1
|
Vaglienti MV, Subirada PV, Joray MB, Bonacci G, Sánchez MC. Protective Effect of NO 2-OA on Oxidative Stress, Gliosis, and Pro-Angiogenic Response in Müller Glial Cells. Cells 2023; 12:cells12030494. [PMID: 36766836 PMCID: PMC9914399 DOI: 10.3390/cells12030494] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 02/05/2023] Open
Abstract
Inflammation and oxidative and nitrosative stress are involved in the pathogenesis of proliferative retinopathies (PR). In PR, a loss of balance between pro-angiogenic and anti-angiogenic factors favors the secretion of vascular endothelial growth factor (VEGF). This vascular change results in alterations in the blood-retinal barrier, with extravasation of plasma proteins such as α2-macroglobulin (α2M) and gliosis in Müller glial cells (MGCs, such as MIO-M1). It is well known that MGCs play important roles in healthy and sick retinas, including in PR. Nitro-fatty acids are electrophilic lipid mediators with anti-inflammatory and cytoprotective properties. Our aim was to investigate whether nitro-oleic acid (NO2-OA) is beneficial against oxidative stress, gliosis, and the pro-angiogenic response in MGCs. Pure synthetic NO2-OA increased HO-1 expression in a time- and concentration-dependent manner, which was abrogated by the Nrf2 inhibitor trigonelline. In response to phorbol 12-myristate 13-acetate (PMA) and lipopolysaccharide (LPS), NO2-OA prevented the ROS increase and reduced the gliosis induced by α2M. Finally, when hypoxic MGCs were incubated with NO2-OA, the increase in VEGF mRNA expression was not affected, but under hypoxia and inflammation (IL-1β), NO2-OA significantly reduced VEGF mRNA levels. Furthermore, NO2-OA inhibited endothelial cell (BAEC) tubulogenesis. Our results highlight NO2-OA's protective effect on oxidative damage, gliosis; and the exacerbated pro-angiogenic response in MGCs.
Collapse
Affiliation(s)
- María V. Vaglienti
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| | - Paula V. Subirada
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| | - Mariana B. Joray
- Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Córdoba 5000, Argentina
- Instituto de Investigaciones en Recursos Naturales y Sustentabilidad José Sánchez Labrador J. S., Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| | - Gustavo Bonacci
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
- Correspondence: (G.B.); (M.C.S.)
| | - María C. Sánchez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
- Correspondence: (G.B.); (M.C.S.)
| |
Collapse
|
2
|
Asare-Bediako B, Adu-Agyeiwaah Y, Abad A, Li Calzi S, Floyd JL, Prasad R, DuPont M, Asare-Bediako R, Bustelo XR, Grant MB. Hematopoietic Cells Influence Vascular Development in the Retina. Cells 2022; 11:3207. [PMID: 36291075 PMCID: PMC9601270 DOI: 10.3390/cells11203207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic cells play a crucial role in the adult retina in health and disease. Monocytes, macrophages, microglia and myeloid angiogenic cells (MACs) have all been implicated in retinal pathology. However, the role that hematopoietic cells play in retinal development is understudied. The temporal changes in recruitment of hematopoietic cells into the developing retina and the phenotype of the recruited cells are not well understood. In this study, we used the hematopoietic cell-specific protein Vav1 to track and investigate hematopoietic cells in the developing retina. By flow cytometry and immunohistochemistry, we show that hematopoietic cells are present in the retina as early as P0, and include microglia, monocytes and MACs. Even before the formation of retinal blood vessels, hematopoietic cells localize to the inner retina where they eventually form networks that intimately associate with the developing vasculature. Loss of Vav1 lead to a reduction in the density of medium-sized vessels and an increased inflammatory response in retinal astrocytes. When pups were subjected to oxygen-induced retinopathy, hematopoietic cells maintained a close association with the vasculature and occasionally formed 'frameworks' for the generation of new vessels. Our study provides further evidence for the underappreciated role of hematopoietic cells in retinal vasculogenesis and the formation of a healthy retina.
Collapse
Affiliation(s)
- Bright Asare-Bediako
- Vision Science Graduate Program, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | - Yvonne Adu-Agyeiwaah
- Vision Science Graduate Program, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | - Antonio Abad
- Centro de Investigación del Cáncer de Salamanca, CSIC and University of Salamanca, 37007 Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBER), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | - Jason L. Floyd
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | - Ram Prasad
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | - Mariana DuPont
- Vision Science Graduate Program, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | - Richmond Asare-Bediako
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY 14623, USA
| | - Xose R. Bustelo
- Centro de Investigación del Cáncer de Salamanca, CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Alabama, Birmingham, AL 35294, USA
| |
Collapse
|
3
|
Goenka V, Borkar T, Desai A, Das RK. Therapeutic potential of mesenchymal stem cells in treating both types of diabetes mellitus and associated diseases. J Diabetes Metab Disord 2020; 19:1979-1993. [PMID: 33520872 PMCID: PMC7843693 DOI: 10.1007/s40200-020-00647-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/24/2020] [Indexed: 10/23/2022]
Abstract
Diabetes mellitus is a common lifestyle disease which can be classified into type 1 diabetes mellitus and type 2 diabetes mellitus. While both result in hyperglycemia due to lack of insulin action and further associated chronic ailments, there is a marked distinction in the cause for each type due to which both require a different prophylaxis. As observed, type 1 diabetes is caused due to the autoimmune action of the body resulting in the destruction of pancreatic islet cells. On the other hand, type 2 diabetes is caused either due to insulin resistance of target cells or lack of insulin production as per physiological requirements. Attempts to cure the disease have been made by bringing drastic changes in the patients' lifestyle; parenteral administration of insulin; prescription of drugs such as biguanides, meglitinides, and amylin; pancreatic transplantation; and immunotherapy. While these attempts cause a certain degree of relief to the patient, none of these can cure diabetes mellitus. However, a new treatment strategy led by the discovery of mesenchymal stem cells and their unique immunomodulatory and multipotent properties has inspired therapies to treat diabetes by essentially reversing the conditions causing the disease. The current review aims to enumerate the role of various mesenchymal stem cells and the different approaches to treat both types of diabetes and its associated diseases as well.
Collapse
Affiliation(s)
- Vidul Goenka
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu India
| | - Tanhai Borkar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu India
| | - Aska Desai
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu India
| | - Raunak Kumar Das
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, Tamil Nadu India
| |
Collapse
|
4
|
Bertelli PM, Pedrini E, Guduric-Fuchs J, Peixoto E, Pathak V, Stitt AW, Medina RJ. Vascular Regeneration for Ischemic Retinopathies: Hope from Cell Therapies. Curr Eye Res 2020; 45:372-384. [PMID: 31609636 DOI: 10.1080/02713683.2019.1681004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022]
Abstract
Retinal vascular diseases, such as diabetic retinopathy, retinopathy of prematurity, retinal vein occlusion, ocular ischemic syndrome and ischemic optic neuropathy, are leading causes of vision impairment and blindness. Whilst drug, laser or surgery-based treatments for the late stage complications of many of these diseases are available, interventions that target the early vasodegenerative stages are lacking. Progressive vasculopathy and ensuing ischemia is an underpinning pathology in many of these diseases, leading to hypoperfusion, hypoxia, and ultimately pathological neovascularization and/or edema in the retina and other ocular tissues, such as the optic nerve and iris. Therefore, repairing the retinal vasculature may prevent progression of ischemic retinopathies into late stage vascular complications. Various cell types have been explored for their vascular repair potential. Endothelial progenitor cells, mesenchymal stem cells and induced pluripotent stem cells are studied for their potential to integrate with the damaged retinal vasculature and limit ischemic injury. Clinical trials for some of these cell types have confirmed safety and feasibility in the treatment of ischemic diseases, including some retinopathies. Another promising avenue is mobilization of endogenous endothelial progenitors, whereby reparative cells are moved from their niche to circulating blood to target and home into ischemic tissues. Several aspects and properties of these cell types have yet to be elucidated. Nevertheless, we foresee that cell therapy, whether through delivery of exogenous or enhancement of endogenous reparative cells, will become a valuable and beneficial treatment for ischemic retinopathies.
Collapse
Affiliation(s)
- Pietro Maria Bertelli
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Edoardo Pedrini
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Jasenka Guduric-Fuchs
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Elisa Peixoto
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Varun Pathak
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Alan W Stitt
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Reinhold J Medina
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University Belfast, Belfast, UK
| |
Collapse
|
5
|
Alabi RO, Farber G, Blobel CP. Intriguing Roles for Endothelial ADAM10/Notch Signaling in the Development of Organ-Specific Vascular Beds. Physiol Rev 2019; 98:2025-2061. [PMID: 30067156 DOI: 10.1152/physrev.00029.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The vasculature is a remarkably interesting, complex, and interconnected organ. It provides a conduit for oxygen and nutrients, filtration of waste products, and rapid communication between organs. Much remains to be learned about the specialized vascular beds that fulfill these diverse, yet vital functions. This review was prompted by the discovery that Notch signaling in mouse endothelial cells is crucial for the development of specialized vascular beds found in the heart, kidneys, liver, intestines, and bone. We will address the intriguing questions raised by the role of Notch signaling and that of its regulator, the metalloprotease ADAM10, in the development of specialized vascular beds. We will cover fundamentals of ADAM10/Notch signaling, the concept of Notch-dependent cell fate decisions, and how these might govern the development of organ-specific vascular beds through angiogenesis or vasculogenesis. We will also consider common features of the affected vessels, including the presence of fenestra or sinusoids and their occurrence in portal systems with two consecutive capillary beds. We hope to stimulate further discussion and study of the role of ADAM10/Notch signaling in the development of specialized vascular structures, which might help uncover new targets for the repair of vascular beds damaged in conditions like coronary artery disease and glomerulonephritis.
