1
|
Sadikan MZ, Abdul Nasir NA, Lambuk L, Mohamud R, Reshidan NH, Low E, Singar SA, Mohmad Sabere AS, Iezhitsa I, Agarwal R. Diabetic retinopathy: a comprehensive update on in vivo, in vitro and ex vivo experimental models. BMC Ophthalmol 2023; 23:421. [PMID: 37858128 PMCID: PMC10588156 DOI: 10.1186/s12886-023-03155-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023] Open
Abstract
Diabetic retinopathy (DR), one of the leading causes of visual impairment and blindness worldwide, is one of the major microvascular complications in diabetes mellitus (DM). Globally, DR prevalence among DM patients is 25%, and 6% have vision-threatening problems among them. With the higher incidence of DM globally, more DR cases are expected to be seen in the future. In order to comprehend the pathophysiological mechanism of DR in humans and discover potential novel substances for the treatment of DR, investigations are typically conducted using various experimental models. Among the experimental models, in vivo models have contributed significantly to understanding DR pathogenesis. There are several types of in vivo models for DR research, which include chemical-induced, surgical-induced, diet-induced, and genetic models. Similarly, for the in vitro models, there are several cell types that are utilised in DR research, such as retinal endothelial cells, Müller cells, and glial cells. With the advancement of DR research, it is essential to have a comprehensive update on the various experimental models utilised to mimic DR environment. This review provides the update on the in vitro, in vivo, and ex vivo models used in DR research, focusing on their features, advantages, and limitations.
Collapse
Affiliation(s)
- Muhammad Zulfiqah Sadikan
- Department of Pharmacology, Faculty of Medicine, Manipal University College Malaysia (MUCM), Bukit Baru, 75150, Melaka, Malaysia
| | - Nurul Alimah Abdul Nasir
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, 47000, Sungai Buloh, Selangor, Malaysia.
| | - Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nur Hidayah Reshidan
- School of Biology, Faculty of Applied Sciences, Universiti Teknologi MARA, 40450, Shah Alam, Selangor, Malaysia
| | - Evon Low
- Ageing Biology Centre, Newcastle University, NE1 7RU, Newcastle upon Tyne, UK
| | - Saiful Anuar Singar
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, 32306, Tallahassee, FL, USA
| | - Awis Sukarni Mohmad Sabere
- Kulliyyah of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Bandar Indera Mahkota, 25200, Kuantan, Pahang, Malaysia
| | - Igor Iezhitsa
- School of Medicine, International Medical University, 57000, Bukit Jalil, Kuala Lumpur, Malaysia
- Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Pavshikh Bortsov sq. 1, 400131 , Volgograd, Russian Federation
| | - Renu Agarwal
- School of Medicine, International Medical University, 57000, Bukit Jalil, Kuala Lumpur, Malaysia
| |
Collapse
|
2
|
Tun SBB, Barathi VA. Akimba Proliferative Diabetic Retinopathy Model: Understanding Molecular Mechanism and Drug Screening for the Progression of Diabetic Retinopathy. Methods Mol Biol 2023; 2678:13-26. [PMID: 37326702 DOI: 10.1007/978-1-0716-3255-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
As the prevalence of diabetes has reached epidemic proportions worldwide, diabetic retinopathy incidence is increasing rapidly. An advanced diabetic retinopathy (DR) stage can lead to a sight-threatening form. There is growing evidence showing diabetes causes a range of metabolic changes that subsequently lead to pathological modifications in the retina and retinal blood vessels. To understand the complex mechanism of the pathophysiology of DR, a precise model is not readily available. By crossbreeding the Akita and Kimba strains, a suitable proliferative DR model was acquired. This new Akimba strain manifests marked hyperglycemia and vascular changes, which resemble the early and advanced stage of DR.Here, we describe the breeding method, colony screening for experiments, and imaging techniques widely used to investigate the DR progression in this model. We elaborate step-by-step protocols to set up and perform fundus, fluorescein angiography, optical coherence tomography, and optical coherence tomography-angiogram to study retinal structural changes and vascular abnormalities. In addition, we show a method to label the leukocytes with fluorescence and laser speckle flowgraphy to examine the inflammation in the retina and retinal vessel blood flow speed, respectively. Lastly, we describe electroretinogram to evaluate the functional aspect of the DR transformations.
Collapse
Affiliation(s)
- Sai Bo Bo Tun
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, Singapore, Singapore
- Karolinska Institutet, Stockholm, Sweden
| | - Veluchamy Amutha Barathi
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, Singapore, Singapore.
- ACP in Ophthalmology & Visual Sciences, DUKE-NUS Graduate Medical School, Singapore, Singapore.
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Guidolin D, Tortorella C, Ribatti D. Detection of Possible Symmetries in Vascular Networks by Computer-Assisted Image Analysis. Methods Mol Biol 2023; 2572:167-180. [PMID: 36161416 DOI: 10.1007/978-1-0716-2703-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The possible forms that vascular networks may assume are significantly constrained by complex demands in terms of efficient delivery of oxygen and resources throughout the entire body. Because of these constraints the search for systematic patterns in the structural features of vascular networks and of their correlation with physiological needs and pathological conditions (such as tumor angiogenesis) represents an important line of morphological research. In this context, symmetry properties of vascular trees received limited attention, although symmetry is a widespread phenomenon, visible in all forms and scales in natural environments, and represents a significant information to describe a shape. In the present chapter three, image analysis-based methods allowing for the detection of possible symmetry features exhibited by vascular trees will be detailed and discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova Medical School, Padova, Italy.
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova Medical School, Padova, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
4
|
Pirri C, Petrelli L, Fede C, Guidolin D, Tiengo C, De Caro R, Stecco C. Blood supply to the superficial fascia of the abdomen: An anatomical study. Clin Anat 2022; 36:570-580. [PMID: 36576229 DOI: 10.1002/ca.23993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/21/2022] [Accepted: 12/11/2022] [Indexed: 12/29/2022]
Abstract
The aim of this study was to examine data demonstrating that Scarpa's fascia, a superficial fascia of the anterior abdominal wall, is a vascularized tissue. Specimens of the fascia of seven volunteers undergoing abdominoplasty surgical procedures at the Plastic Surgery Unit of the University of Padova Medical Center were collected. Fractal analysis and quantitative assessment of the vascular network of the fascia was carried out, exploiting the presence of blood in the vessels. Each sample was divided and processed for histological/immunohistochemical analysis (into 5 micron-paraffin embedded sections and cryo-sectioned free-floating samples) as well as for electron microscopy study. A rich vascular pattern forming a fine, dense meshwork with an area percentage of 6.20% ± 2.10% von Willebrand factor stained vessels was noted in all the specimens of the fascia examined; the area percentage of the αSMA-stained vessels was 2.93% ± 1.80%. The diameters of the vessels fell between the 13 and 65 μm range; the network was composed of arteries, veins, capillaries and lymphatic segments. Topological results showed that the vascular network within Scarpa's fascia is well branched (segments: 6615 ± 3070 and 8.40 ± 3.40 per mm2 ; crossing points: 3092 ± 1490 and 3.40 ± 1.90 per mm2 ). Fractal analysis (fractal dimension = 1.063 ± 0.10; lacunarity = 0.60 ± 0.10) revealed that this particular vascular network has an optimal spatial distribution and homogeneity occupying the entire space of the superficial fascia. These findings could undoubtedly be useful to plastic surgeons as well as to pain management specialists.
Collapse
Affiliation(s)
- Carmelo Pirri
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Lucia Petrelli
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Caterina Fede
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Diego Guidolin
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Cesare Tiengo
- Department of Neurosciences, The Plastic Surgery Unit of the University of Padova Medical Center, Padova, Italy
| | - Raffaele De Caro
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| | - Carla Stecco
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padova, Italy
| |
Collapse
|
5
|
Determining the role of SGLT2 inhibition with Empagliflozin in the development of diabetic retinopathy. Biosci Rep 2022; 42:230970. [PMID: 35234250 PMCID: PMC8891593 DOI: 10.1042/bsr20212209] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus is a chronic metabolic disease that occurs when the pancreas is not producing enough insulin or when the insulin that it does produce is not able to be used effectively in the body. This results in hyperglycemia and if the blood sugars are not controlled, then it can lead to serious damage of various body systems, especially the nerves and the blood vessels. Uncontrolled diabetes is a major cause of kidney failure, heart attacks, stroke and amputation. One of the most devastating complications for patients is diabetic retinopathy (DR) which represents the leading cause of preventable vision loss in people between 20 and 65 years of age. Sodium glucose transporter 2 (SGLT2) inhibitors have been shown to reduce the risk for cardiovascular and renal events, however literature highlighting their potential role to prevent DR is limited. We therefore used a relevant mouse model (Akimba) to explore the effects of the SGLT2 inhibitor, Empagliflozin (EMPA), on the development of diabetic retinal changes. Here we show that when given in the early stages of type 1 diabetes (T1D), EMPA reduced the weight loss usually associated with T1D, decreased diabetes-associated polydipsia, lowered fasting blood glucose levels, decreased kidney-to-body weight ratios and, most importantly in the current context, substantially reduced retinal abnormalities associated with DR. We show that EMPA reduces vascular leakage indicated by lower albumin staining in the vitreous humor and diminishes expression of the pathogenic factor VEGF in the retina. Additionally, EMPA significantly alters the retinal genetic signature. Our findings suggest that SGLT2 inhibition may be a useful therapeutic approach to prevent the development of DR and its severity if given early in the disease process.
