epithelium, and its depletion leads to ectopic differentiation into fiber cells, dramatically altering lens structure and ultimately leading to microphthalmia and aphakia.
Collapse
Affiliation(s)
- Oliver H Tam
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia
| | - David Pennisi
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | - Lorine Wilkinson
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | - Melissa H Little
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | - Fatima Wazin
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia
| | - Victor L Wan
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia
| | - Frank J Lovicu
- Save Sight Institute and Anatomy & Histology, Bosch Institute, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
8
|
Joyal JS, Gantner ML, Smith LEH. Retinal energy demands control vascular supply of the retina in development and disease: The role of neuronal lipid and glucose metabolism. Prog Retin Eye Res 2017; 64:131-156. [PMID: 29175509 DOI: 10.1016/j.preteyeres.2017.11.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/11/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Jean-Sébastien Joyal
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Canada.
| | - Marin L Gantner
- The Lowy Medical Research Institute, La Jolla, United States
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston MA 02115, United States.
| |
Collapse
|
9
|
Gupta S, Gogia V, T R, Sen S, Venkatesh P. Posterior lens capsular neovascularization of young: management using endodiathermy assisted biopsy. Can J Ophthalmol 2015; 50:e4-7. [PMID: 25677299 DOI: 10.1016/j.jcjo.2014.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 09/23/2014] [Accepted: 10/01/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Shikha Gupta
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India.
| | - Varun Gogia
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Roshan T
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Seema Sen
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Pradeep Venkatesh
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
10
|
Hartsock A, Lee C, Arnold V, Gross JM. In vivo analysis of hyaloid vasculature morphogenesis in zebrafish: A role for the lens in maturation and maintenance of the hyaloid. Dev Biol 2014; 394:327-39. [PMID: 25127995 DOI: 10.1016/j.ydbio.2014.07.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 01/11/2023]
Abstract
Two vascular networks nourish the embryonic eye as it develops - the hyaloid vasculature, located at the anterior of the eye between the retina and lens, and the choroidal vasculature, located at the posterior of the eye, surrounding the optic cup. Little is known about hyaloid development and morphogenesis, however. To begin to identify the morphogenetic underpinnings of hyaloid formation, we utilized in vivo time-lapse confocal imaging to characterize morphogenesis of the zebrafish hyaloid through 5 days post fertilization (dpf). Our data segregate hyaloid formation into three distinct morphogenetic stages: Stage I: arrival of hyaloid cells at the lens and formation of the hyaloid loop; Stage II: formation of a branched hyaloid network; Stage III: refinement of the hyaloid network. Utilizing fixed and dissected tissues, distinct Stage II and Stage III aspects of hyaloid formation were quantified over time. Combining in vivo imaging with microangiography, we demonstrate that the hyaloid system becomes fully enclosed by 5dpf. To begin to identify the molecular and cellular mechanisms underlying hyaloid morphogenesis, we identified a recessive mutation in the mab21l2 gene, and in a subset of mab21l2 mutants the lens does not form. Utilizing these "lens-less" mutants, we determined whether the lens was required for hyaloid morphogenesis. Our data demonstrate that the lens is not required for Stage I of hyaloid formation; however, Stages II and III of hyaloid formation are disrupted in the absence of a lens, supporting a role for the lens in hyaloid maturation and maintenance. Taken together, this study provides a foundation on which the cellular, molecular and embryologic mechanisms underlying hyaloid morphogenesis can be elucidated.
Collapse
Affiliation(s)
- Andrea Hartsock
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Chanjae Lee
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Victoria Arnold
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States
| | - Jeffrey M Gross
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
11
|
Sema3A maintains corneal avascularity during development by inhibiting Vegf induced angioblast migration. Dev Biol 2014; 391:241-50. [PMID: 24809797 DOI: 10.1016/j.ydbio.2014.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/14/2014] [Accepted: 04/23/2014] [Indexed: 12/25/2022]
Abstract
Corneal avascularity is important for optical clarity and normal vision. However, the molecular mechanisms that prevent angioblast migration and vascularization of the developing cornea are not clear. Previously we showed that periocular angioblasts and forming ocular blood vessels avoid the presumptive cornea despite dynamic ingression of neural crest cells. In the current study, we investigate the role of Semaphorin3A (Sema3A), a cell guidance chemorepellent, on angioblast migration and corneal avascularity during development. We show that Sema3A, Vegf, and Nrp1 are expressed in the anterior eye during cornea development. Sema3A mRNA transcripts are expressed at significantly higher levels than Vegf in the lens that is positioned adjacent to the presumptive cornea. Blockade of Sema3A signaling via lens removal or injection of a synthetic Sema3A inhibitor causes ectopic migration of angioblasts into the cornea and results in its subsequent vascularization. In addition, using bead implantation, we demonstrate that exogenous Sema3A protein inhibits Vegf-induced vascularization of the cornea. In agreement with these findings, loss of Sema/Nrp1 signaling in Nrp1(Sema-) mutant mice results in ectopic angioblasts and vascularization of the embryonic mouse corneas. Altogether, our results reveal Sema3A signaling as an important cue during the establishment of corneal avascularity in both chick and mouse embryos. Our study introduces cornea development as a new model for studying the mechanisms involved in vascular patterning during embryogenesis and it also provides new insights into therapeutic potential for Sema3A in neovascular diseases.
Collapse
|
12
|
Al-Shabrawey M, Elsherbiny M, Nussbaum J, Othman A, Megyerdi S, Tawfik A. Targeting Neovascularization in Ischemic Retinopathy: Recent Advances. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 8:267-286. [PMID: 25598837 DOI: 10.1586/eop.13.17] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pathological retinal neovascularization (RNV) is a common micro-vascular complication in several retinal diseases including retinopathy of prematurity, diabetic retinopathy, age-related macular degeneration and central vein occlusion. The current therapeutic modalities of RNV are invasive and although they may slow or halt the progression of the disease they are unlikely to restore normal acuity. Therefore, there is an urgent need to develop treatment modalities, which are less invasive and therefore associated with fewer procedural complications and systemic side effects. This review article summarizes our understanding of the pathophysiology and current treatment of RNV in ischemic retinopathies; lists potential therapeutic targets; and provides a framework for the development of future treatment modalities.