Collapse
Affiliation(s)
- Rolake O Alabi
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, New York ; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York ; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York ; Department of Medicine, Weill Cornell Medicine, New York, New York ; and Institute for Advanced Study, Technical University Munich , Munich , Germany
| | - Gregory Farber
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, New York ; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York ; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York ; Department of Medicine, Weill Cornell Medicine, New York, New York ; and Institute for Advanced Study, Technical University Munich , Munich , Germany
| | - Carl P Blobel
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, New York ; Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York ; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York ; Department of Medicine, Weill Cornell Medicine, New York, New York ; and Institute for Advanced Study, Technical University Munich , Munich , Germany
| |
Collapse
|
6
|
Galectin-1 expression imprints a neurovascular phenotype in proliferative retinopathies and delineates responses to anti-VEGF. Oncotarget 2018; 8:32505-32522. [PMID: 28455954 PMCID: PMC5464805 DOI: 10.18632/oncotarget.17129] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
Neovascular retinopathies are leading causes of irreversible blindness. Although vascular endothelial growth factor (VEGF) inhibitors have been established as the mainstay of current treatment, clinical management of these diseases is still limited. As retinal impairment involves abnormal neovascularization and neuronal degeneration, we evaluated here the involvement of galectin-1 in vascular and non-vascular alterations associated with retinopathies, using the oxygen-induced retinopathy (OIR) model. Postnatal day 17 OIR mouse retinas showed the highest neovascular profile and exhibited neuro-glial injury as well as retinal functional loss, which persisted until P26 OIR. Concomitant to VEGF up-regulation, galectin-1 was highly expressed in P17 OIR retinas and it was mainly localized in neovascular tufts. In addition, OIR induced remodelling of cell surface glycophenotype leading to exposure of galectin-1-specific glycan epitopes. Whereas VEGF returned to baseline levels at P26, increased galectin-1 expression persisted until this time period. Remarkably, although anti-VEGF treatment in P17 OIR improved retinal vascularization, neither galectin-1 expression nor non-vascular and functional alterations were attenuated. However, this functional defect was partially prevented in galectin-1-deficient (Lgals1-/-) OIR mice, suggesting the importance of targeting both VEGF and galectin-1 as non-redundant independent pathways. Supporting the clinical relevance of these findings, we found increased levels of galectin-1 in aqueous humor from patients with proliferative diabetic retinopathy and neovascular glaucoma. Thus, using an OIR model and human samples, we identified a role for galectin-1 accompanying vascular and non-vascular retinal alterations in neovascular retinopathies.
Collapse
|
7
|
Wang JD, An Y, Zhang JS, Wan XH, Jonas JB, Xu L, Zhang W. Human bone marrow mesenchymal stem cells for retinal vascular injury. Acta Ophthalmol 2017; 95:e453-e461. [PMID: 27807930 DOI: 10.1111/aos.13154] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/10/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE To examine the potential of intravitreally implanted human bone marrow-derived mesenchymal stem cells (BMSCs) to affect vascular repair and the blood-retina barrier in mice and rats with oxygen-induced retinopathy, diabetic retinopathy or retinal ischaemia-reperfusion damage. METHODS Three study groups (oxygen-induced retinopathy group: 18 C57BL/6J mice; diabetic retinopathy group: 15 rats; retinal ischaemia-reperfusion model: 18 rats) received BMSCs injected intravitreally. Control groups (oxygen-induced retinopathy group: 12 C57BL/6J mice; diabetic retinopathy group: 15 rats; retinal ischaemia-reperfusion model: 18 rats) received an intravitreal injection of phosphate-buffered saline. We applied immunohistological techniques to measure retinal vascularization, spectroscopic measurements of intraretinally extravasated fluorescein-conjugated dextran to quantify the blood-retina barrier breakdown, and histomorphometry to assess retinal thickness and retinal ganglion cell count. RESULTS In the oxygen-induced retinopathy model, the study group with intravitreally injected BMSCs as compared with the control group showed a significantly (p = 0.001) smaller area of retinal neovascularization. In the diabetic retinopathy model, study group and control group did not differ significantly in the amount of intraretinally extravasated dextran. In the retinal ischaemia-reperfusion model, on the 7th day after retina injury, the retina was significantly thicker in the study group than in the control group (p = 0.02), with no significant difference in the retinal ganglion cell count (p = 0.36). CONCLUSIONS Intravitreally implanted human BMSCs were associated with a reduced retinal neovascularization in the oxygen-induced retinopathy model and with a potentially cell preserving effect in the retinal ischaemia-reperfusion model. Intravitreal BMSCs may be of potential interest for the therapy of retinal vascular disorders.
Collapse
Affiliation(s)
- Jin-Da Wang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Ying An
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Jing-Shang Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Xiu-Hua Wan
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Jost B. Jonas
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
- Department of Ophthalmology; Medical Faculty Mannheim of the Ruprecht-Karls-University Heidelberg; Seegartenklinik; Heidelberg Germany
| | - Liang Xu
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| | - Wei Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Tongren Hospital of Capital Medical University; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing China
| |
Collapse
|
8
|
Wang JD, An Y, Zhang JS, Wan XH, Zhang W, Lanza R, Lu SJ, Jonas JB, Xu L. Retinal vascular injuries and intravitreal human embryonic stem cell-derived haemangioblasts. Acta Ophthalmol 2017. [PMID: 28636206 DOI: 10.1111/aos.13477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To investigate whether intravitreally applied haemangioblasts (HB) derived from human embryonic stem cells (hESCs) are helpful for the repair of vascular damage caused in animals by an oxygen-induced retinopathy (OIR), by an induced diabetic retinopathy (DR) or by an induced retinal ischaemia with subsequent reperfusion. METHODS Human embryonic stem cell-derived HBs were transplanted intravitreally into C57BL/6J mice (OIR model), into male Wistar rats with an induced DR and into male Wistar rats undergoing induced retinal ischaemia with subsequent reperfusion. Control groups of animals received an intravitreal injection of endothelial cells (ECs) or phosphate-buffered saline (PBS). We examined the vasculature integrity in the mice with OIR, the blood-retina barrier in the rats with induced DR, and retinal thickness and retinal ganglion cell density in retina flat mounts of the rats with the retinal ischaemic-reperfusion retinopathy. RESULTS In the OIR model, the study group versus control groups showed a significantly (p < 0.001) smaller retinal avascular area [5.1 ± 2.7%;n = 18 animals versus 12.2 ± 2.8% (PBS group; n = 10 animals) and versus 11.8 ± 3.7% (EC group; n = 8 animals)] and less retinal neovascularization [6.3 ± 2.5%;n = 18 versus 15.2 ± 6.3% (n = 10; PBS group) and versus 15.8 ± 3.3% (n = 8; EC group)]. On retinal flat mounts, hESC-HBs were integrated into damaged retinal vessels and stained positive for PECAM (CD31) as EC marker. In the DR model, the study group versus the EC control group showed a significantly (p = 0.001) better blood-retina barrier function as measured at 2 days after the intravitreal injections [study group: 20.2 ± 12.8 μl/(g × hr); n = 6; versus EC control group: 52.9 ± 9.9 μl/(g × hr; n = 6)]. In the retinal ischaemia-reperfusion model, the groups did not differ significantly in retinal thickness and retinal ganglion cell density at 2, 5 and 7 days after baseline. CONCLUSION By integrating into damaged retinal vessels and differentiating into ECs, intravitreally administered hESC-HBs may have partially repaired a retinal vascular injury caused by OIR model and DR.
Collapse
Affiliation(s)
- Jin-Da Wang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Ying An
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Jing-Shang Zhang
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Xiu-Hua Wan
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| | - Wei Zhang
- University of North Texas Health Science Center; Fort Worth Texas USA
| | - Robert Lanza
- Ocata Therapeutics; Marlborough Massachusetts USA
| | - Shi-Jiang Lu
- Ocata Therapeutics; Marlborough Massachusetts USA
| | - Jost B. Jonas
- Department of Ophthalmology; Medical Faculty Mannheim of the Ruprecht-Karls-University Heidelberg; Mannheim Germany
| | - Liang Xu
- Beijing Institute of Ophthalmology; Beijing Tongren Eye Center; Beijing Key Laboratory of Ophthalmology and Visual Sciences; Beijing Tongren Hospital of Capital Medical University; Beijing China
| |
Collapse
|
9
|
The Role of Myeloid-Derived Suppressor Cells (MDSC) in Cancer Progression. Vaccines (Basel) 2016; 4:vaccines4040036. [PMID: 27827871 PMCID: PMC5192356 DOI: 10.3390/vaccines4040036] [Citation(s) in RCA: 271] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/04/2016] [Accepted: 10/31/2016] [Indexed: 12/27/2022] Open
Abstract
The immunosuppressive tumor microenvironment represents not only one of the key factors stimulating tumor progression but also a strong obstacle for efficient tumor immunotherapy. Immunosuppression was found to be associated with chronic inflammatory mediators including cytokines, chemokines and growth factors produced by cancer and stroma cells. Long-term intensive production of these factors induces the formation of myeloid-derived suppressor cells (MDSCs) representing one of the most important players mediating immunosuppression. Moreover, MDSCs could not only inhibit anti-tumor immune reactions but also directly stimulate tumor growth and metastasis. Therefore, understanding the mechanisms of their generation, expansion, recruitment and activation is required for the development of novel strategies for tumor immunotherapy.
Collapse
|
10
|
Çerman E, Akkoç T, Eraslan M, Şahin Ö, Özkara S, Vardar Aker F, Subaşı C, Karaöz E, Akkoç T. Retinal Electrophysiological Effects of Intravitreal Bone Marrow Derived Mesenchymal Stem Cells in Streptozotocin Induced Diabetic Rats. PLoS One 2016; 11:e0156495. [PMID: 27300133 PMCID: PMC4907488 DOI: 10.1371/journal.pone.0156495] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 05/16/2016] [Indexed: 01/09/2023] Open
Abstract
Diabetic retinopathy is the most common cause of legal blindness in developed countries at middle age adults. In this study diabetes was induced by streptozotocin (STZ) in male Wistar albino rats. After 3 months of diabetes, rights eye were injected intravitreally with green fluorescein protein (GFP) labelled bone marrow derived stem cells (BMSC) and left eyes with balanced salt solution (Sham). Animals were grouped as Baseline (n = 51), Diabetic (n = 45), Diabetic+BMSC (n = 45 eyes), Diabetic+Sham (n = 45 eyes), Healthy+BMSC (n = 6 eyes), Healthy+Sham (n = 6 eyes). Immunohistology analysis showed an increased retinal gliosis in the Diabetic group, compared to Baseline group, which was assessed with GFAP and vimentin expression. In the immunofluorescence analysis BMSC were observed to integrate mostly into the inner retina and expressing GFP. Diabetic group had prominently lower oscillatory potential wave amplitudes than the Baseline group. Three weeks after intravitreal injection Diabetic+BMSC group had significantly better amplitudes than the Diabetic+Sham group. Taken together intravitreal BMSC were thought to improve visual function.