Collapse
|
6
|
Pitale PM, Gorbatyuk MS. Diabetic Retinopathy: From Animal Models to Cellular Signaling. Int J Mol Sci 2022; 23:ijms23031487. [PMID: 35163410 PMCID: PMC8835767 DOI: 10.3390/ijms23031487] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/24/2022] Open
Abstract
Diabetic retinopathy (DR) is an ocular complication of diabetes mellitus (DM), a metabolic disorder characterized by elevation in blood glucose level. The pathogenesis of DR includes vascular, neuronal, and inflammatory components leading to activation of complex cellular molecular signaling. If untreated, the disease can culminate in vision loss that eventually leads to blindness. Animal models mimicking different aspects of DM complications have been developed to study the development and progression of DR. Despite the significant contribution of the developed DR models to discovering the mechanisms of DR and the recent achievements in the research field, the sequence of cellular events in diabetic retinas is still under investigation. Partially, this is due to the complexity of molecular mechanisms, although the lack of availability of models that adequately mimic all the neurovascular pathobiological features observed in patients has also contributed to the delay in determining a precise molecular trigger. In this review, we provide an update on the status of animal models of DR to help investigators choose an appropriate system to validate their hypothesis. We also discuss the key cellular and physiological events of DR in these models.
Collapse
Affiliation(s)
- Priyamvada M. Pitale
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Marina S. Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Correspondence: ; Tel.: +1-205-934-6762; Fax: +1-205-934-3425
| |
Collapse
|
7
|
Weigelt CM, Fuchs H, Schönberger T, Stierstorfer B, Strobel B, Lamla T, Ciossek T, Bakker RA, Redemann NH. AAV-Mediated Expression of Human VEGF, TNF-α, and IL-6 Induces Retinal Pathology in Mice. Transl Vis Sci Technol 2021; 10:15. [PMID: 34520511 PMCID: PMC8444492 DOI: 10.1167/tvst.10.11.15] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinopathies display complex pathologies, including vasculopathies, inflammation, and fibrosis, leading ultimately to visual impairment. However, animal models accurately reflecting these pathologies are lacking. In this study, we evaluate the suitability of using Adeno-associated virus (AAV)-mediated long-term expression of cytokines to establish retinal pathology in the murine retina. Methods We administered recombinant, Müller-glia targeted AAV-ShH10 into the mouse vitreous to induce retinal expression of either human vascular endothelial growth factor (VEGF)-A165, tumor necrosis factor alpha (TNF-α), or interleukin-6 (IL-6) and evaluated consequent effects by optical coherence tomography, fluorescein angiography, and histology. Results Intravitreal injection of AAVs resulted in rapid and stable expression of the transgenes within 1 to 6 weeks. Akin to the role of VEGF-A in wet age-related macular degeneration, expression of VEGF-A led to several vasculopathies in mice, including neovascularization and vascular leakage. In contrast, the expression of the proinflammatory cytokines TNF-α or IL-6 induced retinal inflammation, as indicated by microglial activation. Furthermore, the expression of TNF-α, but not of IL-6, induced immune cell infiltration into the vitreous as well as vasculitis, and subsequently induced the development of fibrosis and epiretinal membranes. Conclusions In summary, the long-term expression of human VEGF-A165, TNF-α, or IL-6 in the mouse eye induced specific pathologies within 6 weeks that mimic different aspects of human retinopathies. Translational Relevance AAV-mediated expression of human genes in mice is an attractive approach to provide valuable insights into the underlying molecular mechanisms causing retinopathies and is easily adaptable to other genes and preclinical species supporting drug discovery for retinal diseases.
Collapse
Affiliation(s)
- Carina M Weigelt
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Holger Fuchs
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Tanja Schönberger
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Birgit Stierstorfer
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Benjamin Strobel
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Thorsten Lamla
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Thomas Ciossek
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Remko A Bakker
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Norbert H Redemann
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| |
Collapse
|
8
|
Guidolin D, Tortorella C, Ribatti D. Spatial Statistics-Based Image Analysis Methods for the Study of Vascular Morphogenesis. Methods Mol Biol 2021; 2206:67-88. [PMID: 32754812 DOI: 10.1007/978-1-0716-0916-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Several studies are available addressing the mechanisms of vascular morphogenesis in order to unravel how cooperative cell behavior can follow from the underlying, genetically regulated behavior of endothelial cells and from cell-to-cell and cell-to-extracellular matrix interactions. From the morphological standpoint several aspects of the process are of interest. They include the way the pattern of vessels fills the available tissue space and how the network grows during the angiogenic process, namely how a main trunk divides into smaller branches, and how branching occurs at different distances from the root point of a vascular tree. A third morphological aspect of interest concerns the spatial relationship between vessels and tissue cells able to secrete factors modulating endothelial cells self-organization, thus influencing vascular rearrangement.In the present chapter image analysis methods allowing for a quantitative characterization of these morphological aspects will be detailed and discussed. They are almost based on concepts derived from the theoretical framework represented by spatial statistics.
Collapse
Affiliation(s)
- Diego Guidolin
- Section of Anatomy, Department of Neuroscience, University of Padova Medical School, Padova, Italy.
| | - Cinzia Tortorella
- Section of Anatomy, Department of Neuroscience, University of Padova Medical School, Padova, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
9
|
The retinal tyrosine kinome of diabetic Akimba mice highlights potential for specific Src family kinase inhibition in retinal vascular disease. Exp Eye Res 2020; 197:108108. [PMID: 32590005 DOI: 10.1016/j.exer.2020.108108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/11/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022]
Abstract
Although anti-VEGF therapies have radically changed clinical practice, there is still an urgent demand for novel, integrative approaches for sight-threatening retinal vascular diseases. As we hypothesize that protein tyrosine kinases are key signaling mediators in retinal vascular disease, we performed a comprehensive activity-based tyrosine kinome profiling on retinal tissue of 12-week-old Akimba mice, a translational model displaying hallmarks of early and advanced diabetic retinopathy. Western blotting was used to confirm retinal tyrosine kinase activity in Akimba mice. HUVEC tube formation and murine organotypic choroidal sprouting assays were applied to compare tyrosine kinase inhibitors with different specificity profiles. HUVEC toxicity and proliferation were evaluated using the CellTox™ Green Cytotoxicity and PrestoBlue™ Assays. Our results indicate a shift of the Akimba retinal tyrosine kinome towards a hyperactive state. Functional network analysis of significantly hyperphosphorylated peptides and upstream kinase prediction revealed a central role for Src-FAK family kinases. Western blotting confirmed hyperactivity of this signaling node in the retina of Akimba mice. We demonstrated that not only Src but also FAK family kinase inhibitors with different selectivity profiles were able to suppress angiogenesis in vitro and ex vivo. In the latter model, the novel selective Src family kinase inhibitor eCF506 was able to achieve potent reduction of angiogenesis, comparable to the less specific inhibitor Dasatinib. None of the tested compounds demonstrated acute endothelial cell toxicity. Overall, the collected findings provide the first comprehensive overview of retinal tyrosine kinome changes in the Akimba model of diabetic retinopathy and for the first time highlight Src family kinase inhibition using highly specific inhibitors as an attractive therapeutic intervention for retinal vascular pathology.
Collapse
|
10
|
Herat LY, Ward NC, Magno AL, Rakoczy EP, Kiuchi MG, Schlaich MP, Matthews VB. Sodium glucose co-transporter 2 inhibition reduces succinate levels in diabetic mice. World J Gastroenterol 2020; 26:3225-3235. [PMID: 32684737 PMCID: PMC7336319 DOI: 10.3748/wjg.v26.i23.3225] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/19/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is associated with major chronic microvascular complications which contribute significantly to diabetes associated morbidity. The protein primarily responsible for glucose reabsorption in the kidney is sodium glucose co-transporter 2 (SGLT2). Presently, SGLT2 inhibitors are widely used in diabetic patients to improve blood glucose levels and prevent cardiovascular and renal complications. Given the broad therapeutic application of SGLT2 inhibitors, we hypothesised that SGLT2 inhibition may exert its protective effects via alterations of the gut microbiome and tested this in a type 1 diabetic mouse model of diabetic retinopathy.
AIM To determine whether the treatment with two independent SGLT2 inhibitors affects gut health in a type 1 diabetic mouse model.
METHODS The SGLT2 inhibitors empagliflozin or dapagliflozin (25 mg/kg/d) or vehicle dimethylsulfoxide (DMSO) were administered to C57BL/6J, Akita, Kimba and Akimba mice at 10 wk of age for 8 wk via their drinking water. Serum samples were collected and the concentration of succinate and the short chain fatty acid (SCFA) butyric acid was measured using gas chromatography-mass spectrometry. Enzyme-linked immunosorbent assay (ELISA) was performed to determine the concentration of insulin and leptin. Furthermore, the norepinephrine content in kidney tissue was determined using ELISA. Pancreatic tissue was collected and stained with haematoxylin and eosin and analysed using brightfield microscopy.