Collapse
Affiliation(s)
- Mohamed Al-Shabrawey
- Oral Biology/Anatomy, College of Dental Medicine, GeorgiaRegentsUniversity (GRU), Augusta GA, USA ; Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU ; Anatomy, Mansoura Faculty of Medicine, Mansoura University-Egypt ; Vascular Biology Center, Medical College of Georgia, GRU
| | - Mohamed Elsherbiny
- Oral Biology/Anatomy, College of Dental Medicine, GeorgiaRegentsUniversity (GRU), Augusta GA, USA ; Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU ; Anatomy, Mansoura Faculty of Medicine, Mansoura University-Egypt
| | - Julian Nussbaum
- Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU
| | - Amira Othman
- Anatomy, Mansoura Faculty of Medicine, Mansoura University-Egypt
| | - Sylvia Megyerdi
- Oral Biology/Anatomy, College of Dental Medicine, GeorgiaRegentsUniversity (GRU), Augusta GA, USA
| | - Amany Tawfik
- Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU ; Cellular Biology and Anatomy, Medical College of Georgia, GRU
| |
Collapse
|
13
|
Ribatti D, Crivellato E. “Sprouting angiogenesis”, a reappraisal. Dev Biol 2012; 372:157-65. [DOI: 10.1016/j.ydbio.2012.09.018] [Citation(s) in RCA: 209] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/22/2012] [Accepted: 09/24/2012] [Indexed: 01/15/2023]
|
14
|
Watson MG, McDougall SR, Chaplain MAJ, Devlin AH, Mitchell CA. Dynamics of angiogenesis during murine retinal development: a coupled in vivo and in silico study. J R Soc Interface 2012; 9:2351-64. [PMID: 22438490 DOI: 10.1098/rsif.2012.0067] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The manner in which the superficial retinal vascular plexus (RVP) develops in neonatal wild-type mice is relatively well documented and poses an interesting challenge to the mathematical modelling community. Prior to birth, astrocyte sprouting and proliferation begin around the edge of the optic nerve head, and subsequent astrocyte migration in response to a chemotactic gradient of platelet-derived growth factor (PDGF)-A results in the formation of a dense scaffold on the surface of the inner retina. Astrocytes express a variety of chemotactic and haptotactic proteins that subsequently induce endothelial cell sprouting and modulate growth of the RVP. An experimentally informed, two-dimensional hybrid partial differential equation-discrete model is derived to track the outward migration of individual astrocyte and endothelial tip cells in response to the appropriate biochemical cues. Blood perfusion is included throughout the development of the plexus, and the evolving retinal trees are allowed to adapt and remodel by means of several biological stimuli. The resulting wild-type in silico RVP structures are compared with corresponding experimental whole mounts taken at various stages of development, and agreement between the respective vascular morphologies is found to be excellent. Subsequent numerical predictions help elucidate some of the key biological processes underlying retinal development and demonstrate the potential of the virtual retina for the investigation of various vascular-related diseases of the eye.
Collapse
Affiliation(s)
- M G Watson
- Institute of Petroleum Engineering, Heriot-Watt University, Edinburgh, UK.
| | | | | | | | | |
Collapse
|
15
|
Atwood RC, Lee PD, Konerding MA, Rockett P, Mitchell CA. Quantitation of microcomputed tomography-imaged ocular microvasculature. Microcirculation 2010; 17:59-68. [PMID: 20141601 DOI: 10.1111/j.1549-8719.2009.00009.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE To quantitatively assess microvascular dimensions in the eyes of neonatal wild-type and VEGF(120)-tg mice, using a novel combination of techniques which permit three-dimensional (3D) image reconstruction. METHODS A novel combination of techniques was developed for the accurate 3D imaging of the microvasculature and demonstrated on the hyaloid vasculature of the neonatal mouse eye. Vascular corrosion casting is used to create a stable replica of the vascular network and X-ray microcomputed tomography (muCT) to obtain the 3D images. In-house computer-aided image analysis techniques were then used to perform a quantitative morphological analysis of the images. RESULTS With the use of these methods, differences in the numbers of vessel segments, their diameter, and volume of vessels in the vitreous compartment were quantitated in wild-type neonatal mice or littermates over-expressing a labile (nonheparin binding) isoform of vascular endothelial growth factor (VEGF(120)) from the developing lens. This methodology was instructive in demonstrating that hyaloid vascular networks in VEGFA(120) over-expressing mice have a 10-fold increase in blind-ended, a six-fold increase in connected vessel segments, in addition to a sixfold increase (0.0314 versus 0.0051 mm(3)) in total vitreous vessel volume compared with wild type. These parameters are not readily quantified via histological, ultrastructural, or stereological analysis. CONCLUSION The combination of techniques described here provides the first 3D quantitative characterization of vasculature in an organ system; i.e., the neonatal murine intra-ocular vasculature in both wild-type mice and a transgenic model of lens-specific over-expression of VEGF.
Collapse
Affiliation(s)
- Robert C Atwood
- Department of Materials, Imperial College London, London, UK
| | | | | | | | | |
Collapse
|
16
|
Safvati A, Cole N, Hume E, Willcox M. Mediators of neovascularization and the hypoxic cornea. Curr Eye Res 2009; 34:501-14. [PMID: 19899985 DOI: 10.1080/02713680902919557] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The maintenance of corneal avascularity is essential to vision. The mechanisms by which the cornea becomes vascularized in response to inflammation or hypoxic stress are beginning to be elucidated. A detailed understanding of the molecular responses of the cornea to hypoxia is critical for prevention and development of novel treatments for neovascularization in a range of disease states. Here, we have examined the current literature on the major mediators of angiogenesis, which have previously been reported during hypoxia in the cornea in order to better understand the mechanisms by which corneal angiogenesis occurs in circumstances where the available oxygen is reduced. The normal cornea produces angiogenic factors that are regulated by the production of anti-angiogenic molecules. The various cell types of the cornea respond differentially to inflammatory and hypoxic stimuli. An understanding of the factors that may predispose patients to development of corneal blood vessels may provide an opportunity to develop novel prophylactic strategies. The difficulties with extrapolating data from other cell types and animal models to the cornea are also examined.