Collapse
Affiliation(s)
- Eren Çerman
- Marmara University School of Medicine, Department of Ophthalmology, Istanbul, Turkey
- * E-mail:
| | - Tolga Akkoç
- Genetic Engineering and Biotechnology Institution, The Scientific and Technological Research Council of Turkey, Kocaeli, Turkey
| | - Muhsin Eraslan
- Marmara University School of Medicine, Department of Ophthalmology, Istanbul, Turkey
| | - Özlem Şahin
- Marmara University School of Medicine, Department of Ophthalmology, Istanbul, Turkey
| | - Selvinaz Özkara
- Haydarpaşa Numune Education and Research Hospital, Department of Pathology, Istanbul, Turkey
| | - Fugen Vardar Aker
- Haydarpaşa Numune Education and Research Hospital, Department of Pathology, Istanbul, Turkey
| | - Cansu Subaşı
- Kocaeli University Center for Stem Cell and Gene Therapies, Kocaeli, Turkey
| | - Erdal Karaöz
- Kocaeli University Center for Stem Cell and Gene Therapies, Kocaeli, Turkey
| | - Tunç Akkoç
- Marmara University School of Medicine, Department of Pediatric Allergy and Immunology, Istanbul, Turkey
| |
Collapse
|
11
|
Usui Y, Westenskow PD, Murinello S, Dorrell MI, Scheppke L, Bucher F, Sakimoto S, Paris LP, Aguilar E, Friedlander M. Angiogenesis and Eye Disease. Annu Rev Vis Sci 2015; 1:155-184. [DOI: 10.1146/annurev-vision-082114-035439] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yoshihiko Usui
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
| | - Peter D. Westenskow
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
- The Lowy Medical Research Institute, La Jolla, California 92037
| | - Salome Murinello
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
| | - Michael I. Dorrell
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
- The Lowy Medical Research Institute, La Jolla, California 92037
- Department of Biology, Point Loma Nazarene University, San Diego, California 92106
| | - Lea Scheppke
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
| | - Felicitas Bucher
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
| | - Susumu Sakimoto
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
| | - Liliana P. Paris
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
| | - Edith Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
| | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037; , , , , , , , , ,
- The Lowy Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
12
|
Gallina D, Zelinka CP, Cebulla CM, Fischer AJ. Activation of glucocorticoid receptors in Müller glia is protective to retinal neurons and suppresses microglial reactivity. Exp Neurol 2015; 273:114-25. [PMID: 26272753 DOI: 10.1016/j.expneurol.2015.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/05/2015] [Accepted: 08/08/2015] [Indexed: 01/06/2023]
Abstract
Reactive microglia and macrophages are prevalent in damaged retinas. Glucocorticoid signaling is known to suppress inflammation and the reactivity of microglia and macrophages. In the vertebrate retina, the glucocorticoid receptor (GCR) is known to be activated and localized to the nuclei of Müller glia (Gallina et al., 2014). Accordingly, we investigated how signaling through GCR influences the survival of neurons using the chick retina in vivo as a model system. We applied intraocular injections of GCR agonist or antagonist, assessed microglial reactivity, and the survival of retinal neurons following different damage paradigms. Microglial reactivity was increased in retinas from eyes that were injected with vehicle, and this reactivity was decreased by GCR-agonist dexamethasone (Dex) and increased by GCR-antagonist RU486. We found that activation of GCR suppresses the reactivity of microglia and inhibited the loss of retinal neurons resulting from excitotoxicity. We provide evidence that the protection-promoting effects of Dex were maintained when the microglia were selectively ablated. Similarly, intraocular injections of Dex protected ganglion cells from colchicine-treatment and protected photoreceptors from damage caused by retinal detachment. We conclude that activation of GCR promotes the survival of ganglion cells in colchicine-damaged retinas, promotes the survival of amacrine and bipolar cells in excitotoxin-damaged retinas, and promotes the survival of photoreceptors in detached retinas. We propose that suppression of microglial reactivity is secondary to activation of GCR in Müller glia, and this mode of signaling is an effective means to lessen the damage and vision loss resulting from different types of retinal damage.
Collapse
Affiliation(s)
- Donika Gallina
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave., Columbus, OH 43210-1239, USA
| | - Christopher Paul Zelinka
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave., Columbus, OH 43210-1239, USA
| | - Colleen M Cebulla
- Havener Eye Institute, Department of Ophthalmology and Visual Science, College of Medicine, The Ohio State University, 915 Olentangy River Road, Suite 5000, Columbus, OH 43212, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, Wexner Medical Center, The Ohio State University, 4190 Graves Hall, 333 West 10th Ave., Columbus, OH 43210-1239, USA.
| |
Collapse
|
13
|
Li SQ, Wan XD, Zhu S, Han HM, Xu ZS, Lu HJ. Establishment of a new animal model of azithromycin-induced liver injury and study the molecular pathological change during the process. Hum Exp Toxicol 2015. [PMID: 26205530 DOI: 10.1177/0960327115595684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The purpose of the present study is to establish a new animal model of azithromycin (AZ)-induced liver injury and study the molecular pathological change during the process. First, mice were respectively injected intraperitoneally with AZ of different high doses. Our results showed that 800 mg/kg AZ injection significantly induced liver injury in the mice, which reflected an ideal process of liver injury and repair. In this study, we analyzed the molecular pathological changes during the process by hematoxylin and eosin staining, immunohistochemistry, Western blot, and quantitative real-time reverse transcription polymerase chain reaction in the liver of mice at 0, 12, 24, 48, and 72 h after 800 mg/kg injection. Our results showed that the expression of heat shock protein 70, proliferating cell nuclear antigen, vascular endothelial growth factor, caspase 3, and cytochrome P450 2E1 were significantly differently expressed during liver injury induced by 800 mg/kg AZ in mice. Our results will be conducive for further study of the pathogenesis and prevention of drug-induced liver injury.
Collapse
Affiliation(s)
- S-Q Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - X-D Wan
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - S Zhu
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, China
| | - H-M Han
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - Z-S Xu
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People's Republic of China
| | - H-J Lu
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People's Republic of China
| |
Collapse
|
14
|
|
15
|
Li SQ, Wang DM, Zhu S, Ma Z, Li RF, Xu ZS, Han HM. The important role of ADAM8 in the progression of hepatocellular carcinoma induced by diethylnitrosamine in mice. Hum Exp Toxicol 2015; 34:1053-72. [DOI: 10.1177/0960327114567767] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study focuses on investigating the concrete role of a disintegrin and metalloproteinase 8 (ADAM8) in the progression of hepatocellular carcinoma (HCC). Mice received anti-ADAM8 monoclonal antibody (mAb) of 100 μg/100 μl, 200 μg/100 μl or 300 μg/100 μl, respectively, in phosphate-buffered saline (PBS) or PBS intervention during the progression of HCC induced by diethylnitrosamine. The survival rate, body weight, and relative liver weight were determined in the mice. Serum aspartate aminotransferase (AST), alanine aminotransferase (ALT) and α-fetoprotein (AFP) level, hematoxylin–eosin staining, the expression level of vascular endothelial growth factor A (VEGF-A), proliferating cell nuclear antigen (PCNA), caspase 3 (Casp3), B cell leukemia 2 (Bcl2), B cell leukemia 2-associated X protein (Bax), protein p53 (P53), and ADAM8 were detected in the mice at the end of the 24th week. Our results showed that anti-ADAM8 mAb intervention effectively improved the survival rate, reduced the body weight loss and increased the relative liver weight in mice in a dose-dependent manner ( p < 0.05 or p < 0.01). Anti-ADAM8 mAb intervention also significantly lowered serum AST, ALT, and AFP levels ( p < 0.05 or p < 0.01), slowed the progression of HCC ( p < 0.05 or p < 0.01), induced the expression of Casp3, Bax, and P53 ( p < 0.05 or p < 0.01), and inhibited the expression of VEGF-A, PCNA, and Bcl2 in the liver of mice ( p < 0.05 or p < 0.01) in a dose-dependent manner compared with the mice receiving PBS intervention. Our study suggested that ADAM8 might promote the progression of HCC by regulating the expression of these factors. Anti-ADAM8 mAb intervention might be suitable as a potential method for HCC therapy.
Collapse
Affiliation(s)
- S-Q Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - D-M Wang
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - S Zhu
- Department of Microbiology Immunology, College of Basic Medical Sciences, Zhengzhou University, People’s Republic of China
| | - Z Ma
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - R-F Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Z-S Xu
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - H-M Han
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| |
Collapse
|
16
|
Li SQ, Zhu S, Wan XD, Xu ZS, Ma Z. Neutralization of ADAM8 ameliorates liver injury and accelerates liver repair in carbon tetrachloride-induced acute liver injury. J Toxicol Sci 2014; 39:339-51. [PMID: 24646716 DOI: 10.2131/jts.39.339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Although some studies have described the function of ADAM8 (a disintegrin and metalloprotease 8) related with rheumatoid arthritis, cancer and asthma, etc., the concrete role of ADAM8 in acute liver injury is still unknown. So mice respectively received anti-ADAM8 monoclonal antibody (mAb) of 100 μg/100 μl, 200 μg/100 μl or 300 μg/100 μl in PBS or PBS pre-injection. Then acute liver injury was induced in the mice by intraperitoneal (i.p.) injection of carbon tetrachloride (CCl₄). Serum AST and ALT level, Haematoxylin-eosin (H&E) staining, the expression level of vascular endothelial growth factor (VEGF), cytochrome P450 1A2 (CYP1A2) and proliferating cell nuclear antigen (PCNA) were detected in the mice after CCl4 administration. Our results showed that anti-ADAM8 mAb pre-injection could effectively lower AST and ALT levels (P < 0.05 or P < 0.01) and reduce liver injury (P < 0.05 or P <0.01), induce the expression of VEGF, CYP1A2 and PCNA (P <0.05 or P < 0.01) in dose-dependent manner compared with the control mice which received PBS pre-injection. In summary, our study suggested that ADAM8 might promote liver injury by inhibiting the proliferation of hepatocytes, angiogenesis and affecting the metabolism function of liver during acute liver injury induced by CCl₄. Anti-ADAM8 mAb injection might be suitable as a potential method for acute liver injury therapy.