RESULTS Due to the presence of the Akita allele, both Akita and Akimba mice showed a reduction in insulin production compared to C57BL/6J and Kimba mice. Furthermore, Akita mice also showed the presence of apoptotic bodies within the pancreatic islets. The acinar cells of Akita and Akimba mice showed swelling which is indicative of acute injury or pancreatitis. After 8 wk of SGLT2 inhibition with dapagliflozin, the intermediate metabolite of gut metabolism known as succinate was significantly reduced in Akimba mice when compared to DMSO treated mice. In addition, empagliflozin resulted in suppression of succinate levels in Akimba mice. The beneficial SCFA known as butyric acid was significantly increased in Akita mice after treatment with dapagliflozin when compared to vehicle treated mice. The norepinephrine content in the kidney was significantly reduced with both dapagliflozin and empagliflozin therapy in Akita mice and was significantly reduced in Akimba mice treated with empagliflozin. In non-diabetic C57BL/6J and Kimba mice, serum leptin levels were significantly reduced after dapagliflozin therapy.
CONCLUSION The inhibition of SGLT2 reduces the intermediate metabolite succinate, increases SCFA butyric acid levels and reduces norepinephrine content in mouse models of T1D. Collectively, these improvements may represent an important mechanism underlying the potential benefits of SGLT2 inhibition in T1D and its complications.
Collapse
Affiliation(s)
- Lakshini Y Herat
- School of Biomedical Sciences, Dobney Hypertension Centre, Royal Perth Hospital Unit, University of Western Australia, Perth 6000, Australia
| | - Natalie C Ward
- Faculty of Health and Medical Sciences, University of Western Australia, Crawley 6009, Australia
- Faculty of Health Sciences, School of Public Health, Curtin University, Bentley 6102, Australia
| | - Aaron L Magno
- Research Centre, Royal Perth Hospital, Perth 6000, Australia
| | - Elizabeth P Rakoczy
- Department of Molecular Ophthalmology, University of Western Australia, Crawley 6009, Australia
| | - Marcio G Kiuchi
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, University of Western Australia, Perth 6000, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, University of Western Australia, Perth 6000, Australia
- Department of Cardiology and Department of Nephrology, Royal Perth Hospital, Perth 6000, Australia
| | - Vance B Matthews
- School of Biomedical Sciences, Dobney Hypertension Centre, Royal Perth Hospital Unit, University of Western Australia, Perth 6000, Australia
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Diabetic retinopathy (DR) is one of the most common complications associated with chronic hyperglycemia seen in patients with diabetes mellitus. While many facets of DR are still not fully understood, animal studies have contributed significantly to understanding the etiology and progression of human DR. This review provides a comprehensive discussion of the induced and genetic DR models in different species and the advantages and disadvantages of each model. RECENT FINDINGS Rodents are the most commonly used models, though dogs develop the most similar morphological retinal lesions as those seen in humans, and pigs and zebrafish have similar vasculature and retinal structures to humans. Nonhuman primates can also develop diabetes mellitus spontaneously or have focal lesions induced to simulate retinal neovascular disease observed in individuals with DR. DR results in vascular changes and dysfunction of the neural, glial, and pancreatic β cells. Currently, no model completely recapitulates the full pathophysiology of neuronal and vascular changes that occur at each stage of diabetic retinopathy; however, each model recapitulates many of the disease phenotypes.
Collapse
Affiliation(s)
- Ana Maria Olivares
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114 USA
| | - Kristen Althoff
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114 USA
| | - Gloria Fanghua Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114 USA
| | - Siqi Wu
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114 USA
| | | | | | - Neena Haider
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA 02114 USA
| |
Collapse
|
12
|
Imai A, Toriyama Y, Iesato Y, Hirabayashi K, Sakurai T, Kamiyoshi A, Ichikawa-Shindo Y, Kawate H, Tanaka M, Liu T, Xian X, Zhai L, Dai K, Tanimura K, Liu T, Cui N, Yamauchi A, Murata T, Shindo T. Adrenomedullin Suppresses Vascular Endothelial Growth Factor-Induced Vascular Hyperpermeability and Inflammation in Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:999-1015. [PMID: 28322199 DOI: 10.1016/j.ajpath.2017.01.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/19/2017] [Indexed: 11/29/2022]
Abstract
Diabetic macular edema (DME) is caused by blood-retinal barrier breakdown associated with retinal vascular hyperpermeability and inflammation, and it is the major cause of visual dysfunction in diabetic retinopathy. Adrenomedullin (ADM) is an endogenous peptide first identified as a strong vasodilator. ADM is expressed in the eyes and is up-regulated in various eye diseases, although the pathophysiological significance is largely unknown. We investigated the effect of ADM on DME. In Kimba mice, which overexpress human vascular endothelial growth factor in their retinas, the capillary dropout, vascular leakage, and vascular fragility characteristic of diabetic retinopathy were observed. Intravitreal or systemic administration of ADM to Kimba mice ameliorated both the capillary dropout and vascular leakage. Evaluation of the transendothelial electrical resistance and fluorescein isothiocyanate-dextran permeability of an endothelial cell monolayer using TR-iBRB retinal capillary endothelial cells revealed that vascular endothelial growth factor enhanced vascular permeability but that co-administration of ADM suppressed the effect, in part by enhancing tight junction formation between endothelial cells. In addition, a comprehensive PCR array analysis showed that ADM administration suppressed various molecules related to inflammation and NF-κB signaling within retinas. From these results, we suggest that by exerting inhibitory effects on retinal inflammation, vascular permeability, and blood-retinal barrier breakdown, ADM could serve as a novel therapeutic agent for the treatment of DME.
Collapse
Affiliation(s)
- Akira Imai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Yuichi Toriyama
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Yasuhiro Iesato
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Kazutaka Hirabayashi
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan; Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Akiko Kamiyoshi
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Yuka Ichikawa-Shindo
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Hisaka Kawate
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Megumu Tanaka
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Tian Liu
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Xian Xian
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Liuyu Zhai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Kun Dai
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Keiya Tanimura
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Teng Liu
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | - Nanqi Cui
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan
| | | | - Toshinori Murata
- Department of Ophthalmology, Shinshu University School of Medicine, Nagano, Japan
| | - Takayuki Shindo
- Department of Cardiovascular Research, Shinshu University Graduate School of Medicine, Nagano, Japan.
| |
Collapse
|
13
|
Binz N, Rakoczy EP, Ali Rahman IS, Vagaja NN, Lai CM. Biomarkers for Diabetic Retinopathy - Could Endothelin 2 Be Part of the Answer? PLoS One 2016; 11:e0160442. [PMID: 27482904 PMCID: PMC4970817 DOI: 10.1371/journal.pone.0160442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 07/19/2016] [Indexed: 12/17/2022] Open
Abstract
Purpose The endothelins are a family of three highly conserved and homologous vasoactive peptides that are expressed across all organ systems. Endothelin (Edn) dysregulation has been implicated in a number of pathophysiologies, including diabetes and diabetes-related complications. Here we examined Edn2 and endothelin receptor B (Endrb) expression in retinae of diabetic mouse models and measured serum Edn2 to assess its biomarker potential. Materials and Methods Edn2 and Ednrb mRNA and Edn2 protein expression were assessed in young (8wk) and mature (24wk) C57Bl/6 (wild type; wt), Kimba (model of retinal neovascularisation, RNV), Akita (Type 1 diabetes; T1D) and Akimba mice (T1D plus RNV) by qRT-PCR and immunohistochemistry. Edn2 protein concentration in serum was measured using ELISA. Results Fold-changes in Edn2 and Ednrb mRNA were seen only in young Kimba (Edn2: 5.3; Ednrb: 6.0) and young Akimba (Edn2: 7.9, Ednrb: 8.8) and in mature Kimba (Edn2:9.2, Ednrb:11.2) and mature Akimba (Edn2:14.0, Ednrb:17.5) mice. Co-localisation of Edn2 with Müller-cell-specific glutamine synthetase demonstrated Müller cells and photoreceptors as the major cell types for Edn2 expression in all animal models. Edn2 serum concentrations in young Kimba, Akita and Akimba mice were not elevated compared to wt. However, in mature mice, Edn2 serum concentration was increased in Akimba (6.9pg/mg total serum protein) compared to wt, Kimba and Akita mice (3.9, 4.6, and 3.8pg/mg total serum protein, respectively; p<0.05). Conclusions These results demonstrated that long-term hyperglycaemia in conjunction with VEGF-driven RNV increased Edn2 serum concentration suggesting Edn2 might be a candidate biomarker for vascular changes in diabetic retinopathy.