Collapse
Affiliation(s)
- Aidin Safvati
- Vision Cooperative Research Centre and School of Optometry and Vision Science, The University of New South Wales, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
17
|
Potier E, Ferreira E, Dennler S, Mauviel A, Oudina K, Logeart-Avramoglou D, Petite H. Desferrioxamine-driven upregulation of angiogenic factor expression by human bone marrow stromal cells. J Tissue Eng Regen Med 2008; 2:272-8. [PMID: 18512268 DOI: 10.1002/term.92] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone marrow stromal cells (BMSCs) are the subject of intense research because of their biological properties and potential use for the repair of damaged tissues. Success of BMSC-based therapies, however, relies on a number of methodological improvements, including the establishment of a vascular network providing nutrients and oxygen to the transplanted cells and ensuring their immediate survival and long-term functionality. We described a method to enhance the autocrine expression of angiogenic factors by BMSCs. For this purpose, human BMSCs were treated with desferrioxamine (DFX). No PDGF-BB, VEGF-R1 or -R2 mRNA expression was detected under any of the conditions tested. mRNA and protein expression levels of TGFbeta1 were similar in BMSCs, whether they were exposed to DFX (50 microM) or to control conditions under normoxia for 48 h. In comparison with the results obtained with control conditions under normoxia, exposure of BMSCs to DFX for 48 h resulted in upregulation of bFGF at the protein (26-fold) but not at the mRNA levels and VEGF at both the mRNA (1.5-fold) and protein levels (4.5-fold). In comparison with the results obtained with control conditions under hypoxia, DFX induced a 50% increase in VEGF secretion but led to the same level of hypoxia inducible factor-1alpha protein expression (a transduction factor involved in angiogenic factor expression and known to be activated by DFX). Exposure of BMSCs to DFX resulted in oversecretion of angiogenic factors, suggesting that DFX-treated BMSCs could be used to supply angiogenic factors.
Collapse
Affiliation(s)
- Esther Potier
- Université Denis Diderot Paris VII, Laboratoire de Recherches Orthopédiques (B2OA), UMR CNRS 7052, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
18
|
Saint-Geniez M, Kurihara T, D'Amore PA. Role of cell and matrix-bound VEGF isoforms in lens development. Invest Ophthalmol Vis Sci 2008; 50:311-21. [PMID: 18757513 DOI: 10.1167/iovs.08-2461] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To determine the role of vascular endothelial growth factor (VEGF) in embryonic eye development and lens differentiation. METHODS Expression of components of the VEGF signaling pathway during lens development and in adults was characterized by beta-galactosidase staining of VEGF-LacZ mice, immunohistochemistry, and real-time (q) PCR. Embryonic eyes from wild-type mice and VEGF120/120 mice were analyzed by light microscopy and immunohistochemistry. VEGF function during lens development was analyzed using eye explants treated with VEGF-neutralizing antibody. Direct function of VEGF was demonstrated on the human lens epithelial cell line, HLE-B3. RESULTS Embryonic lens epithelium and posterior lens fibers expressed VEGF and VEGFR2. qPCR revealed VEGF164 as the major isoform in embryonic lens. Transgenic mice expressing only VEGF120 (VEGF120/120 mice) showed major defects in eye development, including microphthalmia, failed lens differentiation, and hyperplastic hyaloid vessels. The lens displayed abnormal cell patterning and differentiation associated with altered c-Maf, Prox1, and p57 expression pattern in the anterior epithelium. The number of proliferating epithelial cells was drastically reduced in VEGF120/120 lenses. Altered MIP26 cellular localization and reduced E-cadherin expression in the lens epithelium were observed. VEGF-neutralization led to reduced fiber elongation of eye explants. Exogenous VEGF increased survival and proliferation of HLE-B3 cell in a dose-dependent manner. CONCLUSIONS Abnormalities in ocular development in VEGF120/120 mice suggest a role for VEGF not only in the formation of ocular vascular beds but also in the differentiation of the lens itself.
Collapse
|
19
|
Garcia CM, Shui YB, Kamath M, DeVillar J, Johnson RS, Gerber HP, Ferrara N, Robinson ML, Beebe DC. The function of VEGF-A in lens development: formation of the hyaloid capillary network and protection against transient nuclear cataracts. Exp Eye Res 2008; 88:270-6. [PMID: 18782574 DOI: 10.1016/j.exer.2008.07.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Revised: 06/23/2008] [Accepted: 07/28/2008] [Indexed: 10/21/2022]
Abstract
A network of capillaries branches from the hyaloid vascular system and surrounds the mammalian lens throughout much of its embryonic development. These vessels are presumed to be important for the growth and maturation of the lens, although the lenses of non-mammalian vertebrates have no comparable vessels. Over expression of VEGF-A in the lens increases the extent of these capillaries, but it is not known whether VEGF-A from the lens is necessary for their formation or survival. To address this question, we deleted Vegfa in the lens. This prevented the formation of the capillary networks adjacent to the lens capsule, but did not alter nearby hyaloid vessels at the surface of the retina. Postnatal lenses lacking Vegfa were smaller than wild type and, by 1 month of age, many had mild nuclear opacities. These opacities regressed with age. The lens is hypoxic throughout most of life and VEGF-A expression is often regulated by the transcription factor, hypoxia inducible factor-1. Lenses lacking Hif1a were of apparently normal size, had markedly reduced levels of mRNA for VEGF-A and glyceraldehyde-3-phosphate dehydrogenase, but had normal-appearing capillaries covering their surface. We conclude that VEGF-A from the lens is necessary for the formation of the normal hyaloid vascular system and that lack of these capillaries was the most likely cause of growth retardation during fetal and early postnatal lens development. In the absence of HIF-1 function, sufficient VEGF-A is produced by the lens to promote capillary formation. Further study is needed to explain the formation of the mild opacities seen in some lenses lacking Vegfa and their regression later in life.
Collapse
Affiliation(s)
- Claudia M Garcia
- Department of Ophthalmology and Visual Science, Washington University, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Chen Y, Doughman YQ, Gu S, Jarrell A, Aota SI, Cvekl A, Watanabe M, Dunwoodie SL, Johnson RS, van Heyningen V, Kleinjan DA, Beebe DC, Yang YC. Cited2 is required for the proper formation of the hyaloid vasculature and for lens morphogenesis. Development 2008; 135:2939-48. [PMID: 18653562 DOI: 10.1242/dev.021097] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cited2 is a transcriptional modulator with pivotal roles in different biological processes. Cited2-deficient mouse embryos manifested two major defects in the developing eye. An abnormal corneal-lenticular stalk was characteristic of Cited2(-/-) developing eyes, a feature reminiscent of Peters' anomaly, which can be rescued by increased Pax6 gene dosage in Cited2(-/-) embryonic eyes. In addition, the hyaloid vascular system showed hyaloid hypercellularity consisting of aberrant vasculature, which might be correlated with increased VEGF expression in the lens. Deletion of Hif1a (which encodes HIF-1alpha) in Cited2(-/-) lens specifically eliminated the excessive accumulation of cellular mass and aberrant vasculature in the developing vitreous without affecting the corneal-lenticular stalk phenotype. These in vivo data demonstrate for the first time dual functions for Cited2: one upstream of, or together with, Pax6 in lens morphogenesis; and another in the normal formation of the hyaloid vasculature through its negative modulation of HIF-1 signaling. Taken together, our study provides novel mechanistic revelation for lens morphogenesis and hyaloid vasculature formation and hence might offer new insights into the etiology of Peters' anomaly and ocular hypervascularity.