Collapse
Affiliation(s)
- San-Qiang Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, China
| | | | | | | | | |
Collapse
|
17
|
Maretzky T, Blobel CP, Guaiquil V. Characterization of oxygen-induced retinopathy in mice carrying an inactivating point mutation in the catalytic site of ADAM15. Invest Ophthalmol Vis Sci 2014; 55:6774-82. [PMID: 25249606 DOI: 10.1167/iovs.14-14472] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Retinal neovascularization is found in diseases such as macular degeneration, diabetic retinopathy, or retinopathy of prematurity and is usually caused by alterations in oxygen supply. We have previously described that mice lacking the membrane-anchored metalloproteinase ADAM15 (a Disintegrin and Metalloprotease 15) have decreased pathological neovascularization of the retina in the oxygen-induced retinopathy (OIR) model. The main purpose of the present study was to determine the contribution of the catalytic activity of ADAM15 to OIR. METHODS To address this question, we generated knock-in mice carrying an inactivating Glutamate to Alanine (E>A) point mutation in the catalytic site of ADAM15 (Adam15E>A mice) and subjected these animals to the OIR model and a heterotopic tumor model. Moreover, we used cell-based assays to determine whether ADAM15 can process cell surface receptors involved in angiogenesis. RESULTS We found that pathological neovascularization in the OIR model in Adam15E>A mice was comparable to that observed in wild type mice, but tumor implantation by heterotopically injected melanoma cells was reduced. In cell-based assays, overexpressed ADAM15 could process the FGFR2iiib, but was unable to process several receptors with roles in angiogenesis. CONCLUSIONS Collectively, these results suggest that the catalytic activity of ADAM15 is not crucial for its function in promoting pathological neovascularization in the mouse OIR model, most likely because of the very limited substrate repertoire of ADAM15. Instead, other noncatalytic functions of ADAM15 must be important for its role in the OIR model.
Collapse
Affiliation(s)
- Thorsten Maretzky
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, United States
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, United States Departments of Medicine and of Physiology, Biophysics and Systems Biology, Weill-Cornell Medical College of Cornell University, New York, New York, United States
| | - Victor Guaiquil
- The Margaret M. Dyson Vision Research Institute, Department of Ophthalmology, Weill-Cornell Medical College of Cornell University, New York, New York, United States
| |
Collapse
|
18
|
Fischer AJ, Zelinka C, Gallina D, Scott MA, Todd L. Reactive microglia and macrophage facilitate the formation of Müller glia-derived retinal progenitors. Glia 2014; 62:1608-28. [PMID: 24916856 DOI: 10.1002/glia.22703] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/11/2014] [Accepted: 05/16/2014] [Indexed: 12/12/2022]
Abstract
In retinas where Müller glia have been stimulated to become progenitor cells, reactive microglia are always present. Thus, we investigated how the activation or ablation of microglia/macrophage influences the formation of Müller glia-derived progenitor cells (MGPCs) in the retina in vivo. Intraocular injections of the Interleukin-6 (IL6) stimulated the reactivity of microglia/macrophage, whereas other types of retinal glia appear largely unaffected. In acutely damaged retinas where all of the retinal microglia/macrophage were ablated, the formation of proliferating MGPCs was greatly diminished. With the microglia ablated in damaged retinas, levels of Notch and related genes were unchanged or increased, whereas levels of ascl1a, TNFα, IL1β, complement component 3 (C3) and C3a receptor were significantly reduced. In the absence of retinal damage, the combination of insulin and Fibroblast growth factor 2 (FGF2) failed to stimulate the formation of MGPCs when the microglia/macrophage were ablated. In addition, intraocular injections of IL6 and FGF2 stimulated the formation of MGPCs in the absence of retinal damage, and this generation of MGPCs was blocked when the microglia/macrophage were absent. We conclude that the activation of microglia and/or infiltrating macrophage contributes to the formation of proliferating MGPCs, and these effects may be mediated by components of the complement system and inflammatory cytokines.
Collapse
Affiliation(s)
- Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio
| | | | | | | | | |
Collapse
|
19
|
Rajashekhar G. Mesenchymal stem cells: new players in retinopathy therapy. Front Endocrinol (Lausanne) 2014; 5:59. [PMID: 24795699 PMCID: PMC4006021 DOI: 10.3389/fendo.2014.00059] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/10/2014] [Indexed: 12/20/2022] Open
Abstract
Retinopathies in human and animal models have shown to occur through loss of pericytes resulting in edema formation, excessive immature retinal angiogenesis, and neuronal apoptosis eventually leading to blindness. In recent years, the concept of regenerating terminally differentiated organs with a cell-based therapy has evolved. The cells used in these approaches are diverse and include tissue-specific endogenous stem cells, endothelial progenitor (EPC), embryonic stem cells, induced pluripotent stem cells (iPSC) and mesenchymal stem cells (MSC). Recently, MSC derived from the stromal fraction of adipose tissue have been shown to possess pluripotent differentiation potential in vitro. These adipose stromal cells (ASC) have been differentiated in a number of laboratories to osteogenic, myogenic, vascular, and adipocytic cell phenotypes. In vivo, ASC have been shown to have functional and phenotypic overlap with pericytes lining microvessels in adipose tissues. Furthermore, these cells either in paracrine mode or physical proximity with endothelial cells, promoted angiogenesis, improved ischemia-reperfusion, protected from myocardial infarction, and were neuroprotective. Owing to the easy isolation procedure and abundant supply, fat-derived ASC are a more preferred source of autologous mesenchymal cells compared to bone marrow MSC. In this review, we present evidence that these readily available ASC from minimally invasive liposuction will facilitate translation of ASC research into patients with retinal diseases in the near future.
Collapse
Affiliation(s)
- Gangaraju Rajashekhar
- Indiana Center for Vascular Biology and Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Vascular and Cardiac Center for Adult Stem Cell Therapy, Indiana University School of Medicine, Indianapolis, IN, USA
- VA Center for Regenerative Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- *Correspondence: Gangaraju Rajashekhar, Eugene and Marilyn Glick Eye Institute, Indiana Center for Vascular Biology and Medicine, Vascular and Cardiac Center for Adult Stem Cell Therapy, 975 West, Walnut Street IB442B, Indianapolis, IN 46202, USA e-mail:
| |
Collapse
|
20
|
Park TS, Bhutto I, Zimmerlin L, Huo JS, Nagaria P, Miller D, Rufaihah AJ, Talbot C, Aguilar J, Grebe R, Merges C, Reijo-Pera R, Feldman RA, Rassool F, Cooke J, Lutty G, Zambidis ET. Vascular progenitors from cord blood-derived induced pluripotent stem cells possess augmented capacity for regenerating ischemic retinal vasculature. Circulation 2013; 129:359-72. [PMID: 24163065 DOI: 10.1161/circulationaha.113.003000] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The generation of vascular progenitors (VPs) from human induced pluripotent stem cells (hiPSCs) has great potential for treating vascular disorders such as ischemic retinopathies. However, long-term in vivo engraftment of hiPSC-derived VPs into the retina has not yet been reported. This goal may be limited by the low differentiation yield, greater senescence, and poor proliferation of hiPSC-derived vascular cells. To evaluate the potential of hiPSCs for treating ischemic retinopathies, we generated VPs from a repertoire of viral-integrated and nonintegrated fibroblast and cord blood (CB)-derived hiPSC lines and tested their capacity for homing and engrafting into murine retina in an ischemia-reperfusion model. METHODS AND RESULTS VPs from human embryonic stem cells and hiPSCs were generated with an optimized vascular differentiation system. Fluorescence-activated cell sorting purification of human embryoid body cells differentially expressing endothelial/pericytic markers identified a CD31(+)CD146(+) VP population with high vascular potency. Episomal CB-induced pluripotent stem cells (iPSCs) generated these VPs with higher efficiencies than fibroblast-iPSC. Moreover, in contrast to fibroblast-iPSC-VPs, CB-iPSC-VPs maintained expression signatures more comparable to human embryonic stem cell VPs, expressed higher levels of immature vascular markers, demonstrated less culture senescence and sensitivity to DNA damage, and possessed fewer transmitted reprogramming errors. Luciferase transgene-marked VPs from human embryonic stem cells, CB-iPSCs, and fibroblast-iPSCs were injected systemically or directly into the vitreous of retinal ischemia-reperfusion-injured adult nonobese diabetic-severe combined immunodeficient mice. Only human embryonic stem cell- and CB-iPSC-derived VPs reliably homed and engrafted into injured retinal capillaries, with incorporation into damaged vessels for up to 45 days. CONCLUSIONS VPs generated from CB-iPSCs possessed augmented capacity to home, integrate into, and repair damaged retinal vasculature.
Collapse
Affiliation(s)
- Tea Soon Park
- Institute for Cell Engineering, and Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD (T.S.P., L.Z., J.S.H., J.A., E.T.Z.); Wilmer Ophthalmological Institute, Johns Hopkins University School of Medicine, Baltimore, MD (I.B., R.G., C.M., G.L.); Department of Radiation Oncology (P.N., F.R.) and Department of Microbiology/Immunology (D.M., R.A.F.), University of Maryland School of Medicine, Baltimore, MD; Department of Cardiovascular Medicine (A.J.R., J.C.) and Institute for Stem Cell Biology and Regenerative Medicine (A.J.R., R.R.-P., J.C.), Stanford University, Palo Alto, CA; and Institute for Basic Biomedical Science at Johns Hopkins School of Medicine, Baltimore, MD (C.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Blenkinsop TA, Corneo B, Temple S, Stern JH. Ophthalmologic stem cell transplantation therapies. Regen Med 2013; 7:32-9. [PMID: 23210809 DOI: 10.2217/rme.12.77] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vision loss is a major social issue, with more than 20 million people over the age of 18 years affected in the USA alone. Loss of vision is feared more than premature death or cardiovascular disease, according to a recent Society for Consumer Research group survey. The annual direct cost of medical care for the most prevalent eye disease, age-related macular degeneration, was estimated at US$255 billion in 2010 with an additional economic impact of US$88 billion due to lost productivity and the burden of family and community care for visual disability. With the blossoming of human stem cell research, regenerative treatments are now being developed that can help reduce this burden. Positive results from animal studies demonstrate that stem cell-based transplants can preserve and potentially improve vision. This has led to new clinical trials for several eye diseases that are yielding encouraging results. In the next few years, additional trials and longer-term results are anticipated to further develop ocular regenerative therapies, with the potential to revolutionize our approach to ophthalmic disease and damage.