Collapse
MESH Headings
- Animals
- Biomarkers/blood
- Blood Glucose/metabolism
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/diagnosis
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/pathology
- Disease Models, Animal
- Endothelin-2/blood
- Endothelin-2/genetics
- Ependymoglial Cells/metabolism
- Ependymoglial Cells/pathology
- Gene Expression
- Glycated Hemoglobin/metabolism
- Hyperglycemia/blood
- Hyperglycemia/diagnosis
- Hyperglycemia/genetics
- Hyperglycemia/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Photoreceptor Cells, Vertebrate/metabolism
- Photoreceptor Cells, Vertebrate/pathology
- RNA, Messenger/blood
- RNA, Messenger/genetics
- Receptor, Endothelin B/blood
- Receptor, Endothelin B/genetics
- Retinal Neovascularization/blood
- Retinal Neovascularization/diagnosis
- Retinal Neovascularization/genetics
- Retinal Neovascularization/pathology
- Vascular Endothelial Growth Factor A/blood
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Nicolette Binz
- Molecular Ophthalmology, Lions Eye Institute, Nedlands, WA, 6009, Australia
| | - Elizabeth P. Rakoczy
- Molecular Ophthalmology, Lions Eye Institute, Nedlands, WA, 6009, Australia
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, 6009, Australia
- * E-mail:
| | - Ireni S. Ali Rahman
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Nermina N. Vagaja
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Chooi-May Lai
- Molecular Ophthalmology, Lions Eye Institute, Nedlands, WA, 6009, Australia
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, WA, 6009, Australia
| |
Collapse
|
14
|
Lange C, Storkebaum E, de Almodóvar CR, Dewerchin M, Carmeliet P. Vascular endothelial growth factor: a neurovascular target in neurological diseases. Nat Rev Neurol 2016; 12:439-54. [PMID: 27364743 DOI: 10.1038/nrneurol.2016.88] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain function critically relies on blood vessels to supply oxygen and nutrients, to establish a barrier for neurotoxic substances, and to clear waste products. The archetypal vascular endothelial growth factor, VEGF, arose in evolution as a signal affecting neural cells, but was later co-opted by blood vessels to regulate vascular function. Consequently, VEGF represents an attractive target to modulate brain function at the neurovascular interface. On the one hand, VEGF is neuroprotective, through direct effects on neural cells and their progenitors and indirect effects on brain perfusion. In accordance, preclinical studies show beneficial effects of VEGF administration in neurodegenerative diseases, peripheral neuropathies and epilepsy. On the other hand, pathologically elevated VEGF levels enhance vessel permeability and leakage, and disrupt blood-brain barrier integrity, as in demyelinating diseases, for which blockade of VEGF may be beneficial. Here, we summarize current knowledge on the role and therapeutic potential of VEGF in neurological diseases.
Collapse
Affiliation(s)
- Christian Lange
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| | - Erik Storkebaum
- Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, D-48149 Muenster, Germany.,Faculty of Medicine, University of Muenster, Roentgenstrasse 20, D-48149 Muenster, Germany
| | | | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Department of Oncology (KU Leuven) and Vesalius Research Center (VIB), Campus Gasthuisberg O&N4, Herestraat 49 - 912, B-3000, Leuven, Belgium
| |
Collapse
|
15
|
Fu X, Gens JS, Glazier JA, Burns SA, Gast TJ. Progression of Diabetic Capillary Occlusion: A Model. PLoS Comput Biol 2016; 12:e1004932. [PMID: 27300722 PMCID: PMC4907516 DOI: 10.1371/journal.pcbi.1004932] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 04/20/2016] [Indexed: 12/14/2022] Open
Abstract
An explanatory computational model is developed of the contiguous areas of retinal capillary loss which play a large role in diabetic maculapathy and diabetic retinal neovascularization. Strictly random leukocyte mediated capillary occlusion cannot explain the occurrence of large contiguous areas of retinal ischemia. Therefore occlusion of an individual capillary must increase the probability of occlusion of surrounding capillaries. A retinal perifoveal vascular sector as well as a peripheral retinal capillary network and a deleted hexagonal capillary network are modelled using Compucell3D. The perifoveal modelling produces a pattern of spreading capillary loss with associated macular edema. In the peripheral network, spreading ischemia results from the progressive loss of the ladder capillaries which connect peripheral arterioles and venules. System blood flow was elevated in the macular model before a later reduction in flow in cases with progression of capillary occlusions. Simulations differing only in initial vascular network structures but with identical dynamics for oxygen, growth factors and vascular occlusions, replicate key clinical observations of ischemia and macular edema in the posterior pole and ischemia in the retinal periphery. The simulation results also seem consistent with quantitative data on macular blood flow and qualitative data on venous oxygenation. One computational model applied to distinct capillary networks in different retinal regions yielded results comparable to clinical observations in those regions.
Collapse
Affiliation(s)
- Xiao Fu
- The Biocomplexity Institute, Indiana University, Bloomington, Indiana, United States of America
- Department of Physics, Indiana University, Bloomington, Indiana, United States of America
| | - John Scott Gens
- The Biocomplexity Institute, Indiana University, Bloomington, Indiana, United States of America
- Department of Physics, Indiana University, Bloomington, Indiana, United States of America
| | - James A. Glazier
- The Biocomplexity Institute, Indiana University, Bloomington, Indiana, United States of America
- Department of Physics, Indiana University, Bloomington, Indiana, United States of America
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, United States of America
| | - Stephen A. Burns
- School of Optometry, Indiana University, Bloomington, Indiana, United States of America
| | - Thomas J. Gast
- School of Optometry, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
16
|
Kim YR, Hong SH. Association between the polymorphisms of the vascular endothelial growth factor gene and metabolic syndrome. Biomed Rep 2015; 3:319-326. [PMID: 26137230 DOI: 10.3892/br.2015.423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/27/2015] [Indexed: 01/12/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a major angiogenic factor. Increased levels of VEGF have been reported in patients with metabolic syndrome (MetS). The role of VEGF polymorphisms in MetS susceptibility, however, has not been reported previously. Thus, the present study was performed to analyze the associations between the VEGF -634G>C and 936C>T polymorphisms and the patients with MetS. A total of 320 patients with MetS (mean age, 49.86±11.76 years) and 320 healthy subjects (mean age, 50.94±8.43 years) were enrolled in the study. The VEGF -634G>C polymorphism in the 5'-untranslated region (UTR) and 936C>T polymorphism in 3'-UTR were analyzed by polymerase chain reaction-restriction fragment length polymorphism. The VEGF -634G>C polymorphism significantly affected MetS susceptibility. The CC genotype of the -634G>C polymorphism was significantly associated with an increased risk of MetS [adjusted odds ratio (AOR)=3.973; 95% confidence interval (CI), 2.321-6.799; P<0.0001]. AORs of the dominant (GG vs. GC+CC) and recessive models (GG+GC vs. CC) between the cases and controls were 2.569 (95% CI, 1.657-3.983; P<0.0001) and 2.163 (95% CI, 1.475-3.171; P=0.0001), respectively. Haplotypes of -634G>C and 936C>T were also associated with MetS susceptibility. When the haplotype data were stratified by gender, the association remained only in males. The -634G>C polymorphism was also associated with the subgroups of MetS risk components by the stratification analysis. The 936C>T polymorphism was, however, not associated with the MetS susceptibility. The present study demonstrates that the VEGF -634G>C polymorphism and haplotypes may be a genetic determinant for the MetS susceptibility. To the best of our knowledge, this is the first study on the significant association of the VEGF polymorphisms in MetS patients. To confirm the effects of the VEGF polymorphisms on MetS, further functional and population studies are required.
Collapse
Affiliation(s)
- Young Ree Kim
- Department of Laboratory Medicine, School of Medicine, Jeju National University, Jeju 690-756, Republic of Korea
| | - Seung-Ho Hong
- Department of Science Education, Teachers College, Jeju National University, Jeju 690-781, Republic of Korea
| |
Collapse
|
17
|
Shen W, Chung SH, Irhimeh MR, Li S, Lee SR, Gillies MC. Systemic administration of erythropoietin inhibits retinopathy in RCS rats. PLoS One 2014; 9:e104759. [PMID: 25119659 PMCID: PMC4132022 DOI: 10.1371/journal.pone.0104759] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/12/2014] [Indexed: 12/17/2022] Open
Abstract
Objective Royal College of Surgeons (RCS) rats develop vasculopathy as photoreceptors degenerate. The aim of this study was to examine the effect of erythropoietin (EPO) on retinopathy in RCS rats. Methods Fluorescein angiography was used to monitor retinal vascular changes over time. Changes in retinal glia and vasculature were studied by immunostaining. To study the effects of EPO on retinal pathology, EPO (5000 IU/kg) was injected intraperitoneally in 14 week old normal and RCS rats twice a week for 4 weeks. Changes in the retinal vasculature, glia and microglia, photoreceptor apoptosis, differential expression of p75 neurotrophin receptor (p75NTR), pro-neurotrophin 3 (pro-NT3), tumour necrosis factor-α (TNFα), pigment epithelium derived factor (PEDF) and vascular endothelial growth factor-A (VEGF-A), the production of CD34+ cells and mobilization of CD34+/VEGF-R2+ cells as well as recruitment of CD34+ cells into the retina were examined after EPO treatment. Results RCS rats developed progressive capillary dropout and subretinal neovascularization which were accompanied by retinal gliosis. Systemic administration of EPO stabilized the retinal vasculature and inhibited the development of focal vascular lesions. Further studies showed that EPO modulated retinal gliosis, attenuated photoreceptor apoptosis and p75NTR and pro-NT3 upregulation, promoted the infiltration of ramified microglia and stimulated VEGF-A expression but had little effect on TNFα and PEDF expression. EPO stimulated the production of red and white blood cells and CD34+ cells along with effective mobilization of CD34+/VEGF-R2+ cells. Immunofluorescence study demonstrated that EPO enhanced the recruitment of CD34+ cells into the retina. Conclusions Our results suggest that EPO has therapeutic potentials in treatment of neuronal and vascular pathology in retinal disease. The protective effects of EPO on photoreceptors and the retinal vasculature may involve multiple mechanisms including regulation of retinal glia and microglia, inhibition of p75NTR-pro-NT3 signaling together with stimulation of production and mobilization of bone marrow derived cells.