Collapse
Affiliation(s)
- Yu Chen
- Department of Biochemistry and Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Beebe DC. Maintaining transparency: a review of the developmental physiology and pathophysiology of two avascular tissues. Semin Cell Dev Biol 2007; 19:125-33. [PMID: 17920963 DOI: 10.1016/j.semcdb.2007.08.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Revised: 08/22/2007] [Accepted: 08/24/2007] [Indexed: 12/13/2022]
Abstract
The lens and cornea are transparent and usually avascular. Controlling nutrient supply while maintaining transparency is a physiological challenge for both tissues. During sleep and with contact lens wear the endothelial layer of the cornea may become hypoxic, compromising its ability to maintain corneal transparency. The mechanism responsible for establishing the avascular nature of the corneal stroma is unknown. In several pathological conditions, the stroma can be invaded by abnormal, leaky vessels, leading to opacification. Several molecules that are likely to help maintain the avascular nature of the corneal stroma have been identified, although their relative contributions remain to be demonstrated. The mammalian lens is surrounded by capillaries early in life. After the fetal vasculature regresses, the lens resides in a hypoxic environment. Hypoxia is likely to be required to maintain lens transparency. The vitreous body may help to maintain the low oxygen level around the lens. The hypothesis is presented that many aspects of the aging of the lens, including increased hardening, loss of accommodation (presbyopia), and opacification of the lens nucleus, are caused by exposure to oxygen. Testing this hypothesis may lead to prevention for nuclear cataract and insight into the mechanisms of lens aging. Although they are both transparent, corneal pathology is associated with an insufficient supply of oxygen, while lens pathology may involve excessive exposure to oxygen.
Collapse
Affiliation(s)
- David C Beebe
- Department of Ophthalmology and Visual Sciences, Washington University, St Louis, MO 63110, USA.
| |
Collapse
|
22
|
Abstract
Migration of endothelial precursor cells (so-called "angioblasts" in embryos and "endothelial progenitor cells" in adults) during vasculogenesis is a requirement for the formation of a primary vascular plexus. The migration is initiated by the change of endothelial precursors to their migratory phenotype. The endothelial precursor cells are then guided to the position where the primary vascular plexus is formed. Migration is stopped by the reversion of the cells to their nonmigratory phenotype. A combination of regulatory mechanisms and factors controls this process. These include gradients of soluble factors, extracellular matrix-cell interaction and cell-cell interaction. In this review, we give an overview of the regulation of angioblast migration during embryonic vasculogenesis and its relationship to the migration of endothelial progenitors during postnatal vascular development.
Collapse
Affiliation(s)
- Annette Schmidt
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| | | | | |
Collapse
|
23
|
Potier E, Ferreira E, Andriamanalijaona R, Pujol JP, Oudina K, Logeart-Avramoglou D, Petite H. Hypoxia affects mesenchymal stromal cell osteogenic differentiation and angiogenic factor expression. Bone 2007; 40:1078-87. [PMID: 17276151 DOI: 10.1016/j.bone.2006.11.024] [Citation(s) in RCA: 227] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 11/17/2006] [Accepted: 11/22/2006] [Indexed: 01/02/2023]
Abstract
Mesenchymal stromal cells (MSCs) seeded onto biocompatible scaffolds have been proposed for repairing bone defects. When transplanted in vivo, MSCs (expanded in vitro in 21% O(2)) undergo temporary oxygen deprivation due to the lack of pre-existing blood vessels within these scaffolds. In the present study, the effects of temporary (48 h) exposure to hypoxia (<or=1% O(2)) on primary human MSC survival and osteogenic potential were investigated. Temporary exposure of MSCs to hypoxia had no effect on MSC survival, but resulted in (i) persistent (up to 14 days post exposure) down-regulation of cbfa-1/Runx2, osteocalcin and type I collagen and (ii) permanent (up to 28 days post exposure) up-regulation of osteopontin mRNA expressions. Since angiogenesis is known to contribute crucially to alleviating hypoxia, the effects of temporary hypoxia on angiogenic factor expression by MSCs were also assessed. Temporary hypoxia led to a 2-fold increase in VEGF expression at both the mRNA and protein levels. Other growth factors and cytokines secreted by MSCs under control conditions (namely bFGF, TGFbeta1 and IL-8) were not affected by temporary exposure to hypoxia. All in all, these results indicate that temporary exposure of MSCs to hypoxia leads to limited stimulation of angiogenic factor secretion but to persistent down-regulation of several osteoblastic markers, which suggests that exposure of MSCs transplanted in vivo to hypoxia may affect their bone forming potential. These findings prompt for the development of appropriate cell culture or in vivo transplantation conditions preserving the full osteogenic potential of MSCs.
Collapse
Affiliation(s)
- Esther Potier
- Laboratoire de Recherches Orthopédiques (B2OA), UMR CNRS 7052, Faculté de Médecine Lariboisière-Saint-Louis, 10 Avenue de Verdun, 75010 Paris, France
| | | | | | | | | | | | | |
Collapse
|
24
|
Zhang SX, Ma JX. Ocular neovascularization: Implication of endogenous angiogenic inhibitors and potential therapy. Prog Retin Eye Res 2007; 26:1-37. [PMID: 17074526 DOI: 10.1016/j.preteyeres.2006.09.002] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ocular neovascularization (NV) is the primary cause of blindness in a wide range of ocular diseases, such as diabetic retinopathy (DR) and age-related macular degeneration (AMD). The exact mechanism underlying the pathogenesis of ocular NV is not yet well understood, and as a consequence, there is no satisfactory therapy for ocular NV. In the last 10 years, a number of studies provided increasing evidence demonstrating that the imbalance between angiogenic stimulating factors and angiogenic inhibitors is a major contributor to the angiogenesis induced by various insults, such as hypoxia or ischemia, inflammation and tumor. The angiogenic inhibitors alone or in combination with other existing therapies are, therefore, believed to be promising in the treatment of ocular NV in the near future. This article reviews recent progress in studies on the mechanisms and treatment of ocular NV, focusing on the implication and therapeutic potential of endogenous angiogenic inhibitors in ocular NV.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Medicine Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
25
|
Mitchell CA, Rutland CS, Walker M, Nasir M, Foss AJE, Stewart C, Gerhardt H, Konerding MA, Risau W, Drexler HCA. Unique vascular phenotypes following over-expression of individual VEGFA isoforms from the developing lens. Angiogenesis 2006; 9:209-24. [PMID: 17109192 DOI: 10.1007/s10456-006-9056-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Accepted: 10/13/2006] [Indexed: 01/13/2023]
Abstract
Formation of a correctly organised vasculature and subsequently embryonic survival is critically dependent on the dosage and site-specific expression of VEGF. Murine VEGF exists in three common isoforms (viz. 120, 164 and 188 amino acids) having different organ specific distribution levels. Gene knock-in studies show that expression of any of the individual isoforms of VEGF extends survival until birth, although each is associated with distinct organ-specific abnormalities. Comparison of the effects of VEGF isoform expression is complicated by the general lethality of mis-expression, in addition to cumulative effects of adjacent tissues from the inappropriately patterned vasculature. Here we investigate the effects of over-expression of individual VEGFA isoforms from the lens-specific alphaA-Crystallin promoter and characterise their effects on the vessel morphology of the hyaloid and developing retinal vasculature. Since the hyaloid vasculature is an anatomically distinct, transient vasculature of the eye, comprising 3 cell types (endothelium, pericytes and macrophages) it is possible to more readily interpret the role of individual VEGF-A isoforms in vascular pattern formation in this model. The severity of the vascular phenotype, characterised by a hyperplastic hyaloid at E13.5 and subsequently retinal vascular patterning and ocular defects, is most severe in transgenics over-expressing the more diffusible forms of VEGFA (120 and 164), whereas in VEGFA(188) transgenics the hyaloid vascular defects partially resolve post-natally. The results of this study indicate that individual isoforms of VEGFA induce distinct vascular phenotypes in the eye during embryonic development and that their relative doses provide instructive cues for vascular patterning.