Collapse
Affiliation(s)
- Timothy A Blenkinsop
- Neural Stem Cell Institute, Regenerative Research Foundation, One Discovery Drive, Rensselaer, NY12144, USA
| | | | | | | |
Collapse
|
22
|
Avraham-Lubin BCR, Goldenberg-Cohen N, Sadikov T, Askenasy N. VEGF induces neuroglial differentiation in bone marrow-derived stem cells and promotes microglia conversion following mobilization with GM-CSF. Stem Cell Rev Rep 2013; 8:1199-210. [PMID: 22810360 DOI: 10.1007/s12015-012-9396-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE Evaluation of potential tropic effects of vascular endothelial growth factor (VEGF) on the incorporation and differentiation of bone-marrow-derived stem cells (BMSCs) in a murine model of anterior ischemic optic neuropathy (AION). METHODS In the first approach, small-sized subset of BMCs were isolated from GFP donors mice by counterflow centrifugal elutriation and depleted of hematopoietic lineages (Fr25lin(-)). These cells were injected into a peripheral vein (1 × 10(6) in 0.2 ml) or inoculated intravitreally (2 × 10(5)) to syngeneic mice, with or without intravitreal injection of 5 μg/2μL VEGF, simultaneously with AION induction. In a second approach, hematopoietic cells were substituted by myelablative transplant of syngeseic GFP + bone marrow cells. After 3 months, progenitors were mobilized with granulocyte-macrophage colony-stimulating factor (GM-CSF) followed by VEGF inoculation into the vitreous body and AION induction . Engraftment and phenotype were examined by immunohistochemistry and FISH at 4 and 24 weeks post-transplantation, and VEGF receptors were determined by real time PCR. RESULTS VEGF had no quantitative effect on incorporation of elutriated cells in the injured retina, yet it induced early expression of neuroal markers in cells incorporated in the RGC layer and promoted durable gliosis, most prominent perivascular astrocytes. These effects were mediated by VEGF-R1/Flt-1, which is constitutively expresses in the elutriated fraction of stem cells. Mobilization with GM-CSF limited the differentiation of bone marrow progenitors to microglia, which was also fostered by VEGF. CONCLUSIONS VEGF signaling mediated by Flt-1 induces early neural and sustained astrocytic differentiation of stem cells elutriated from adult bone-marrow, with significant contribution to stabilization retinal architecture following ischemic injury.
Collapse
Affiliation(s)
- Bat-Chen R Avraham-Lubin
- The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Tel Aviv University, Petach Tikva, Israel
| | | | | | | |
Collapse
|
23
|
Caballero S, Hazra S, Bhatwadekar A, Li Calzi S, Paradiso LJ, Miller LP, Chang LJ, Kern TS, Grant MB. Circulating mononuclear progenitor cells: differential roles for subpopulations in repair of retinal vascular injury. Invest Ophthalmol Vis Sci 2013; 54:3000-9. [PMID: 23572102 DOI: 10.1167/iovs.12-10280] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE We examined effect on retinal vascular homing of exogenous CD34(+) and CD14(+) progenitor cells using mouse models of chronic (streptozotocin [STZ]-induced diabetes) and acute (ischemia-reperfusion [I/R]) ocular vascular injury. METHODS STZ-treated mice of short or long duration (≤4, ≥11 months) diabetes, along with age- and sex-matched controls, were given intravitreous injections of human CD34(+) and CD14(+) cells isolated from healthy or diabetic donors alone or in combination. I/R injured mice were given diabetic or nondiabetic CD34(+) cells with mesenchymal stem cells (MSCs) or diabetic CD34(+) cells manipulated by ex vivo fucosylation with ASC-101. Injected cells were localized by fluorescent immunocytochemistry, and the degree of retinal vascular colocalization quantified morphometrically. Permeability was assessed by fluorescent albumin leakage. RESULTS Diabetic CD14(+) cells associated with vessels to a greater degree than diabetic CD34(+) cells. Vascular permeability was reduced only by nondiabetic cells and only at the highest number of cells tested. Diabetic CD34(+) cells consistently demonstrated reduced migration. There was a 2-fold or 4-fold increase over control in the specific localization of diabetic CD34(+) cells within the vasculature when these cells were co-administered with MSCs or ex vivo fucosylated prior to injection, respectively. CONCLUSIONS Diabetic CD14(+) cells, unlike diabetic CD34(+) cells, retain robust homing characteristics. CD34(+) or CD14(+) subsets rather than whole bone marrow or peripheral blood cells may prove more beneficial in autologous cell therapy for diabetics. Co-administration with MSCs or ex vivo fucosylation may enhance utility of CD34(+) cells in cell therapy for diabetic ocular conditions like macular ischemia and retinal nonperfusion.
Collapse
Affiliation(s)
- Sergio Caballero
- Program in Stem Cell Biology and Department of Pharmacology & Therapeutics, University of Florida, Gainesville, Florida 32610-0267, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Girdin and Its Phosphorylation Dynamically Regulate Neonatal Vascular Development and Pathological Neovascularization in the Retina. THE AMERICAN JOURNAL OF PATHOLOGY 2013. [DOI: 10.1016/j.ajpath.2012.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
25
|
Liang X, Zhou H, Ding Y, Li J, Yang C, Luo Y, Li S, Sun G, Liao X, Min W. TMP prevents retinal neovascularization and imparts neuroprotection in an oxygen-induced retinopathy model. Invest Ophthalmol Vis Sci 2012; 53:2157-69. [PMID: 22410554 DOI: 10.1167/iovs.11-9315] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE To evaluate the effects of tetramethylpyrazine (TMP) on retinal neovascularization (NV) and neuroprotection in an oxygen-induced retinopathy (OIR) model. METHODS Neonatal C57BL/6J mice were subjected to 75% oxygen from postnatal day 7 (P7) to P12 and then returned to room air. TMP (200 mg/kg) or normal saline was given daily from P12 to P17. Immunostaining, HE staining, TUNEL assay, and RT-PCR were used to assess the effects of TMP on retinal neurovascular repair. RESULTS TMP effectively prevented pathologic NV and accelerated physiologic revascularization by enhancing the formation of endothelial tip cells at the edges of the repairing capillary networks and preserving the astrocytic template in the avascular retina. TMP also prevented morphologic changes and significantly decreased TUNEL-positive cells in the avascular retina by rescuing neurons such as amacrine, rod bipolar, horizontal, and Müller cells. In TMP-treated mice retinas, there was a less obvious loss of amacrine cell bodies and their distinct bands; the number of both rod bipolar and horizontal cell bodies, as well as the density of their dendrites in the outer plexiform layer, was greater than that in OIR control mice. TMP not only decreased the loss of alignment of Müller cell bodies and distortion of processes but reduced the reactive expression of GFAP in Müller cells. Furthermore, HIF-1α and VEGF mRNA expression were downregulated in TMP-treated mice retinas. CONCLUSIONS TMP improved neurovascular recovery by preventing NV and protecting retinal astroglia cells and neurons from ischemia-induced cell death partially due to its downregulation of HIF-1α and VEGF mRNA expression.
Collapse
Affiliation(s)
- Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Benoit E, O'Donnell TF, Patel AN. Safety and efficacy of autologous cell therapy in critical limb ischemia: a systematic review. Cell Transplant 2012; 22:545-62. [PMID: 22490340 DOI: 10.3727/096368912x636777] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Researchers have accumulated a decade of experience with autologous cell therapy in the treatment of critical limb ischemia (CLI). We conducted a systematic review of clinical trials in the literature to determine the safety and efficacy of cell therapy in CLI. We searched the literature for clinical trials of autologous cell therapy in CLI, including observational series of five or more patients to accrue a large pool of patients for safety analysis. Safety analysis included evaluation of death, cancer, unregulated angiogenesis, and procedural adverse events such as bleeding. Efficacy analysis included the clinical endpoints amputation and death as well as functional and surrogate endpoints. We identified 45 clinical trials, including seven RCTs, and 1,272 patients who received cell therapy. The overall adverse event rate was low (4.2%). Cell therapy patients did not have a higher mortality rate than control patients and demonstrated no increase in cancer incidence when analyzed against population rates. With regard to efficacy, cell therapy patients had a significantly lower amputation rate than control patients (OR 0.36, p = 0.0004). Cell therapy also demonstrated efficacy in a variety of functional and surrogate outcomes. Clinical trials differed in the proportion of patients with risk factors for clinical outcomes, and these influenced rates of amputation and death. Cell therapy presents a favorable safety profile with a low adverse event rate and no increase in severe events such as mortality and cancer and treatment with cell therapy decreases the risk of amputation. Cell therapy has a positive benefit-to-risk ratio in CLI and may be a valuable treatment option, particularly for those challenging patients who cannot undergo arterial reconstruction.
Collapse
Affiliation(s)
- Eric Benoit
- Department of Surgery, Tufts Medical Center, Boston, MA, USA
| | | | | |
Collapse
|
27
|
Dou GR, Wang L, Wang YS, Han H. Notch signaling in ocular vasculature development and diseases. Mol Med 2012; 18:47-55. [PMID: 21989947 PMCID: PMC3269647 DOI: 10.2119/molmed.2011.00256] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 10/04/2011] [Indexed: 01/10/2023] Open
Abstract
Ocular angiogenesis, characterized by the formation of new blood vessels in the avascular area in eyes, is a highly coordinated process involved in retinal vasculature formation and several ocular diseases such as age-related macular degeneration, proliferative diabetic retinopathy and retinopathy of prematurity. This process is orchestrated by complicated cellular interactions and vascular growth factors, during which endothelial cells acquire heterogeneous phenotypes and distinct cellular destinations. To date, while the vascular endothelial growth factor has been identified as the most critical angiogenic agent with a remarkable therapeutic value, the Notch signaling pathway appears to be a similarly important regulator in several angiogenic steps. Recent progress has highlighted the involvement, mechanisms and therapeutic potential of Notch signaling in retinal vasculature development and pathological angiogenesis-related eye disorders, which may cause irreversible blindness.