Collapse
Affiliation(s)
- Weiyong Shen
- Save Sight Institute, the University of Sydney, Sydney, Australia
| | - Sook H Chung
- Save Sight Institute, the University of Sydney, Sydney, Australia
| | | | - Shiying Li
- Save Sight Institute, the University of Sydney, Sydney, Australia; Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, China
| | - So-Ra Lee
- Save Sight Institute, the University of Sydney, Sydney, Australia
| | - Mark C Gillies
- Save Sight Institute, the University of Sydney, Sydney, Australia
| |
Collapse
|
18
|
Wisniewska-Kruk J, Klaassen I, Vogels IMC, Magno AL, Lai CM, Van Noorden CJF, Schlingemann RO, Rakoczy EP. Molecular analysis of blood-retinal barrier loss in the Akimba mouse, a model of advanced diabetic retinopathy. Exp Eye Res 2014; 122:123-31. [PMID: 24703908 DOI: 10.1016/j.exer.2014.03.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 03/06/2014] [Accepted: 03/17/2014] [Indexed: 12/31/2022]
Abstract
The molecular mechanisms of vascular leakage in diabetic macular edema and proliferative retinopathy are poorly understood, mainly due to the lack of reliable in vivo models. The Akimba (Ins2(Akita)VEGF(+/-)) mouse model combines retinal neovascularization with hyperglycemia, and in contrast to other models, displays the majority of signs of advanced clinical diabetic retinopathy (DR). To study the molecular mechanism that underlies the breakdown of the blood-retinal barrier (BRB) in diabetic macular edema and proliferative diabetic retinopathy, we investigated the retinal vasculature of Akimba and its parental mice Kimba (trVEGF029) and Akita (Ins2(Akita)). Quantitative PCR, immunohistochemistry and fluorescein angiography were used to characterize the retinal vasculature with special reference to the inner BRB. Correlations between the degree of fluorescein leakage and retinal gene expression were tested by calculating the Spearman's correlation coefficient. Fluorescein leakage demonstrating BRB loss was observed in Kimba and Akimba, but not in Akita or wild type mice. In Kimba and Akimba mice fluorescein leakage was associated with focal angiogenesis and correlated significantly with Plvap gene expression. PLVAP is an endothelial cell-specific protein that is absent in intact blood-retinal barrier, but its expression significantly increases in pathological conditions such as DR. Furthermore, in Akimba mice BRB disruption was linked to decreased expression of endothelial junction proteins, pericyte dropout and vessel loss. Despite fluorescein leakage, no alteration in BRB protein levels or pericyte coverage was detected in retinas of Kimba mice. In summary, our data not only demonstrate that hyperglycemia sensitizes retinal vasculature to the effects of VEGF, leading to more severe microvascular changes, but also confirm an important role of PLVAP in the regulation of BRB permeability.
Collapse
Affiliation(s)
- Joanna Wisniewska-Kruk
- Ocular Angiogenesis Group, Departments of Ophthalmology and Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Ilse M C Vogels
- Ocular Angiogenesis Group, Departments of Ophthalmology and Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Aaron L Magno
- Department of Molecular Ophthalmology, Lions Eye Institute, Nedlands, Western Australia, Australia.
| | - Chooi-May Lai
- Centre for Ophthalmology and Visual Sciences, The University of Western Australia, Nedlands, Western Australia, Australia.
| | - Cornelis J F Van Noorden
- Ocular Angiogenesis Group, Departments of Ophthalmology and Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Clinical and Molecular Ophthalmogenetics, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Science (KNAW), Amsterdam, The Netherlands.
| | - Elizabeth P Rakoczy
- Centre for Ophthalmology and Visual Sciences, The University of Western Australia, Nedlands, Western Australia, Australia.
| |
Collapse
|
19
|
Shen W, Zhu L, Lee SR, Chung SH, Gillies MC. Involvement of NT3 and P75(NTR) in photoreceptor degeneration following selective Müller cell ablation. J Neuroinflammation 2013; 10:137. [PMID: 24224958 PMCID: PMC3831588 DOI: 10.1186/1742-2094-10-137] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 11/01/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurotrophins can regulate opposing functions that result in cell survival or apoptosis, depending on which form of the protein is secreted and which receptor and signaling pathway is activated. We have recently developed a transgenic model in which inducible and patchy Müller cell ablation leads to photoreceptor degeneration. This study aimed to examine the roles of mature neurotrophin-3 (NT3), pro-NT3 and p75 neurotrophin receptor (P75(NTR)) in photoreceptor degeneration in this model. METHODS Transgenic mice received tamoxifen to induce Müller cell ablation. Changes in the status of Müller and microglia cells as well as expression of mature NT3, pro-NT3 and P75(NTR) were examined by immunohistochemistry and Western blot analysis. Recombinant mature NT3 and an antibody neutralizing 75(NTR) were injected intravitreally 3 and 6 days after Müller cell ablation to examine their effects on photoreceptor degeneration and microglial activation. RESULTS We found that patchy loss of Müller cells was associated with activation of surviving Müller cells and microglial cells, concurrently with reduced expression of mature NT3 and upregulation of pro-NT3 and P75(NTR). Intravitreal injection of mature NT3 and a neutralizing antibody to P75NTR, either alone or in combination, attenuated photoreceptor degeneration and the beneficial effect was associated with inhibition of microglial activation. CONCLUSIONS Our data suggest that Müller cell ablation alters the balance between the protective and deleterious effects of mature NT3 and pro-NT3. Modulation of the neuroprotective action of mature NT3 and pro-apoptotic pro-NT3/P75(NTR) signaling may represent a novel pharmacological strategy for photoreceptor protection in retinal disease.
Collapse
Affiliation(s)
- Weiyong Shen
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - Ling Zhu
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - So-Ra Lee
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - Sook H Chung
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| | - Mark C Gillies
- Save Sight Institute, the University of Sydney, 8 Macquarie Street, Sydney 2000, Australia
| |
Collapse
|
20
|
Qiu M, Xiong W, Liao H, Li F. VEGF -634G>C polymorphism and diabetic retinopathy risk: a meta-analysis. Gene 2013; 518:310-5. [PMID: 23353010 DOI: 10.1016/j.gene.2013.01.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 01/09/2013] [Accepted: 01/11/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF), a multifunctional cytokine that promotes angiogenesis and is a potent mediator of microvascular permeability, which is critical for the development of diabetic retinopathy (DR). It has demonstrated that VEGF -634G>C (rs2010963) polymorphism alters the transcriptional activity of the gene. However, studies on the association between VEGF -634G>C polymorphism and DR in type 2 diabetes have reported conflicting results. Thus, the aim of the present study was to investigate whether VEGF -634G>C polymorphism is associated with the risk of DR in type 2 diabetes. METHODS A systematic search of electronic databases (PubMed, Embase and Web of Science) and reference lists of relevant articles was carried out until September 15, 2012. The pooled odds ratios (ORs) and their corresponding 95% confidence interval (CI) were calculated by a fixed effect model. RESULTS A total of 1525 DR cases and 1422 diabetic without retinopathy (DWR) controls in 9 independent studies were included in the meta-analysis. A significant relationship between VEGF -634G>C polymorphism and DR was found in an allelic genetic model (OR: 1.13, 95% CI: 1.01 to 1.25, P=0.03) and a recessive genetic model (OR: 1.26, 95% CI: 1.02 to 1.55, P=0.03). CONCLUSION Our research confirmed the association between the VEGF -634G>C polymorphism and DR in subjects with type 2 diabetes. Well-designed studies with larger sample size and more ethnic groups are required to further validate the results.
Collapse
Affiliation(s)
- Min Qiu
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave, Wuhan 430030, People's Republic of China
| | | | | | | |
Collapse
|
21
|
Conditional Müllercell ablation causes independent neuronal and vascular pathologies in a novel transgenic model. J Neurosci 2013; 32:15715-27. [PMID: 23136411 DOI: 10.1523/jneurosci.2841-12.2012] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Müller cells are the major glia of the retina that serve numerous functions essential to retinal homeostasis, yet the contribution of Müller glial dysfunction to retinal diseases remains largely unknown. We have developed a transgenic model using a portion of the regulatory region of the retinaldehyde binding protein 1 gene for conditional Müller cell ablation and the consequences of primary Müller cell dysfunction have been studied in adult mice. We found that selective ablation of Müller cells led to photoreceptor apoptosis, vascular telangiectasis, blood-retinal barrier breakdown and, later, intraretinal neovascularization. These changes were accompanied by impaired retinal function and an imbalance between vascular endothelial growth factor-A (VEGF-A) and pigment epithelium-derived factor. Intravitreal injection of ciliary neurotrophic factor inhibited photoreceptor injury but had no effect on the vasculopathy. Conversely, inhibition of VEGF-A activity attenuated vascular leak but did not protect photoreceptors. Our findings show that Müller glial deficiency may be an important upstream cause of retinal neuronal and vascular pathologies in retinal diseases. Combined neuroprotective and anti-angiogenic therapies may be required to treat Müller cell deficiency in retinal diseases and in other parts of the CNS associated with glial dysfunction.