Collapse
Affiliation(s)
- Christopher A Mitchell
- Department of Obstetrics and Gynaecology, University of Nottingham, City Hospital, Hucknall Rd, Nottingham, NG5 1PB, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Since the days of Hans Spemann, the ocular lens has served as one of the most important developmental systems for elucidating fundamental processes of induction and differentiation. More recently, studies in the lens have contributed significantly to our understanding of cell cycle regulation and apoptosis. Over 20 years of accumulated evidence using several different vertebrate species has suggested that fibroblast growth factors (FGFs) and/or fibroblast growth factor receptors (FGFRs) play a key role in lens development. FGFR signaling has been implicated in lens induction, lens cell proliferation and survival, lens fiber differentiation and lens regeneration. Here we will review and discuss historical and recent evidence suggesting that (FGFR) signaling plays a vital and universal role in multiple aspects of lens development.
Collapse
|
27
|
Wu Z, Wang S, Sorenson CM, Sheibani N. Attenuation of retinal vascular development and neovascularization in transgenic mice over-expressing thrombospondin-1 in the lens. Dev Dyn 2006; 235:1908-20. [PMID: 16691615 DOI: 10.1002/dvdy.20837] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Thrombospondin-1 (TSP1) is an endogenous inhibitor of angiogenesis and induces endothelial cell (EC) apoptosis. To study the role TSP1 plays during vascular development and neovascularization, we assessed the effects of ectopic TSP1 expression in the lens on retinal vascularization in transgenic mice. The TSP1 over-expressing mice showed abnormalities in the development of retinal vasculature. There was a dramatic decrease in the density of superficial and deep vascular plexuses of the retina in transgenic mice. The retinal vessels in TSP1 transgenic mice also appeared nonuniform and abnormal in maturation. We detected an increase in the number of EC undergoing apoptosis, which was compensated, in part, by an increase in cell proliferation in retinal vasculature of TSP1 transgenic mice. The TSP1 transgenic mice also exhibited increased levels of vessel obliteration and a limited preretinal neovascularization during oxygen-induced ischemic retinopathy (OIR). Our results indicate increased expression of TSP1 attenuates normal retinal vascularization and preretinal neovascularization during OIR. Therefore, modulation of TSP1 expression may provide an effective mechanism for regulation of ocular angiogenesis.
Collapse
Affiliation(s)
- Zhifeng Wu
- Department of Ophthalmology, University of Wisconsin Medical School, Madison, Wisconsin 53792-4673, USA
| | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Susan X Hsiong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
29
|
Ohlmann A, Scholz M, Goldwich A, Chauhan BK, Hudl K, Ohlmann AV, Zrenner E, Berger W, Cvekl A, Seeliger MW, Tamm ER. Ectopic norrin induces growth of ocular capillaries and restores normal retinal angiogenesis in Norrie disease mutant mice. J Neurosci 2005; 25:1701-10. [PMID: 15716406 PMCID: PMC6725931 DOI: 10.1523/jneurosci.4756-04.2005] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Norrie disease is an X-linked retinal dysplasia that presents with congenital blindness, sensorineural deafness, and mental retardation. Norrin, the protein product of the Norrie disease gene (NDP), is a secreted protein of unknown biochemical function. Norrie disease (Ndp(y/-)) mutant mice that are deficient in norrin develop blindness, show a distinct failure in retinal angiogenesis, and completely lack the deep capillary layers of the retina. We show here that the transgenic expression of ectopic norrin under control of a lens-specific promoter restores the formation of a normal retinal vascular network in Ndp(y/-) mutant mice. The improvement in structure correlates with restoration of neuronal function in the retina. In addition, lenses of transgenic mice with ectopic expression of norrin show significantly more capillaries in the hyaloid vasculature that surrounds the lens during development. In vitro, lenses of transgenic mice in coculture with microvascular endothelial cells induce proliferation of the cells. Transgenic mice with ectopic expression of norrin show more bromodeoxyuridine-labeled retinal progenitor cells at embryonic day 14.5 and thicker retinas at postnatal life than wild-type littermates, indicating a putative direct neurotrophic effect of norrin. These data provide direct evidence that norrin induces growth of ocular capillaries and that pharmacologic modulation of norrin might be used for treatment of the vascular abnormalities associated with Norrie disease or other vascular disorders of the retina.