Collapse
Affiliation(s)
- Guo-Rui Dou
- Department of Ophthalmology, Xijing Hospital, Xi’an, China
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| | - Lin Wang
- Department of Hepatic Surgery, Xijing Hospital, Xi’an, China
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| | - Yu-Sheng Wang
- Department of Ophthalmology, Xijing Hospital, Xi’an, China
| | - Hua Han
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
28
|
Yanai R, Thanos A, Connor KM. Complement involvement in neovascular ocular diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 946:161-83. [PMID: 21948368 DOI: 10.1007/978-1-4614-0106-3_10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pathological neovascularization (NV) is a hallmark of late stage neovascular age-related macular degeneration (AMD), diabetic retinopathy (DR), and retinopathy of prematurity (ROP). There is accumulating evidence that alterations in inflammatory and immune system pathways that arise from genetic differences, injury, and disease can predispose individuals to retinal neovascular eye diseases. Yet the mechanism of disease progression with respect to the complement system in these maladies is not fully understood. Recent studies have implicated the complement system as an emerging player in the etiology of several retinal diseases. We will summarize herein several of the complement system pathways known to be involved in ocular neovascular pathologies. Current treatment for many neovascular eye diseases focuses on suppression of NV with laser ablation, photodynamic therapy, or anti-VEGF angiogenic inhibitors. However, these treatments do not address the underlying cause of many of these diseases. A clear understanding of the cellular and molecular mechanisms could bring a major shift in our approach to disease treatment and prevention.
Collapse
Affiliation(s)
- Ryoji Yanai
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA.
| | | | | |
Collapse
|
29
|
Early results and lessons learned from a multicenter, randomized, double-blind trial of bone marrow aspirate concentrate in critical limb ischemia. J Vasc Surg 2011; 54:1650-8. [DOI: 10.1016/j.jvs.2011.06.118] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 06/28/2011] [Accepted: 06/30/2011] [Indexed: 11/23/2022]
|
30
|
Goldenberg-Cohen N, Avraham-Lubin BCR, Sadikov T, Goldstein RS, Askenasy N. Primitive stem cells derived from bone marrow express glial and neuronal markers and support revascularization in injured retina exposed to ischemic and mechanical damage. Stem Cells Dev 2011; 21:1488-500. [PMID: 21905921 DOI: 10.1089/scd.2011.0366] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ischemic or mechanical injury to the optic nerve is an irreversible cause of vision loss, associated with limited regeneration and poor response to neuroprotective agents. The aim of this study was to assess the capacity of adult bone marrow cells to participate in retinal regeneration following the induction of anterior ischemic optic neuropathy (AION) and optic nerve crush (ONC) in a rodent model. The small-sized subset of cells isolated by elutriation and lineage depletion (Fr25lin(-)) was found to be negative for the neuroglial markers nestin and glial fibrillary acidic protein (GFAP). Syngeneic donor cells, identified by genomic marker in sex-mismatched transplants and green fluorescent protein, incorporated into the injured retina (AION and ONC) at a frequency of 0.35%-0.45% after intravenous infusion and 1.8%-2% after intravitreous implantation. Perivascular cells with astrocytic morphology expressing GFAP and vimentin were of the predominant lineage that engrafted after AION injury; 10%-18% of the donor cells incorporated in the retinal ganglion cell layer and expressed NeuN, Thy-1, neurofilament, and beta-tubulin III. The Fr25lin(-) cells displayed an excellent capacity to migrate to sites of tissue disruption and developed coordinated site-specific morphological and phenotypic neural and glial markers. In addition to cellular reconstitution of the injured retinal layers, these cells contributed to endothelial revascularization and apparently supported remodeling by secretion of insulin-like growth factor-1. These results suggest that elutriated autologous adult bone marrow-derived stem cells may serve as an accessible source for cellular reconstitution of the retina following injury.
Collapse
Affiliation(s)
- Nitza Goldenberg-Cohen
- Krieger Eye Research Laboratory, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
| | | | | | | | | |
Collapse
|
31
|
Abstract
Retinal degenerative disease has limited therapeutic options and the possibility of stem cell-mediated regenerative treatments is being actively explored for these blinding retinal conditions. The relative accessibility of this central nervous system tissue and the ability to visually monitor changes after transplantation make the retina and adjacent retinal pigment epithelium prime targets for pioneering stem cell therapeutics. Prior work conducted for several decades indicated the promise of cell transplantation for retinal disease, and new strategies that combine these established surgical approaches with stem cell-derived donor cells is ongoing. A variety of tissue-specific and pluripotent-derived donor cells are being advanced to replace lost or damaged retinal cells and/or to slow the disease processes by providing neuroprotective factors, with the ultimate aim of long-term improvement in visual function. Clinical trials are in the early stages, and data on safety and efficacy are widely anticipated. Positive outcomes from these stem cell-based clinical studies would radically change the way that blinding disorders are approached in the clinic.
Collapse
|
32
|
Huang Y, Enzmann V, Ildstad ST. Stem cell-based therapeutic applications in retinal degenerative diseases. Stem Cell Rev Rep 2011; 7:434-45. [PMID: 20859770 DOI: 10.1007/s12015-010-9192-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Retinal degenerative diseases that target photoreceptors or the adjacent retinal pigment epithelium (RPE) affect millions of people worldwide. Retinal degeneration (RD) is found in many different forms of retinal diseases including retinitis pigmentosa (RP), age-related macular degeneration (AMD), diabetic retinopathy, cataracts, and glaucoma. Effective treatment for retinal degeneration has been widely investigated. Gene-replacement therapy has been shown to improve visual function in inherited retinal disease. However, this treatment was less effective with advanced disease. Stem cell-based therapy is being pursued as a potential alternative approach in the treatment of retinal degenerative diseases. In this review, we will focus on stem cell-based therapies in the pipeline and summarize progress in treatment of retinal degenerative disease.
Collapse
Affiliation(s)
- Yiming Huang
- Institute for Cellular Therapeutics, University of Louisville, 570 S. Preston Street, Suite 404, Louisville, KY 40202-1760, USA
| | | | | |
Collapse
|
33
|
Kielczewski JL, Hu P, Shaw LC, Li Calzi S, Mames RN, Gardiner TA, McFarland E, Chan-Ling T, Grant MB. Novel protective properties of IGFBP-3 result in enhanced pericyte ensheathment, reduced microglial activation, increased microglial apoptosis, and neuronal protection after ischemic retinal injury. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1517-28. [PMID: 21435441 DOI: 10.1016/j.ajpath.2010.12.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/14/2010] [Accepted: 12/22/2010] [Indexed: 12/25/2022]
Abstract
This study was conducted to determine the perivascular cell responses to increased endothelial cell expression of insulin-like growth factor binding protein-3 (IGFBP-3) in mouse retina. The contribution of bone marrow cells in the IGFBP-3-mediated response was examined using green fluorescent protein-positive (GFP(+)) adult chimeric mice subjected to laser-induced retinal vessel occlusion injury. Intravitreal injection of an endothelial-specific IGFBP-3-expressing plasmid resulted in increased differentiation of GFP(+) hematopoietic stem cells (HSCs) into pericytes and astrocytes as determined by immunohistochemical analysis. Administration of IGFBP-3 plasmid to mouse pups that underwent the oxygen-induced retinopathy model resulted in increased pericyte ensheathment and reduced pericyte apoptosis in the developing retina. Increased IGFBP-3 expression reduced the number of activated microglial cells and decreased apoptosis of neuronal cells in the oxygen-induced retinopathy model. In summary, IGFBP-3 increased differentiation of GFP(+) HSCs into pericytes and astrocytes while increasing vascular ensheathment of pericytes and decreasing apoptosis of pericytes and retinal neurons. All of these cytoprotective effects exhibited by IGFBP-3 overexpression can result in a more stable retinal vascular bed. Thus, endothelial expression of IGFBP-3 may represent a physiologic response to injury and may represent a therapeutic strategy for the treatment of ischemic vascular eye diseases, such as diabetic retinopathy and retinopathy of prematurity.
Collapse
Affiliation(s)
- Jennifer L Kielczewski
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610-0267, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Evidence of hematopoietic differentiation, vasculogenesis and angiogenesis in the formation of human choroidal blood vessels. Exp Eye Res 2011; 92:361-76. [PMID: 21354137 DOI: 10.1016/j.exer.2011.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 12/01/2010] [Accepted: 02/15/2011] [Indexed: 11/20/2022]
Abstract
Human fetal eyes 8-40 weeks gestation (WG) were examined using markers to hematopoietic stem cells (HSC), vascular precursor cells (VPC), monocytes/macrophages and endothelial cells (EC). Electron microscopy and bromo-deoxyuridene labeling were undertaken to confirm the existence of solid vascular cords and to demonstrate vasculogenesis and angiogenesis in developing choroidal tissue. Our results demonstrated that the earliest incipient choroid consisted of vimentin(+) mesenchymal precursor cells which downregulated vimentin expression with maturation. Our observations lead us to conclude that these vimentin(-)/CD34(+)/CD44(+)/CD133(+) HSCs then differentiated into three distinct lineages: single isolated CD34(-)/CD39(+) VPCs that formed solid vascular cords which lumenized and became lined with CD34(+) vascular ECs; CD34(--+)/CD14(+)/CD68(+) monocytes that differentiated into tissue macrophages; and CD133(+)/CD34(--+)/α-smooth muscle actin(+) mural precursor cells that matured into smooth muscle cells and pericytes. Blood vessel formation occurred throughout the whole choroid simultaneously, indicative of in situ differentiation. Vasculogenesis, as evidenced by lumenization of solid vascular cords, was responsible for the formation of the entire choroidal area with angiogenesis, in all three layers of the choroid, only adding to vascular density. These results suggest that formation of the human choroid involves three processes: HSC differentiation, vasculogenesis and angiogenesis. Since vasculogenesis takes place independently of VEGF(165), further insights regarding the molecular mechanisms of vasculogenesis are required to better inform future treatments of choroidal neovascularization.