Collapse
|
22
|
Bharadwaj AS, Appukuttan B, Wilmarth PA, Pan Y, Stempel AJ, Chipps TJ, Benedetti EE, Zamora DO, Choi D, David LL, Smith JR. Role of the retinal vascular endothelial cell in ocular disease. Prog Retin Eye Res 2013; 32:102-80. [PMID: 22982179 PMCID: PMC3679193 DOI: 10.1016/j.preteyeres.2012.08.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 08/23/2012] [Accepted: 08/24/2012] [Indexed: 12/14/2022]
Abstract
Retinal endothelial cells line the arborizing microvasculature that supplies and drains the neural retina. The anatomical and physiological characteristics of these endothelial cells are consistent with nutritional requirements and protection of a tissue critical to vision. On the one hand, the endothelium must ensure the supply of oxygen and other nutrients to the metabolically active retina, and allow access to circulating cells that maintain the vasculature or survey the retina for the presence of potential pathogens. On the other hand, the endothelium contributes to the blood-retinal barrier that protects the retina by excluding circulating molecular toxins, microorganisms, and pro-inflammatory leukocytes. Features required to fulfill these functions may also predispose to disease processes, such as retinal vascular leakage and neovascularization, and trafficking of microbes and inflammatory cells. Thus, the retinal endothelial cell is a key participant in retinal ischemic vasculopathies that include diabetic retinopathy and retinopathy of prematurity, and retinal inflammation or infection, as occurs in posterior uveitis. Using gene expression and proteomic profiling, it has been possible to explore the molecular phenotype of the human retinal endothelial cell and contribute to understanding of the pathogenesis of these diseases. In addition to providing support for the involvement of well-characterized endothelial molecules, profiling has the power to identify new players in retinal pathologies. Findings may have implications for the design of new biological therapies. Additional progress in this field is anticipated as other technologies, including epigenetic profiling methods, whole transcriptome shotgun sequencing, and metabolomics, are used to study the human retinal endothelial cell.
Collapse
Affiliation(s)
| | | | - Phillip A. Wilmarth
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University
| | - Yuzhen Pan
- Casey Eye Institute, Oregon Health & Science University
| | | | | | | | | | - Dongseok Choi
- Department of Public Health and Preventive Medicine, Oregon Health & Science University
| | - Larry L. David
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University
| | - Justine R. Smith
- Casey Eye Institute, Oregon Health & Science University
- Department of Cell & Developmental Biology, Oregon Health & Science University
| |
Collapse
|
23
|
Lai AKW, Lo ACY. Animal models of diabetic retinopathy: summary and comparison. J Diabetes Res 2013; 2013:106594. [PMID: 24286086 PMCID: PMC3826427 DOI: 10.1155/2013/106594] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 09/02/2013] [Accepted: 09/02/2013] [Indexed: 12/16/2022] Open
Abstract
Diabetic retinopathy (DR) is a microvascular complication associated with chronic exposure to hyperglycemia and is a major cause of blindness worldwide. Although clinical assessment and retinal autopsy of diabetic patients provide information on the features and progression of DR, its underlying pathophysiological mechanism cannot be deduced. In order to have a better understanding of the development of DR at the molecular and cellular levels, a variety of animal models have been developed. They include pharmacological induction of hyperglycemia and spontaneous diabetic rodents as well as models of angiogenesis without diabetes (to compensate for the absence of proliferative DR symptoms). In this review, we summarize the existing protocols to induce diabetes using STZ. We also describe and compare the pathological presentations, in both morphological and functional aspects, of the currently available DR animal models. The advantages and disadvantages of using different animals, ranging from zebrafish, rodents to other higher-order mammals, are also discussed. Until now, there is no single model that displays all the clinical features of DR as seen in human. Yet, with the understanding of the pathological findings in these animal models, researchers can select the most suitable models for mechanistic studies or drug screening.
Collapse
Affiliation(s)
- Angela Ka Wai Lai
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Amy C. Y. Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- *Amy C. Y. Lo:
| |
Collapse
|
24
|
Binz N, Ali Rahman IS, Chinnery HR, McKeone R, Simpson KM, Speed TP, Lai CM, Rakoczy PE. Effect of vascular endothelial growth factor upregulation on retinal gene expression in the Kimba mouse. Clin Exp Ophthalmol 2012; 41:251-62. [PMID: 22788671 DOI: 10.1111/j.1442-9071.2012.02845.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The Kimba mouse carries a human vascular endothelial growth factor transgene causing retinal neovascularisation similar to that seen in diabetic retinopathy. Here, we examine the relationship between differential gene expression induced by vascular endothelial growth factor overexpression and the architectural changes that occur in the retinae of these mice. METHODS Retinal gene expression changes in juvenile and adult Kimba mice were assayed by microarray and compared with age-matched wild-type littermates. Transcription of selected genes was validated by quantitative real-time polymerase chain reaction. Protein translation was determined using immunohistochemistry and enzyme-linked immunosorbent assay. RESULTS Semaphorin 3C was upregulated, and nuclear receptor subfamily 2, group 3, member 3 (Nr2e3) was downregulated in juvenile Kimba mice. Betacellulin and endothelin 2 were upregulated in adults. Semaphorin 3C colocalized with glial fibrillary acidic protein in Müller cells of Kimba retinae at greater signal intensities than in wild type. Endothelin 2 colocalised to Müller cell end feet and extended into the outer limiting membrane. Endothelin receptor type B staining was most pronounced in the inner nuclear layer, the region containing Müller cell somata. CONCLUSIONS An early spike in vascular endothelial growth factor induced significant long-term retinal neovascularisation associated with changes to the retinal ganglion, photoreceptor and Müller cells. Overexpression of vascular endothelial growth factor led to dysregulation of photoreceptor metabolism through differential expression of Nr2e3, endothelin 2, betacellulin and semaphorin 3C. Alterations in the expression of these genes may therefore play key roles in the pathological mechanisms that result from retinal neovascularisation.
Collapse
Affiliation(s)
- Nicolette Binz
- Department of Molecular Ophthalmology, Lions Eye Institute, Nedlands Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Western Australia
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kim YH, Park SY, Park J, Kim YS, Hwang EM, Park JY, Roh GS, Kim HJ, Kang SS, Cho GJ, Choi WS. Reduction of experimental diabetic vascular leakage and pericyte apoptosis in mice by delivery of αA-crystallin with a recombinant adenovirus. Diabetologia 2012; 55:2835-2844. [PMID: 22772798 DOI: 10.1007/s00125-012-2625-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 05/23/2012] [Indexed: 01/27/2023]
Abstract
AIMS/HYPOTHESIS The study aimed to evaluate the efficacy of recombinant adenovirus expressing αA-crystallin (Ad-αAc-Gfp) in reducing pericyte loss within retinal vasculature in early diabetes. METHODS Diabetes was induced by streptozotocin injection into C57BL/6 mice. Ad-αAc-Gfp was delivered by intravitreous injection to the right eyes of mice 2 weeks before induction of diabetes. Vascular leakage was determined by fluorescent angiography, Evans Blue leakage assay and leucocyte adhesion test. Production of αA-crystallin was analysed by immunoblotting and double immunostaining and pericyte loss was analysed by pericyte count. RESULTS Vessel leakage and pericyte loss were observed in the streptozotocin-induced diabetic retina. Decreased abundance of αA-crystallin in retinas 2 and 6 months after the induction of diabetes was confirmed by two-dimensional electrophoretic analysis, immunoblotting and RT-PCR. Double immunofluorescence staining for αA-crystallin and NG2 chondroitin sulphate proteoglycan revealed that αA-crystallin was predominantly produced in the retinal pericyte and that the number of αA-crystallin-producing pericytes decreased in the diabetic retina. Retinal infection with Ad-αAc-Gfp led to decreased pericyte loss and vascular leakage compared with control. CONCLUSIONS/INTERPRETATION Intravitreal delivery of Ad-αAc-Gfp protects against vascular leakage in the streptozotocin-induced model of diabetes. This effect is associated with the inhibition of diabetic retinal pericyte loss in early diabetes, suggesting that αA-crystallin has a role in preventing the pathogenesis of early diabetic retinopathy.
Collapse
Affiliation(s)
- Y H Kim
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - S Y Park
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - J Park
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - Y S Kim
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - E M Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - J Y Park
- Department of Physiology, School of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - G S Roh
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - H J Kim
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - S S Kang
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - G J Cho
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea
| | - W S Choi
- Department of Anatomy and Neurobiology, Medical Research Center for Neural Dysfunction, Institute of Health Science, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam, 660-751, Republic of Korea.
| |
Collapse
|
26
|
Ali Rahman IS, Li CR, Lai CM, Rakoczy EP. In VivoMonitoring of VEGF-Induced Retinal Damage in the Kimba Mouse Model of Retinal Neovascularization. Curr Eye Res 2011; 36:654-62. [DOI: 10.3109/02713683.2010.551172] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
27
|
Placental growth factor contributes to micro-vascular abnormalization and blood-retinal barrier breakdown in diabetic retinopathy. PLoS One 2011; 6:e17462. [PMID: 21408222 PMCID: PMC3049767 DOI: 10.1371/journal.pone.0017462] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 02/03/2011] [Indexed: 12/28/2022] Open
Abstract
Objective There are controversies regarding the pro-angiogenic activity of placental growth factor (PGF) in diabetic retinopathy (DR). For a better understanding of its role on the retina, we have evaluated the effect of a sustained PGF over-expression in rat ocular media, using ciliary muscle electrotransfer (ET) of a plasmid encoding rat PGF-1 (pVAX2-rPGF-1). Materials and Methods pVAX2-rPGF-1 ET in the ciliary muscle (200 V/cm) was achieved in non diabetic and diabetic rat eyes. Control eyes received saline or naked plasmid ET. Clinical follow up was carried out over three months using slit lamp examination and fluorescein angiography. After the control of rPGF-1 expression, PGF-induced effects on retinal vasculature and on the blood-external barrier were evaluated respectively by lectin and occludin staining on flat-mounts. Ocular structures were visualized through histological analysis. Results After fifteen days of rPGF-1 over-expression in normal eyes, tortuous and dilated capillaries were observed. At one month, microaneurysms and moderate vascular sprouts were detected in mid retinal periphery in vivo and on retinal flat-mounts. At later stages, retinal pigmented epithelial cells demonstrated morphological abnormalities and junction ruptures. In diabetic retinas, PGF expression rose between 2 and 5 months, and, one month after ET, rPGF-1 over-expression induced glial activation and proliferation. Conclusion This is the first demonstration that sustained intraocular PGF production induces vascular and retinal changes similar to those observed in the early stages of diabetic retinopathy. PGF and its receptor Flt-1 may therefore be looked upon as a potential regulatory target at this stage of the disease.