Collapse
Affiliation(s)
- Andreas Ohlmann
- Department of Anatomy, University of Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kobayashi S, Shinohara H, Tsuneki H, Nagai R, Horiuchi S. N(epsilon)-(carboxymethyl)lysine proliferated CD34(+) cells from rat choroidal explant in culture. Biol Pharm Bull 2005; 27:1382-7. [PMID: 15340223 DOI: 10.1248/bpb.27.1382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Action of N(epsilon)-(carboxymethyl)lysine-human serum albumin (CML-HSA) on neovascularization was investigated in cultured rat choroidal explant. Choroidal explants of normal male Wistar rats were cultured in fibrin gel with Dulbecco's modified Eagle medium containing fetal bovine serum in the presence or absence of CML-HSA. Migrated cells were budded from 2nd day in culture and developed from cultured choroidal explants in a time-dependent manner. Budded and developed cells from the choroidal explant had a feature of fibroblasts, which had attenuated long cytoplasmic processes, long ellipsoid nuclei and numerous membrane-bound polymorphic vesicles. Immunostaining of the attenuated cells in fibrin bed with CD34 (a marker protein of vascular endothelial cells and endothelial progenitor cells) failed to disclose positive result. However the cells which were isolated from fibrin bed by collagenase were specifically stained with anti-CD34 antibody. The isolated cells did not form tube-like structures on collagen gel by 3 weeks in culture. CML-HSA significantly increased the number of total isolated cells and CD34(+) cells as well as the number of vessel-like structures. These results indicate that CML-HSA overproduced immature blood vessels from cultured choroidal explants in fibrin gel, which consisted of CD34(+) cells. The CML-HSA-induced formation of immature blood vessel may be implicated in various choroidal diseases such as age-related macular degeneration.
Collapse
Affiliation(s)
- Shinjiro Kobayashi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokuriku University, 3-Ho Kanagawa-Machi, Kanazawa 920-1181, Japan.
| | | | | | | | | |
Collapse
|
31
|
Vokes SA, Yatskievych TA, Heimark RL, McMahon J, McMahon AP, Antin PB, Krieg PA. Hedgehog signaling is essential for endothelial tube formation during vasculogenesis. Development 2004; 131:4371-80. [PMID: 15294868 DOI: 10.1242/dev.01304] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
During embryonic development, the first blood vessels are formed through the aggregation and subsequent assembly of angioblasts (endothelial precursors) into a network of endothelial tubes, a process known as vasculogenesis. These first vessels generally form in mesoderm that is adjacent to endodermal tissue. Although specification of the angioblast lineage is independent of endoderm interactions, a signal from the endoderm is necessary for angioblasts to assemble into a vascular network and to undergo vascular tube formation. In this study, we show that endodermally derived sonic hedgehog is both necessary and sufficient for vascular tube formation in avian embryos. We also show that Hedgehog signaling is required for vascular tube formation in mouse embryos, and for vascular cord formation in cultured mouse endothelial cells. These results demonstrate a previously uncharacterized role for Hedgehog signaling in vascular development, and identify Hedgehog signaling as an important component of the molecular pathway leading to vascular tube formation.
Collapse
Affiliation(s)
- Steven A Vokes
- Department of Cell Biology and Anatomy, University of Arizona Health Sciences Center, 1501 North Campbell Avenue, Tucson, AZ 85724-5044, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Peterson RT, Shaw SY, Peterson TA, Milan DJ, Zhong TP, Schreiber SL, MacRae CA, Fishman MC. Chemical suppression of a genetic mutation in a zebrafish model of aortic coarctation. Nat Biotechnol 2004; 22:595-9. [PMID: 15097998 DOI: 10.1038/nbt963] [Citation(s) in RCA: 324] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2003] [Accepted: 02/13/2004] [Indexed: 11/08/2022]
Abstract
Conventional drug discovery approaches require a priori selection of an appropriate molecular target, but it is often not obvious which biological pathways must be targeted to reverse a disease phenotype. Phenotype-based screens offer the potential to identify pathways and potential therapies that influence disease processes. The zebrafish mutation gridlock (grl, affecting the gene hey2) disrupts aortic blood flow in a region and physiological manner akin to aortic coarctation in humans. Here we use a whole-organism, phenotype-based, small-molecule screen to discover a class of compounds that suppress the coarctation phenotype and permit survival to adulthood. These compounds function during the specification and migration of angioblasts. They act to upregulate expression of vascular endothelial growth factor (VEGF), and the activation of the VEGF pathway is sufficient to suppress the gridlock phenotype. Thus, organism-based screens allow the discovery of small molecules that ameliorate complex dysmorphic syndromes even without targeting the affected gene directly.
Collapse
|
33
|
Ozawa CR, Banfi A, Glazer NL, Thurston G, Springer ML, Kraft PE, McDonald DM, Blau HM. Microenvironmental VEGF concentration, not total dose, determines a threshold between normal and aberrant angiogenesis. J Clin Invest 2004; 113:516-27. [PMID: 14966561 PMCID: PMC338257 DOI: 10.1172/jci18420] [Citation(s) in RCA: 384] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2003] [Accepted: 12/16/2003] [Indexed: 12/13/2022] Open
Abstract
Use of long-term constitutive expression of VEGF for therapeutic angiogenesis may be limited by the growth of abnormal blood vessels and hemangiomas. We investigated the relationship between VEGF dosage and the morphology and function of newly formed blood vessels by implanting retrovirally transduced myoblasts that constitutively express VEGF164 into muscles of adult mice. Reducing VEGF dosage by decreasing the total number of VEGF myoblasts implanted did not prevent vascular abnormalities. However, when clonal populations of myoblasts homogeneously expressing different levels of VEGF were implanted, a threshold between normal and aberrant angiogenesis was found. Clonal myoblasts that expressed low to medium levels of VEGF induced growth of stable, pericyte-coated capillaries of uniform size that were not leaky and became VEGF independent, as shown by treatment with the potent VEGF blocker VEGF-TrapR1R2. In contrast, clones that expressed high levels of VEGF induced hemangiomas. Remarkably, when different clonal populations were mixed, even a small proportion of cells with high production of VEGF was sufficient to cause hemangioma growth. These results show for the first time to our knowledge that the key determinant of whether VEGF-induced angiogenesis is normal or aberrant is the microenvironmental amount of growth factor secreted, rather than the overall dose. Long-term continuous delivery of VEGF, when maintained below a threshold microenvironmental level, can lead to normal angiogenesis without other exogenous growth factors.