Collapse
|
35
|
Ye XZ, Yu SC, Bian XW. Contribution of myeloid-derived suppressor cells to tumor-induced immune suppression, angiogenesis, invasion and metastasis. J Genet Genomics 2011; 37:423-30. [PMID: 20659706 DOI: 10.1016/s1673-8527(09)60061-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/14/2010] [Accepted: 05/20/2010] [Indexed: 02/03/2023]
Abstract
Growing evidence suggests that myeloid-derived suppressor cells (MDSCs), which have been named "immature myeloid cells" or "myeloid suppressor cells" (MSCs), play a critical role during the progression of cancer in tumor-bearing mice and cancer patients. As their name implies, these cells are derived from bone marrow and have a tremendous potential to suppress immune responses. Recent studies indicated that these cells also have a crucial role in tumor progression. MDSCs can directly incorporate into tumor endothelium. They secret many pro-angiogenic factors as well. In addition, they play an essential role in cancer invasion and metastasis through inducing the production of matrix metalloproteinases (MMPs), chemoattractants and creating a pre-metastatic environment. Increasing evidence supports the idea that cancer stem cells (CSCs) are responsible for tumorigenesis, resistance to therapies, invasion and metastasis. Here, we hypothesize that CSCs may "hijack" MDSCs for use as alternative niche cells, leading to the maintenance of stemness and enhanced chemo- and radio-therapy resistance. The countermeasure that directly targets to MDSCs may be useful for against angiogenesis and preventing cancer from invasion and metastasis. Therefore, the study of MDSCs is important to understand tumor progression and to enhance the therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Xian-Zong Ye
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | |
Collapse
|
36
|
Hasan A, Pokeza N, Shaw L, Lee HS, Lazzaro D, Chintala H, Rosenbaum D, Grant MB, Chaqour B. The matricellular protein cysteine-rich protein 61 (CCN1/Cyr61) enhances physiological adaptation of retinal vessels and reduces pathological neovascularization associated with ischemic retinopathy. J Biol Chem 2011; 286:9542-54. [PMID: 21212276 DOI: 10.1074/jbc.m110.198689] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Retinal vascular damages are the cardinal hallmarks of retinopathy of prematurity (ROP), a leading cause of vision impairment and blindness in childhood. Both angiogenesis and vasculogenesis are disrupted in the hyperoxia-induced vaso-obliteration phase, and recapitulated, although aberrantly, in the subsequent ischemia-induced neovessel formation phase of ROP. Yet, whereas the histopathological features of ROP are well characterized, many key modulators with a therapeutic potential remain unknown. The CCN1 protein also known as cysteine-rich protein 61 (Cyr61) is a dynamically expressed, matricellular protein required for proper angiogenesis and vasculogenesis during development. The expression of CCN1 becomes abnormally reduced during the hyperoxic and ischemic phases of ROP modeled in the mouse eye with oxygen-induced retinopathy (OIR). Lentivirus-mediated re-expression of CCN1 enhanced physiological adaptation of the retinal vasculature to hyperoxia and reduced pathological angiogenesis following ischemia. Remarkably, injection into the vitreous of OIR mice of hematopoietic stem cells (HSCs) engineered to express CCN1 harnessed ischemia-induced neovessel outgrowth without adversely affecting the physiological adaptation of retinal vessels to hyperoxia. In vitro exposure of HSCs to recombinant CCN1 induced integrin-dependent cell adhesion, migration, and expression of specific endothelial cell markers as well as many components of the Wnt signaling pathway including Wnt ligands, their receptors, inhibitors, and downstream targets. CCN1-induced Wnt signaling mediated, at least in part, adhesion and endothelial differentiation of cultured HSCs, and inhibition of Wnt signaling interfered with normalization of the retinal vasculature induced by CCN1-primed HSCs in OIR mice. These newly identified functions of CCN1 suggest its possible therapeutic utility in ischemic retinopathy.
Collapse
Affiliation(s)
- Adeel Hasan
- Department of Cell Biology, Downstate Medical Center, Brooklyn, New York 11203, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fischer AJ, Zelinka C, Scott MA. Heterogeneity of glia in the retina and optic nerve of birds and mammals. PLoS One 2010; 5:e10774. [PMID: 20567503 PMCID: PMC2887354 DOI: 10.1371/journal.pone.0010774] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 05/04/2010] [Indexed: 11/21/2022] Open
Abstract
We have recently described a novel type of glial cell that is scattered across the inner layers of the avian retina [1]. These cells are stimulated by insulin-like growth factor 1 (IGF1) to proliferate, migrate distally into the retina, and up-regulate the nestin-related intermediate filament transitin. These changes in glial activity correspond with increased susceptibility of neurons to excitotoxic damage. This novel cell-type has been termed the Non-astrocytic Inner Retinal Glia-like (NIRG) cells. The purpose of the study was to investigate whether the retinas of non-avian species contain cells that resemble NIRG cells. We assayed for NIRG cells by probing for the expression of Sox2, Sox9, Nkx2.2, vimentin and nestin. NIRG cells were distinguished from astrocytes by a lack of expression for Glial Fibrilliary Acidic Protein (GFAP). We examined the retinas of adult mice, guinea pigs, dogs and monkeys (Macaca fasicularis). In the mouse retina and optic nerve head, we identified numerous astrocytes that expressed GFAP, S100β, Sox2 and Sox9; however, we found no evidence for NIRG-like cells that were positive for Nkx2.2, nestin, and negative for GFAP. In the guinea pig retina, we did not find astrocytes or NIRG cells in the retina, whereas we identified astrocytes in the optic nerve. In the eyes of dogs and monkeys, we found astrocytes and NIRG-like cells scattered across inner layers of the retina and within the optic nerve. We conclude that NIRG-like cells are present in the retinas of canines and non-human primates, whereas the retinas of mice and guinea pigs do not contain NIRG cells.
Collapse
Affiliation(s)
- Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA.
| | | | | |
Collapse
|
38
|
Stahl A, Connor KM, Sapieha P, Chen J, Dennison RJ, Krah NM, Seaward MR, Willett KL, Aderman CM, Guerin KI, Hua J, Löfqvist C, Hellström A, Smith LEH. The mouse retina as an angiogenesis model. Invest Ophthalmol Vis Sci 2010; 51:2813-26. [PMID: 20484600 DOI: 10.1167/iovs.10-5176] [Citation(s) in RCA: 474] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The mouse retina has been used extensively over the past decades to study both physiologic and pathologic angiogenesis. Over time, various mouse retina models have evolved into well-characterized and robust tools for in vivo angiogenesis research. This article is a review of the angiogenic development of the mouse retina and a discussion of some of the most widely used vascular disease models. From the multitude of studies performed in the mouse retina, a selection of representative works is discussed in more detail regarding their role in advancing the understanding of both the ocular and general mechanisms of angiogenesis.
Collapse
Affiliation(s)
- Andreas Stahl
- Department of Ophthalmology, Harvard Medical School, Children's Hospital Boston, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Fischer AJ, Scott MA, Zelinka C, Sherwood P. A novel type of glial cell in the retina is stimulated by insulin-like growth factor 1 and may exacerbate damage to neurons and Müller glia. Glia 2010; 58:633-49. [PMID: 19941335 DOI: 10.1002/glia.20950] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent studies have demonstrated that insulin can have profound affects on the survival of neurons within the retina. The purpose of this study was to determine how insulin-like growth factor 1 (IGF1) influences retinal cells; in particular, the glial cells. We identify a novel type of glial cell in the avian retina and provide evidence that these cells can respond to acute damage and IGF1. In normal retinas, we found a distinct cell-type, scattered across the ganglion cell and inner plexiform layers that express Sox2, Sox9, Nkx2.2, vimentin, and transitin, the avian homologue of mammalian nestin. These glial cells have a unique immunohistochemical profile, morphology, and distribution that are distinct among other known types of retinal glia, including microglia, oligodendrocytes, astrocytes, and Muller glia. We termed these cells nonastrocytic inner retinal glia-like (NIRG) cells. We found that the NIRG cells may express the IGF1 receptor and respond to IGF1 by proliferating, migrating distally into the retina, and upregulating transitin. In addition, IGF1 stimulated microglia to become reactive and upregulate lysosomal membrane glycoprotein and CD45. With microglia and NIRG cells stimulated by IGF1 there were elevated levels of cell death and numerous focal detachments across the retina in response to excitotoxic damage. Cell death was prominent within the areas of detachment coinciding with a stark loss of Müller glia and accumulation of NIRG cells. We conclude that NIRG cells are a novel type of retinal glia that is sensitive to IGF1 and whose activity may impact the survival of neurons and Müller glia.
Collapse
Affiliation(s)
- Andy J Fischer
- Department of Neuroscience, The Ohio State University, College of Medicine, Columbus, OH 43210-1239, USA.
| | | | | | | |
Collapse
|
40
|
Fadini GP, Avogaro A. Cell-based methods for ex vivo evaluation of human endothelial biology. Cardiovasc Res 2010; 87:12-21. [DOI: 10.1093/cvr/cvq119] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
41
|
Shimada N, Ohno-Matsui K, Iseki S, Koike M, Uchiyama Y, Wang J, Yoshida T, Sato T, Peters C, Mochizuki M, Morita I. Cathepsin L in bone marrow-derived cells is required for retinal and choroidal neovascularization. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2571-80. [PMID: 20304958 DOI: 10.2353/ajpath.2010.091027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Many vision-threatening diseases are characterized by intraocular neovascularization, (e.g., proliferative diabetic retinopathy and age-related macular degeneration). Although a new therapy with anti-VEGF antibodies is being used to treat these intraocular neovascular disorders, the visual recovery is limited, mainly because of the remnants of fibrovascular tissues. The ideal goal of the treatment is to prevent the invasion of new vessels into the avascular tissue through a matrix barrier. The purpose of this study was to determine the role played by cathepsin L, a matrix degrading enzyme, on intraocular angiogenesis. Used established animal models of retinal and choroidal neovascularization, we demonstrated that an inhibition of cathepsin L by specific inhibitors resulted in a significant decrease of intraocular neovascularization. A similar decrease of neovascularization was found in cathepsin L-deficient mice. Transplantation of bone marrow from cathepsin L-deficient mice into wild-type mice significantly reduced the degree of intraocular neovascularization. In addition, immunocytochemical analyses demonstrated that VE cadherin-positive endothelial progenitor cells, but not CD43-positive or Iba-1-positive cells, were the major cells contributing to the production of cathepsin L. These data indicate that cathepsin L expressed in endothelial progenitor cells plays a critical role in intraocular angiogenesis and suggest a potential therapeutic approach of targeting cathepsin L for neovascular ocular diseases.
Collapse
Affiliation(s)
- Noriaki Shimada
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
ADAM8 is a negative regulator of retinal neovascularization and of the growth of heterotopically injected tumor cells in mice. J Mol Med (Berl) 2010; 88:497-505. [PMID: 20119708 DOI: 10.1007/s00109-010-0591-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 12/11/2009] [Accepted: 12/25/2009] [Indexed: 01/20/2023]
Abstract
ADAM8 is a member of the "a disintegrin and metalloproteinase" (ADAM) family of membrane-anchored metalloproteinases. ADAM8-deficient mice have no evident spontaneous developmental or pathological defects, and little is currently known about the role of ADAM8 in disease. Here, we investigated the contribution of ADAM8 to pathological neovascularization in mice using an oxygen-induced retinopathy (OIR) model and heterotopical injection of tumor cells. We found an increase in retinal re-vascularization but fewer neovascular tufts in the OIR model and increased growth of heterotopically injected tumor cells in Adam8-/- mice compared with wild-type controls. These results suggest that ADAM8 functions to limit both of these processes in wild-type mice. In cell-based assays, overexpression of ADAM8 increased the ectodomain shedding of several co-expressed membrane proteins with roles in angiogenesis (CD31, Tie-2, Flk-1, Flt-1, EphrinB2, EphB4, VE-cadherin, KL-1, E-selectin, and neuregulin-1beta2). Thus, dysregulated expression of ADAM8 in endothelial cells in vivo could potentially increase the processing of these and other substrate proteins. Taken together, our findings suggest that inhibiting ADAM8 could be useful for promoting re-vascularization and thereby preventing formation of neovascular tufts in proliferative retinopathies. On the other hand, blocking ADAM8 could be detrimental in the context of rapidly growing tumors.