Collapse
|
28
|
Rakoczy EP, Ali Rahman IS, Binz N, Li CR, Vagaja NN, de Pinho M, Lai CM. Characterization of a mouse model of hyperglycemia and retinal neovascularization. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2659-70. [PMID: 20829433 DOI: 10.2353/ajpath.2010.090883] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
One of the limitations of research into diabetic retinopathy is the lack of suitable animal models. To study how the two important factors--hyperglycemia and vascular endothelial growth factor--interact in diabetic retinopathy, the Akimba mouse (Ins2AkitaVEGF+/-) was generated by crossing the Akita mouse (Ins2Akita) with the Kimba mouse (VEGF+/+). C57Bl/6 and the parental and Akimba mouse lines were characterized by biometric measurements, histology, immunohistochemistry, and Spectralis Heidelberg retinal angiography and optical coherence tomography. The Akimba line not only retained the characteristics of the parental strains, such as developing hyperglycemia and retinal neovascularization, but developed higher blood glucose levels at a younger age and had worse kidney-body weight ratios than the Akita line. With aging, the Akimba line demonstrated enhanced photoreceptor cell loss, thinning of the retina, and more severe retinal vascular pathology, including more severe capillary nonperfusion, vessel constriction, beading, neovascularization, fibroses, and edema, compared with the Kimba line. The vascular changes were associated with major histocompatibility complex class II+ cellular staining throughout the retina. Together, these observations suggest that hyperglycemia resulted in higher prevalences of edema and exacerbated the vascular endothelial growth factor-driven neovascular and retinal changes in the Akimba line. Thus, the Akimba line could become a useful model for studying the interplay between hyperglycemia and vascular endothelial growth factor and for testing treatment strategies for potentially blinding complications, such as edema.
Collapse
Affiliation(s)
- Elizabeth P Rakoczy
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Crawley, Australia.
| | | | | | | | | | | | | |
Collapse
|
29
|
Wang J, Xu X, Elliott MH, Zhu M, Le YZ. Müller cell-derived VEGF is essential for diabetes-induced retinal inflammation and vascular leakage. Diabetes 2010; 59:2297-305. [PMID: 20530741 PMCID: PMC2927953 DOI: 10.2337/db09-1420] [Citation(s) in RCA: 283] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF-A or VEGF) is a major pathogenic factor and therapeutic target for diabetic retinopathy (DR). Since VEGF has been proposed as a survival factor for retinal neurons, defining the cellular origin of pathogenic VEGF is necessary for the effectiveness and safety of long-term anti-VEGF therapies for DR. To determine the significance of Müller cell-derived VEGF in DR, we disrupted VEGF in Müller cells with an inducible Cre/lox system and examined diabetes-induced retinal inflammation and vascular leakage in these conditional VEGF knockout (KO) mice. RESEARCH DESIGN AND METHODS Leukostasis was determined by counting the number of fluorescently labeled leukocytes inside retinal vasculature. Expression of biomarkers for retinal inflammation was assessed by immunoblotting of TNF-alpha, ICAM-1, and NF-kappaB. Vascular leakage was measured by immunoblotting of retinal albumin and fluorescent microscopic analysis of extravascular albumin. Diabetes-induced vascular alterations were examined by immunoblotting and immunohistochemistry for tight junctions, and by trypsin digestion assays for acellular capillaries. Retinal integrity was analyzed with morphologic and morphometric analyses. RESULTS Diabetic conditional VEGF KO mice exhibited significantly reduced leukostasis, expression of inflammatory biomarkers, depletion of tight junction proteins, numbers of acellular capillaries, and vascular leakage compared to diabetic control mice. CONCLUSIONS Müller cell-derived VEGF plays an essential and causative role in retinal inflammation, vascular lesions, and vascular leakage in DR. Therefore, Müller cells are a primary cellular target for proinflammatory signals that mediates retinal inflammation and vascular leakage in DR.
Collapse
Affiliation(s)
- Juanjuan Wang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- Department of Medicine Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Harold Hamm Oklahoma Diabetes Center, University of Oklahoma, Oklahoma City, Oklahoma
| | - Xueliang Xu
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Michael H. Elliott
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Dean A. McGee Eye Institute, Oklahoma City, Oklahoma
| | - Meili Zhu
- Department of Medicine Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Harold Hamm Oklahoma Diabetes Center, University of Oklahoma, Oklahoma City, Oklahoma
| | - Yun-Zheng Le
- Department of Medicine Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Harold Hamm Oklahoma Diabetes Center, University of Oklahoma, Oklahoma City, Oklahoma
- Dean A. McGee Eye Institute, Oklahoma City, Oklahoma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Corresponding author: Yun-Zheng Le,
| |
Collapse
|
30
|
Shen W, Li S, Chung SH, Gillies MC. Retinal vascular changes after glial disruption in rats. J Neurosci Res 2010; 88:1485-99. [PMID: 20029988 DOI: 10.1002/jnr.22317] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Glial dysfunction is found in a number of retinal vascular diseases but its link with blood-retinal barrier (BRB) breakdown remains poorly understood. The present study tested the hypothesis that glial dysfunction is a major contributor to the BRB breakdown that is a hallmark of retinal vascular diseases. We investigated the specificity of the purportedly selective glial toxin, DL-alpha-aminoadipic acid (DL-alpha-AAA) on different types of ocular cells in vitro and then tested the effect of glial disruption on retinal vasculature after intraocular injection of DL-alpha-AAA or siRNA targeting glutamine synthetase (GS) in rats. DL-alpha-AAA was toxic to astrocytes and Müller cells but not to other types of BRB-related cells in vitro. Subretinal injection of DL-alpha-AAA disrupted retinal glial cells, induced vascular telangiectasis and increased vascular permeability from 4 days to over 2 months post-injection. Vascular changes induced by DL-alpha-AAA were observed predominantly in regions of glial disruption, as reflected by reduced expression of GS and increased expression of glial fibrillary acidic protein and vimentin. Confocal microscopy showed changes in all three layers of the retinal vasculature, which co-localised with areas of Müller cell disruption. Double labeling immunohistochemistry revealed that retinal glial disruption after DL-alpha-AAA injection was accompanied by increased expression of vascular endothelial growth factor and reduced expression of the tight junction protein claudin-5. Intravitreal injection of GS siRNA induced similar changes in Müller cells and BRB breakdown. Our data are consistent with the hypothesis that glial dysfunction is a primary contributor to the BRB breakdown in retinal vascular diseases.
Collapse
Affiliation(s)
- Weiyong Shen
- Save Sight Institute, University of Sydney, Sydney, Australia.
| | | | | | | |
Collapse
|
31
|
Bai Y, Ma JX, Guo J, Wang J, Zhu M, Chen Y, Le YZ. Müller cell-derived VEGF is a significant contributor to retinal neovascularization. J Pathol 2010; 219:446-54. [PMID: 19768732 DOI: 10.1002/path.2611] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vascular endothelial growth factor (VEGF-A) is a major pathogenic factor and a therapeutic target for age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity. Despite intensive effort in the field, the cellular mechanisms of VEGF action remain virtually uninvestigated. This situation makes it difficult to design cellular target-based therapeutics for these diseases. In light of the recent finding that VEGF is a potential neurotrophic factor, revealing the cellular mechanisms of VEGF action becomes necessary to preserve its beneficial effect and inhibit its pathological function in long-term anti-VEGF therapeutics for ocular vascular diseases. We therefore generated conditional VEGF knockout mice with an inducible Cre/lox system and determined the significance of Müller cell-derived VEGF in retinal development and maintenance and ischaemia-induced neovascularizartion and vascular leakage. Retinal development in the conditional VEGF knockout mice was analysed by examining retinal and choroidal vasculatures and retinal morphology and function. Ischaemia-induced retinal neovascularization and vascular leakage in the conditional VEGF knockout mice were analysed with fluorescein angiography, quantification of proliferative neovascular cells, immunohistochemistry, and immunoblotting using an oxygen-induced retinopathy model. Our results demonstrated that disruption of Müller cell-derived VEGF resulted in no apparent defects in retinal and choroidal vasculatures and retinal morphology and function, significant inhibition of the ischaemia-induced retinal neovascularization and vascular leakage, and attenuation of the ischaemia-induced breakdown of the blood-retina barrier. These results suggest that the retinal Müller cell-derived VEGF is a major contributor to ischaemia-induced retinal vascular leakage and pre-retinal and intra-retinal neovascularization. The observation that a significant, but not complete, reduction of VEGF in the retina does not cause detectable retinal degeneration suggests that appropriate doses of anti-VEGF agents may be important to the safe treatment of retinal vascular diseases.