Collapse
Affiliation(s)
- Clare R Ozawa
- Baxter Laboratory in Genetic Pharmacology, Stanford University School of Medicine, Stanford, California 94305-5175, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kobayashi S, Suzuki M, Tsuneki H, Nagai R, Horiuchi S, Hagino N. Overproduction of N.EPSILON.-(Carboxymethyl)lysine-Induced Neovascularization in Cultured Choroidal Explant of Streptozotocin-Diabetic Rat. Biol Pharm Bull 2004; 27:1565-71. [PMID: 15467196 DOI: 10.1248/bpb.27.1565] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Action of N(epsilon)-(carboxymethyl)lysine (CML) adduct, an advanced glycation end product, was investigated on neovascularization of cultured choroidal explants in streptozotocin (STZ)-diabetic rat. The choroidal explants of early (4 weeks after an injection of 60 mg/kg STZ) and advanced (8 months after the STZ injection) diabetic rats, and age-matched normal rats were cultured in fibrin gel with Dulbecco's modified Eagle medium containing fetal bovine serum. The number of budded microvessel-like structures was counted and used as an index of in vitro neovascularization. Choroidal explants in the early diabetic stage released vascular endothelial growth factor (VEGF) and tended to increase tumor necrosis factor (TNF) alpha and platelet-derived growth factor (PDGF)-B, and concomitantly facilitated growth of sprout and buds, compared to the normal control. When choroidal explants were stimulated with CML-human serum albumin (HSA), its releasing effect was in the order VEGF>TNFalpha>PDGF-B. CML-HSA and CML-bovine serum albumin augmented the neovascularization in the cultured diabetic explant and their actions did not virtually differ. A monoclonal anti-CML antibody (6D12) inhibited the neovascularization in the advanced diabetes greater than that in the early diabetes. Inhibitory actions of anti-VEGF and anti-TNFalpha antibodies on the neovascularization were similar to that of the anti-CML antibody in the diabetes. In conclusion, CML adducts were accumulated and over-produced the actions of VEGF, TNFalpha and PDGF-B in the choroidal explant during diabetes in an age-dependent manner. TNFalpha and VEGF are likely to play a predominant role for the CML-induced choroidal neovascularization.
Collapse
Affiliation(s)
- Shinjiro Kobayashi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanagawa-Machi, Kanazawa, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Hagedorn M, Balke M, Schmidt A, Bloch W, Kurz H, Javerzat S, Rousseau B, Wilting J, Bikfalvi A. VEGF coordinates interaction of pericytes and endothelial cells during vasculogenesis and experimental angiogenesis. Dev Dyn 2004; 230:23-33. [PMID: 15108306 DOI: 10.1002/dvdy.20020] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Biological activities of vascular endothelial growth factor (VEGF) have been studied extensively in endothelial cells (ECs), but few data are available regarding its effects on pericytes. In murine embryoid body cultures, VEGF-induced expression of desmin and alpha-smooth muscle actin (alpha-SMA) in CD-31+ cells. The number of CD-31+/desmin+ vascular chords increased with VEGF treatment time and peaked during a differentiation window between 6 and 9 days after plating. In vivo, VEGF-induced elongation and migration of desmin-positive pericytes and coverage of angiogenic capillaries, as revealed by analysis of Sambucus nigra lectin-stained vascular beds of the chick chorioallantoic membrane. VEGF also caused significant decrease of intercapillary spaces, an indicator for intussusceptive vascular growth. These VEGF-mediated effects point at a more intricate interaction between ECs and pericytes cells than previously demonstrated and suggest that pericytes may be derived from EC progenitors in vitro and not only stabilize capillaries but also participate in vascular remodeling in vivo.
Collapse
Affiliation(s)
- Martin Hagedorn
- INSERM E0113 Molecular Mechanisms of Angiogenesis, University Bordeaux I, Talence, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Linden T, Katschinski DM, Eckhardt K, Scheid A, Pagel H, Wenger RH. The antimycotic ciclopirox olamine induces HIF-1alpha stability, VEGF expression, and angiogenesis. FASEB J 2003; 17:761-3. [PMID: 12594177 DOI: 10.1096/fj.02-0586fje] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The heterodimeric hypoxia-inducible factor (HIF)-1 is a master regulator of oxygen homeostasis. Protein stability and transactivation function of the alpha subunit are controlled by iron- and oxygen-dependent hydroxylation of proline and asparagine residues. The anti-mycotic ciclopirox olamine (CPX) is a lipophilic bidentate iron chelator that stabilizes HIF-1alpha under normoxic conditions at lower concentrations than other iron chelators, probably by inhibiting HIF-1alpha hydroxylation. As shown by the inhibition of iron-dependent quenching of FITC-labeled deferoxamine (DFX) fluorescence, CPX appears to have an even higher affinity for iron than DFX. Initial observations that treatment with 1% CPX, but not with placebo, occasionally caused reddening of wound margins in a mouse skin wound model prompted us to investigate the capability of CPX to induce angiogenesis. CPX-induced HIF-1-mediated reporter gene activity and endogenous HIF-1 target gene expression, including elevation of transcription, mRNA, and protein levels of the vascular endothelial growth factor (VEGF). In the chick chorioallantoic membrane assay, inert polymer disks containing CPX but not the solvent alone induced angiogenesis. In summary, these results suggest that CPX induces angiogenesis in vivo via HIF-1 and VEGF induction. Therefore, CPX might serve as an alternative to recombinant VEGF treatment or to VEGF gene therapy for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Tobias Linden
- Institute of Physiology, Medical University of Lübeck, Lübeck, Germany
| | | | | | | | | | | |
Collapse
|
37
|
Bikfalvi A, Bicknell R. Recent advances in angiogenesis, anti-angiogenesis and vascular targeting. Trends Pharmacol Sci 2002; 23:576-82. [PMID: 12457776 DOI: 10.1016/s0165-6147(02)02109-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis, the development of new blood vessels, has become a major focus of research. This has been stimulated by the therapeutic opportunities offered by the ability to manipulate the vasculature in pathologies such as cancer. Here, we present an overview of recent advances in angiogenesis. Especially noteworthy is the large volume of information from developmental studies, particularly those that involve transgenic and gene knockout mice. We also discuss the increasing repertoire of drugs with which to manipulate angiogenesis and new endothelial-specific genes with which to target the vasculature.
Collapse
Affiliation(s)
- Andreas Bikfalvi
- INSERM E 0113 Molecular Mechanisms of Angiogenesis and Growth Factor and Cell Differentiation Laboratory, University Bordeaux I, France.