Collapse
|
43
|
Dorrell MI, Aguilar E, Jacobson R, Trauger SA, Friedlander J, Siuzdak G, Friedlander M. Maintaining retinal astrocytes normalizes revascularization and prevents vascular pathology associated with oxygen-induced retinopathy. Glia 2010; 58:43-54. [PMID: 19544395 DOI: 10.1002/glia.20900] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Astrocytes are well known modulators of normal developmental retinal vascularization. However, relatively little is known about the role of glial cells during pathological retinal neovascularization (NV), a leading contributor to vision loss in industrialized nations. We demonstrate that the loss of astrocytes and microglia directly correlates with the development of pathological NV in a mouse model of oxygen-induced retinopathy (OIR). These two distinct glial cell populations were found to have cooperative survival effects in vitro and in vivo. The intravitreal injection of myeloid progenitor cells, astrocytes, or astrocyte-conditioned media rescued endogenous astrocytes from degeneration that normally occurs within the hypoxic, vaso-obliterated retina following return to normoxia. Protection of the retinal astrocytes and microglia was directly correlated with accelerated revascularization of the normal retinal plexuses and reduction of pathological intravitreal NV normally associated with OIR. Using astrocyte-conditioned media, several factors were identified that may contribute to the observed astrocytic protection and subsequent normalization of the retinal vasculature, including vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Injection of VEGF or bFGF at specific doses rescued the retinas from developing OIR-associated pathology, an effect that was also preceded by protection of endogenous glia from hypoxia-induced degeneration. Together, these data suggest that vascular-associated glia are also required for normalized revascularization of the hypoxic retina. Methods developed to target and protect glial cells may provide a novel strategy by which normalized revascularization can be promoted and the consequences of abnormal NV in retinal vascular diseases can be prevented.
Collapse
Affiliation(s)
- Michael I Dorrell
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Fernández MN. Improving the outcome of cord blood transplantation: use of mobilized HSC and other cells from third party donors. Br J Haematol 2009; 147:161-76. [DOI: 10.1111/j.1365-2141.2009.07766.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Basu A, Menicucci G, Maestas J, Das A, McGuire P. Plasminogen activator inhibitor-1 (PAI-1) facilitates retinal angiogenesis in a model of oxygen-induced retinopathy. Invest Ophthalmol Vis Sci 2009; 50:4974-81. [PMID: 19443721 PMCID: PMC3684946 DOI: 10.1167/iovs.09-3619] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Angiogenesis, or the formation of new retinal blood vessels, is a key feature of many proliferative retinal diseases including diabetic retinopathy, retinal vein occlusion, and retinopathy of prematurity. The aim of the present study was to investigate the role of the serine proteinase inhibitor plasminogen activator inhibitor -1 (PAI-1) in facilitating retinal angiogenesis. METHODS The temporal expression of PAI-1 was examined by real-time PCR, Western blot analysis, and immunohistochemistry in retinal tissues from mice with oxygen-induced retinopathy. The requirement for PAI-1 in facilitating the retinal angiogenic response in this model was examined by quantitating the angiogenic response with wild-type and PAI-1 null mice. The mechanism by which PAI-1 mediates angiogenesis was further investigated with isolated human retinal vascular endothelial cells. RESULTS PAI-1 expression was upregulated in the retinas of mice with oxygen-induced retinopathy, which coincided with a significant increase in the expression of vitronectin in the retina of the experimental mice. There was significant reduction in the angiogenic response of PAI-1(-/-) mice compared with wild-type mice. PAI-1 promotes endothelial cell migration in vitro and facilitates the migration of cells on a vitronectin substrate by regulating alpha v integrin cell surface expression. CONCLUSIONS These observations suggest a role for PAI-1 during retinal angiogenesis and point to a potential new therapeutic target in the prevention or treatment of retinal neovascularization seen in many ocular diseases.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Blotting, Western
- Cell Movement
- Cells, Cultured
- Disease Models, Animal
- Endothelium, Vascular
- Flow Cytometry
- Fluorescent Antibody Technique, Indirect
- Gene Expression/physiology
- Humans
- Infant, Newborn
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oxygen/toxicity
- RNA, Messenger/metabolism
- Retinal Neovascularization/etiology
- Retinal Neovascularization/metabolism
- Retinal Neovascularization/pathology
- Retinal Vessels/metabolism
- Retinal Vessels/pathology
- Reverse Transcriptase Polymerase Chain Reaction
- Serpin E2
- Serpins/physiology
- Up-Regulation
- Vitronectin/metabolism
Collapse
Affiliation(s)
- Anupam Basu
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| | | | | | | | | |
Collapse
|
46
|
Bhola R, Purkiss T, Hunter S, Stewart D, Rychwalski PJ. Effect of granulocyte colony-stimulating factor on the incidence of threshold retinopathy of prematurity. J AAPOS 2009; 13:450-3. [PMID: 19840722 DOI: 10.1016/j.jaapos.2009.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 07/16/2009] [Accepted: 07/17/2009] [Indexed: 12/16/2022]
Abstract
PURPOSE To study the effect of granulocyte colony-stimulating factor (GCSF) on the incidence of threshold retinopathy of prematurity (ROP). METHODS A retrospective chart review of all neonates who received GCSF at our neonatal intensive care unit over a period ranging from January 2003 to September 2007 was performed. Of the 213 patients identified, 50 patients with birth weight <1,500 g and gestational age <32 weeks were included in this study. The incidence of threshold ROP in this subset was compared to a control group (n = 161) obtained from Vermont Oxford Network ROP database over the same time period. The two data sets were cross-referenced to exclude any patient receiving GCSF from the control data. RESULTS The average birth weight was 913 g in GCSF patients and 847 g in controls. The average dose of GCSF was 10.4 microg (range, 1.2-21.6 microg). The average age at administration of GCSF was 12 days after birth (range, 0-34 days). Out of 50 patients in the GCSF group, 5 (10%) required laser treatment, whereas in the contol group, 30 (18.6 %) required laser treatment; this difference was not statistically significant. CONCLUSIONS There was a suggestion of a decreased incidence of threshold ROP, leading to reduced need for laser treatment, in patients who received GCSF, but the observed differences were not statistically significant. The authors anticipate that these findings will lead to further study of the upstream regulators of neovascularization.
Collapse
Affiliation(s)
- Rahul Bhola
- Department of Ophthalmology, University of Louisville School of Medicine, Kentucky, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
Pathological ocular neovascularization, caused by diabetic retinopathy, age-related macular degeneration, or retinopathy of prematurity, is a leading cause of blindness, yet much remains to be learned about its underlying causes. Here we used oxygen-induced retinopathy (OIR) and laser-induced choroidal neovascularization (CNV) to assess the contribution of the metalloprotease-disintegrin ADAM9 to ocular neovascularization in mice. Pathological neovascularization in both the OIR and CNV models was significantly reduced in Adam9(-/-) mice compared to wild-type controls. In addition, the level of ADAM9 expression was strongly increased in endothelial cells in pathological vascular tufts in the OIR model. Moreover, tumor growth from heterotopically injected B16F0 melanoma cells was reduced in Adam9(-/-) mice compared to controls. In cell-based assays, the overexpression of ADAM9 enhanced the ectodomain shedding of EphB4, Tie-2, Flk-1, CD40, VCAM, and VE-cadherin, so the enhanced expression of ADAM9 could potentially affect pathological neovascularization by increasing the shedding of these and other membrane proteins from endothelial cells. Finally, we provide the first evidence for the upregulation of ADAM9-dependent shedding by reactive oxygen species, which in turn are known to play a critical role in OIR. Collectively, these results suggest that ADAM9 could be an attractive target for the prevention of proliferative retinopathies, CNV, and cancer.
Collapse
|
48
|
Afzal A, Shaw LC, Ljubimov AV, Boulton ME, Segal MS, Grant MB. Retinal and choroidal microangiopathies: therapeutic opportunities. Microvasc Res 2007; 74:131-44. [PMID: 17585951 DOI: 10.1016/j.mvr.2007.04.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 04/25/2007] [Accepted: 04/25/2007] [Indexed: 12/12/2022]
Abstract
Pathological angiogenesis in the retina and underlying choroid is a major cause of visual impairment in all age groups. The last decade has seen an explosion in the clinical availability of antiangiogenic compounds. Emphasis has been placed on inhibitors of the VEGF signaling pathway and considerable success has been achieved with aptamers and antibodies that bind VEGF. However, regression of neovascularization is rarely permanent and the regrowth of new vessels, often within a few months, requires multiple applications of drug. A number of antiangiogenic factors such as IGFBP3, SDF-1 blockers, PEDF, gamma-secretase, Delta-like ligand 4, and integrin antagonists have been identified, which act either indirectly on the VEGF system or independent of it. The importance of other candidates such as HIF-1alpha and protein kinase CK2, which act as "master" regulators of angiogenesis, offer realistic alternative targets for pharmacological intervention. The concept of combination therapy is rapidly gaining interest in the eye field and co-administration of two angiogenic agents (e.g., a CK2 inhibitor with a somatostatin analog, octreotide) are often significantly more effective at inhibiting retinal angiogenesis than either drug alone. The following review will discuss the current therapies available for aberrant ocular angiogenesis, consider new candidate targets for development of antiangiogenic compounds and emphasize the importance of combinatorial pharmacological agents in the treatment of such a dynamic cellular event as angiogenesis.
Collapse
Affiliation(s)
- A Afzal
- Program in Stem Cell Biology, Department of Pharmacology and Therapeutics, University of Florida, PO Box 100267, Gainesville, FL 32610-0267, USA
| | | | | | | | | | | |
Collapse
|