Collapse
Affiliation(s)
- Yanyan Bai
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Bussolati B, Ribatti D, Munaron L, Bartorelli A, Bussolati G. Anti-angiogenic properties of calcium trifluoroacetate. Microvasc Res 2009; 78:272-7. [PMID: 19631669 DOI: 10.1016/j.mvr.2009.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 07/15/2009] [Accepted: 07/16/2009] [Indexed: 10/20/2022]
Abstract
Endothelial cell proliferation and the formation of new vessels are strictly regulated by angiogenic factors (e.g., VEGF) that induce the activation of signal transduction pathways controlled by calcium dynamics. Using in vitro and in vivo experiments, we investigated the effect of calcium trifluoroacetate (CaTFAc), a complex, poorly dissociated salt that is characterized by its low toxicity, on angiogenesis. In vitro, CaTFAc inhibited VEGF-induced effects on endothelial cell proliferation. In two in vivo models of angiogenesis, a Matrigel plug in mice and a chick embryo chorioallantoic membrane, CaTFAc inhibited the VEGF-induced formation of new vessels. The exact mechanism of action is still under investigation, but in vitro experiments demonstrate that CaTFAc induced a reversible increase in the levels of intracellular calcium under basal conditions and prevented calcium signaling induced by VEGF. These results are the first to suggest that CaTFAc may be useful for the treatment of diseases caused by enhanced angiogenesis.
Collapse
Affiliation(s)
- Benedetta Bussolati
- Department of Internal Medicine, Centre for Molecular Biotechnology, University of Turin, Italy
| | | | | | | | | |
Collapse
|
33
|
Ruiz de Almodovar C, Lambrechts D, Mazzone M, Carmeliet P. Role and therapeutic potential of VEGF in the nervous system. Physiol Rev 2009; 89:607-48. [PMID: 19342615 DOI: 10.1152/physrev.00031.2008] [Citation(s) in RCA: 337] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The development of the nervous and vascular systems constitutes primary events in the evolution of the animal kingdom; the former provides electrical stimuli and coordination, while the latter supplies oxygen and nutrients. Both systems have more in common than originally anticipated. Perhaps the most striking observation is that angiogenic factors, when deregulated, contribute to various neurological disorders, such as neurodegeneration, and might be useful for the treatment of some of these pathologies. The prototypic example of this cross-talk between nerves and vessels is the vascular endothelial growth factor or VEGF. Although originally described as a key angiogenic factor, it is now well established that VEGF also plays a crucial role in the nervous system. We describe the molecular properties of VEGF and its receptors and review the current knowledge of its different functions and therapeutic potential in the nervous system during development, health, disease and in medicine.
Collapse
|
34
|
Kiick KL. Peptide- and protein-mediated assembly of heparinized hydrogels. SOFT MATTER 2008; 4:29-37. [PMID: 19960073 PMCID: PMC2787454 DOI: 10.1039/b711319f] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Polymeric hydrogels have demonstrated significant promise in biomedical applications such as drug delivery and tissue engineering. A continued direction in hydrogel development includes the engineering of the biological responsiveness of these materials, via the inclusion of cell-binding domains and enzyme-sensitive domains. Ligand-receptor interactions offer additional opportunities in the design of responsive hydrogels, and strategies employing protein- polysaccharide interactions as a target may have unique relevance to materials intended to mimic the extracellular matrix (ECM). Accordingly, we have developed approaches for producing hydrogels via noncovalent interactions between heparin and heparin-binding peptides/proteins, and have demonstrated that such matrices are capable of both passive and receptor-mediated growth factor delivery. Further modification of these materials via the integration of these noncovalent strategies with chemical crosslinking methods will expand the range of their potential use and is under exploration. The combination of these approaches offers broad opportunities for the production of responsive matrices for biomedical applications.
Collapse
Affiliation(s)
- Kristi L Kiick
- University of Delaware, Department of Materials Science & Engineering, 201 DuPont Hall, Newark, DE 19716 and the Delaware Biotechnology Institute, 15 Innovation Way, Newark DE 19711, USA
| |
Collapse
|
35
|
Szaflik JP, Wysocki T, Kowalski M, Majsterek I, Borucka AI, Blasiak J, Szaflik J. An association between vascular endothelial growth factor gene promoter polymorphisms and diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2007; 246:39-43. [PMID: 17849138 DOI: 10.1007/s00417-007-0674-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 10/07/2006] [Accepted: 10/15/2006] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Diabetic retinopathy is a highly prevalent cause of visual loss in Western countries. Vascular endothelial growth factor (VEGF) is a potent angiogenic factor implicated in the development of the proliferative stage of this disease. Reports have suggested that polymorphisms at positions -460 and -634 of the 5' untranslated region of the VEGF gene increase its basal promoter activity. METHODS To investigate whether polymorphisms are associated with diabetic retinopathy, 215 patients with type 2 diabetes mellitus (T2DM) were enrolled. Among them, 82 subjects had proliferative diabetic retinopathy (PDR), 72 had non-proliferative diabetic retinopathy (NPDR), and 61 individuals without retinopathy served as controls. Two polymorphisms of the VEGF gene, a G-->C transversion at -634 (the G/C polymorphism) and a C-->T transition at -460 (the C/T polymorphism), were investigated by restriction fragment length polymorphism PCR and allele-specific PCR respectively. RESULTS We did not find any association between the C/T polymorphism and diabetic retinopathy. However, the G/C polymorphism genotype distribution and the frequency of the C allele were significantly higher in the NPDR group than in control patients (OR = 1.69, 95% CI = 1.03-2.79). Analysis of the distribution of combined genotypes of the VEGF gene revealed the prevalence of the C/C-C/C genotype in NPDR patients (OR = 8.26, 95% CI = 1.79-37.99) and C/G-CC in PDR patients (OR = 3.36, 95% CI = 1.39-8.12). CONCLUSIONS Occurrence of the -634C allele appears to be associated with increased VEGF gene promoter activity, and the G/C polymorphism might serve as a predictive factor for the development of diabetic retinopathy.
Collapse
Affiliation(s)
- Jacek P Szaflik
- Department of Ophthalmology, Medical University of Warsaw, Warsaw, Poland.
| | | | | | | | | | | | | |
Collapse
|
36
|
Yamaguchi N, Zhang L, Chae BS, Palla CS, Furst EM, Kiick KL. Growth factor mediated assembly of cell receptor-responsive hydrogels. J Am Chem Soc 2007; 129:3040-1. [PMID: 17315874 PMCID: PMC2606044 DOI: 10.1021/ja0680358] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nori Yamaguchi
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, USA
| | | | | | | | | | | |
Collapse
|
37
|
Kilic Ü, Kilic E, Järve A, Guo Z, Spudich A, Bieber K, Barzena U, Bassetti CL, Marti HH, Hermann DM. Human vascular endothelial growth factor protects axotomized retinal ganglion cells in vivo by activating ERK-1/2 and Akt pathways. J Neurosci 2006; 26:12439-46. [PMID: 17135405 PMCID: PMC6674905 DOI: 10.1523/jneurosci.0434-06.2006] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Based on its trophic effects on neurons and vascular cells, vascular endothelial growth factor (VEGF) is a promising candidate for the treatment of neurodegenerative diseases. To evaluate the therapeutic potential of VEGF, we here examined effects of this growth factor on the degeneration of axotomized retinal ganglion cells (RGCs), which, as CNS-derived neurons, offer themselves in an excellent way to study neuroprotection in vivo. Making use of a transgenic mouse line that constitutively expresses human VEGF under a neuron-specific enolase promoter, we show that (1) the VEGF-transgenic retina overexpresses human VEGF, (2) RGCs carry the VEGF receptor-2, and (3) vascular networks in normal and axotomized VEGF-transgenic (tg) retinas do not differ from control animals. After axotomy, RGCs of VEGF-tg mice were protected against delayed degeneration, as compared with wild-type littermates. Western blots revealed increased phosphorylated ERK-1/2 and Akt and reduced phosphorylated p38 and activated caspase-3 levels in axotomized VEGF-transgenic retinas. Intravitreous injections of pharmacological ERK-1/2 (PD98059) or Akt (LY294002) inhibitors showed that VEGF exerts neuroprotection by dual activation of ERK-1/2 and Akt pathways. In view that axotomy-induced RGC death occurs slowly and considering that RGCs are CNS-derived neurons, we predict the clinical implementation of VEGF in neurodegenerative diseases of both brain and retina.
Collapse
Affiliation(s)
- Ülkan Kilic
- Department of Neurology, University Hospital Zurich, CH-8091 Zurich, Switzerland, and
| | - Ertugrul Kilic
- Department of Neurology, University Hospital Zurich, CH-8091 Zurich, Switzerland, and
| | - Anne Järve
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Zeyun Guo
- Department of Neurology, University Hospital Zurich, CH-8091 Zurich, Switzerland, and
| | - Annett Spudich
- Department of Neurology, University Hospital Zurich, CH-8091 Zurich, Switzerland, and
| | - Katja Bieber
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Uxue Barzena
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Claudio L. Bassetti
- Department of Neurology, University Hospital Zurich, CH-8091 Zurich, Switzerland, and
| | - Hugo H. Marti
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Dirk M. Hermann
- Department of Neurology, University Hospital Zurich, CH-8091 Zurich, Switzerland, and
| |
Collapse
|