| | | |
Collapse
|
38
|
Paradis H, Liu CY, Saika S, Azhar M, Doetschman T, Good WV, Nayak R, Laver N, Kao CWC, Kao WWY, Gendron RL. Tubedown-1 in remodeling of the developing vitreal vasculature in vivo and regulation of capillary outgrowth in vitro. Dev Biol 2002; 249:140-55. [PMID: 12217325 DOI: 10.1006/dbio.2002.0757] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tubedown-1 (tbdn-1) is a mammalian homologue of the N-terminal acetyltransferase subunit NAT1 of Saccharomyces cerevisiae and copurifies with an acetyltransferase activity. Tbdn-1 expression in endothelial cells becomes downregulated during the formation of capillary-like structures in vitro and is regulated in vivo in a manner which suggests a functional role in dampening blood vessel development. Here we show that tbdn-1 is expressed highly in the vitreal vascular network (tunica vasculosa lentis and vasa hyaloidea propria) during the pruning and remodeling phases of this transient structure. The vitreal blood vessels of mice harboring a targeted inactivation of TGF-beta2 fail to remodel and abnormally accumulate, a phenomenon reminiscent of the ocular pathology resembling persistent fetal vasculature (PFV) in humans. Since suppression of normal tbdn-1 expression has been previously observed in retinal vessel proliferation, we analyzed vitreal vascular changes and tbdn-1 expression in TGF-beta2(-/-) eyes. The nuclei of vitreal vessel endothelial cells in TGF-beta2(-/-) eyes express proliferating cell nuclear antigen (PCNA) and exhibit increased levels of active (P42/44)mitogen-activated protein kinase (phospho-(P42/44)MAPK), characteristics consistent with proliferative endothelial cells. In contrast to normal vitreal vessels, collagen IV expression exhibited a disorganized pattern in the TGF-beta2(-/-) vitreal vessels, suggesting vessel disorganization and possibly a breakdown of vessel basal laminae. Moreover, vitreal vessels of TGF-beta2(-/-) mice lack expression of pericyte markers (CD13, alpha smooth muscle actin) and show ultrastructural changes consistent with pericyte degeneration. The accumulating vitreal blood vessels of TGF-beta2(-/-) mice, while maintaining expression of the endothelial marker von Willebrand Factor, show a significant decrease in the expression of tbdn-1. We addressed the functional role of tbdn-1 in the regulation of vitreal blood vessels using an in vitro model of choroid-retina capillary outgrowth. Clones of the RF/6A fetal choroid-retina endothelial cell line showing suppression of tbdn-1 levels after overexpression of an antisense TBDN-1 cDNA display a significant increase in the formation of capillary-like structures in vitro compared with controls. These findings suggest that tbdn-1 inhibits capillary-like formation in vitro and may serve to dampen vitreal blood vessel formation preceding the regression of the vitreal vasculature during development. Our results also suggest that tbdn-1 may participate with TGF-beta2 in regulating normal development of the vitreal vasculature.
Collapse
Affiliation(s)
- H Paradis
- Division of Basic Medical Sciences, Department of Medicine, Memorial University of Newfoundland, St. John's NF, A1B 3V6, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Vokes SA, Krieg PA. Endoderm is required for vascular endothelial tube formation, but not for angioblast specification. Development 2002; 129:775-85. [PMID: 11830576 DOI: 10.1242/dev.129.3.775] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Angioblasts, the precursor cells that comprise the endothelial layer of blood vessels, arise from a purely mesodermal population. Individual angioblasts coalesce to form the primary vascular plexus through a process called vasculogenesis. A number of reports in the literature suggest that signals from the adjacent endoderm are necessary to induce angioblast specification within the mesoderm. We present evidence, using both embryological and molecular techniques, indicating that endoderm is not necessary for the induction of angioblasts. Xenopus embryos that had endoderm physically removed at the onset of gastrulation still express vascular markers. Furthermore, animal caps stimulated with bFGF form angioblasts in the absence of any detectable endodermal markers. These results show that endoderm is not required for the initial formation of angioblasts. While Xenopus embryos lacking endoderm contain aggregates of angioblasts, these angioblasts fail to assemble into endothelial tubes. Endothelial tube formation can be rescued, however, by implantation of endodermal tissue from sibling embryos. Based on these studies in Xenopus, and corroborating experiments using the quail embryo, we conclude that endoderm is not required for angioblast specification, but does play an essential role in the formation of vascular tubes.
Collapse
Affiliation(s)
- Steven A Vokes
- Department of Cell Biology and Anatomy, University of Arizona Health Sciences Center, 1501 N. Campbell Avenue, PO Box 245044, Tucson, AZ 85724, USA
| | | |
Collapse
|
40
|
Kinder SJ, Loebel DA, Tam PP. Allocation and early differentiation of cardiovascular progenitors in the mouse embryo. Trends Cardiovasc Med 2001; 11:177-84. [PMID: 11597828 DOI: 10.1016/s1050-1738(01)00091-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
During gastrulation of the mouse embryo, progenitor cells of the endothelium of blood vessels are allocated to different compartments of the extraembryonic and embryonic tissues in accordance to the timing and the site of recruitment to the mesodermal layer. In the yolk sac, the endothelium and the erythropoietic progenitors are populated by different groups of mesodermal cells, suggesting that they may not be derived from a common pool of progenitors. An orderly pattern of movement of mesodermal cells and the provision of proper intercellular transforming growth factor beta (TGF beta) and vascular endothelial growth factor (VEGF) signaling by neighboring germ layer tissues are essential for normal morphogenesis of the vasculature.
Collapse
Affiliation(s)
- S J Kinder
- Embryology Unit, Children's Medical Research Institute, Wentworth, NSW, Australia
| | | | | |
Collapse
|
41
|
Vinores SA, Seo MS, Okamoto N, Ash JD, Wawrousek EF, Xiao WH, Hudish T, Derevjanik NL, Campochiaro PA. Experimental models of growth factor-mediated angiogenesis and blood-retinal barrier breakdown. GENERAL PHARMACOLOGY 2000; 35:233-9. [PMID: 11888678 DOI: 10.1016/s0306-3623(01)00117-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Following chronic ischemia, vascular endothelial growth factor (VEGF) is induced primarily in the ganglion cell layer of the retina. This often results in neovascularization (NV) that originates from the vascular bed closest to the ganglion cell layer. To study the effects of VEGF, independent lines of transgenic mice that express VEGF in the lens and in the retina have been generated. Expression in the lens results in excessive proliferation and accumulation of angioblasts and endothelial cells in proximity to the lens. However, VEGF expression is not sufficient to direct blood vessel organization or maturation in the prenatal mouse. Abnormal vessels do form on the retinal surface, but not until the second postnatal week. In transgenic mice expressing VEGF in the photoreceptors, NV originates from the deep capillary bed--the vascular bed closest to the photoreceptors. NV is accompanied by localized blood-retinal barrier breakdown. NV is also induced in PDGF-B transgenic mice. PDGF-B expression in the lens occurs prenatally and, during this time, mainly affects the perilenticular vessels. Postnatally, transgenic mice expressing PDGF-B in the lens or photoreceptors show a similar phenotype. In both models, a highly vascularized cell mass containing endothelial cells, pericytes, and glia forms in the superficial retina, and the formation of the deep capillary bed is inhibited. The phenotype suggests that an additional factor is necessary for the maturation and penetration of vascular endothelial cells into the retina to form the deep capillary bed.
Collapse
Affiliation(s)
- S A Vinores
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, 825 Maumenee Building, 600 North Wolfe Street, 21287-9289, Baltimore, MD 21287-9289, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|