1
|
Tierno IS, Agarwal M, Matisioudis N, Chandrakumar S, Ghosh K. Stiffness Measurement of Retinal Capillaries and Subendothelial Matrix using Atomic Force Microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582372. [PMID: 38464329 PMCID: PMC10925338 DOI: 10.1101/2024.02.28.582372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Retinal capillary degeneration is a clinical hallmark of the early stages of diabetic retinopathy (DR). Our recent studies have revealed that diabetes-induced increase in retinal capillary stiffness plays a crucial and previously unrecognized causal role in inflammation-mediated degeneration of retinal capillaries. Retinal capillary stiffening results from overexpression of lysyl oxidase, an enzyme that crosslinks and stiffens the subendothelial matrix. Since tackling DR at the early stage is expected to prevent or slow down DR progression and associated vision loss, subendothelial matrix and capillary stiffness represent relevant and novel therapeutic targets for early DR management. Further, direct measurement of retinal capillary stiffness can serve as a crucial preclinical validation step for the development of new imaging techniques for non-invasive assessment of retinal capillary stiffness in animal and human subjects. With this view in mind, we here provide a detailed protocol for the isolation and stiffness measurement of mouse retinal capillaries and retinal subendothelial matrix using atomic force microscopy.
Collapse
Affiliation(s)
- Irene Santiago Tierno
- Department of Ophthalmology, University of California, Los Angeles, CA, USA
- Doheny Eye Institute, Pasadena, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental PhD Program, University of California, Los Angeles, CA, USA
| | - Mahesh Agarwal
- Department of Ophthalmology, University of California, Los Angeles, CA, USA
- Doheny Eye Institute, Pasadena, CA, USA
| | | | - Sathishkumar Chandrakumar
- Department of Ophthalmology, University of California, Los Angeles, CA, USA
- Doheny Eye Institute, Pasadena, CA, USA
| | - Kaustabh Ghosh
- Department of Ophthalmology, University of California, Los Angeles, CA, USA
- Doheny Eye Institute, Pasadena, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental PhD Program, University of California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Li Y, Kazlauskas A. An Assay to Detect Protection of the Retinal Vasculature from Diabetes-Related Death in Mice. J Vis Exp 2024:10.3791/66123. [PMID: 38284520 PMCID: PMC11282432 DOI: 10.3791/66123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024] Open
Abstract
Diabetic retinopathy (DR) is a complex and progressive ocular disease characterized by two distinct phases in its pathogenesis. The first phase involves the loss of protection from diabetes-induced damage to the retina, while the second phase centers on the accumulation of this damage. Traditional assays primarily focus on evaluating capillary degeneration, which is indicative of the severity of damage, essentially addressing the second phase of DR. However, they only indirectly provide insights into whether the protective mechanisms of the retinal vasculature have been compromised. To address this limitation, a novel approach was developed to directly assess the retina's protective mechanisms - specifically, its resilience against diabetes-induced insults like oxidative stress and cytokines. This protection assay, although initially designed for diabetic retinopathy, holds the potential for broader applications in both physiological and pathological contexts. In summary, understanding the pathogenesis of diabetic retinopathy involves recognizing the dual phases of protection loss and damage accumulation, with this innovative protection assay offering a valuable tool for research and potentially extending to other medical conditions.
Collapse
Affiliation(s)
- Yanliang Li
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago
| | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago; Department of Physiology and Biophysics, University of Illinois at Chicago;
| |
Collapse
|
3
|
Yuan T, Dong L, Pearsall EA, Zhou K, Cheng R, Ma JX. The Protective Role of Microglial PPARα in Diabetic Retinal Neurodegeneration and Neurovascular Dysfunction. Cells 2022; 11:cells11233869. [PMID: 36497130 PMCID: PMC9739170 DOI: 10.3390/cells11233869] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Microglial activation and subsequent pathological neuroinflammation contribute to diabetic retinopathy (DR). However, the underlying mechanisms of microgliosis, and means to effectively suppress pathological microgliosis, remain incompletely understood. Peroxisome proliferator-activated receptor alpha (PPARα) is a transcription factor that regulates lipid metabolism. The present study aimed to determine if PPARα affects pathological microgliosis in DR. In global Pparα mice, retinal microglia exhibited decreased structural complexity and enlarged cell bodies, suggesting microglial activation. Microglia-specific conditional Pparα-/- (PCKO) mice showed decreased retinal thickness as revealed by optical coherence tomography. Under streptozotocin (STZ)-induced diabetes, diabetic PCKO mice exhibited decreased electroretinography response, while diabetes-induced retinal dysfunction was alleviated in diabetic microglia-specific Pparα-transgenic (PCTG) mice. Additionally, diabetes-induced retinal pericyte loss was exacerbated in diabetic PCKO mice and alleviated in diabetic PCTG mice. In cultured microglial cells with the diabetic stressor 4-HNE, metabolic flux analysis demonstrated that Pparα ablation caused a metabolic shift from oxidative phosphorylation to glycolysis. Pparα deficiency also increased microglial STING and TNF-α expression. Taken together, these findings revealed a critical role for PPARα in pathological microgliosis, neurodegeneration, and vascular damage in DR, providing insight into the underlying molecular mechanisms of microgliosis in this context and suggesting microglial PPARα as a potential therapeutic target.
Collapse
Affiliation(s)
- Tian Yuan
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Lijie Dong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Elizabeth A. Pearsall
- Vision Research Center, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Kelu Zhou
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Rui Cheng
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
- Correspondence: (R.C.); (J.-X.M.); Tel.: +1-336-716-3914 (R.C.); +1-336-716-4676 (J.-X.M.)
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
- Correspondence: (R.C.); (J.-X.M.); Tel.: +1-336-716-3914 (R.C.); +1-336-716-4676 (J.-X.M.)
| |
Collapse
|
4
|
Lee YR, Son M, Kim YS, Kim JS, Kim CH, Jung SH. Evaluation of a Rapid and Simple Method for Assessing Retinal Vessel Structures in Adult Zebrafish. Int J Mol Sci 2022; 23:ijms232315069. [PMID: 36499406 PMCID: PMC9739661 DOI: 10.3390/ijms232315069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
The evaluation of retinal vascular structures is important for analyzing various ophthalmic diseases. Conventional trypsin digestion was used for separating retinal vasculatures in mouse, rat, and other animal models; however, the trypsin method alone is technically difficult to perform and has not been reported in zebrafish to date. In this study, we introduced a rapid and convenient method that allows the investigation of fine vessel structures at a cellular level in the relatively intact retinal vasculature of adult zebrafish. Using an anti-ZO-1 antibody, tight junction structures in retinal vessels were examined in detail and several different cell types constituting blood vessels in arterial and capillary areas were identified. In addition, using cell type-specific antibodies, we identified smooth muscle cells, blood cells, and endothelial cells in the retinal vasculature. Finally, using the hyperglycemic model, we observed the dilation of retinal vessels, the downregulation of tight junction proteins, and the reduction in smooth muscle cells. Based on these results, we provide a rapid and convenient method for the study of retinal vasculature disease in the zebrafish animal model.
Collapse
Affiliation(s)
- Yu-Ri Lee
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
- Department of Biology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Myeongjoo Son
- Research Institute for Aerospace Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Young Sook Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
| | - Jin Sook Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
- Julia Laboratory, Suwon 16232, Republic of Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Republic of Korea
- Correspondence: (C.-H.K.); (S.-H.J.)
| | - Seung-Hyun Jung
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Republic of Korea
- Correspondence: (C.-H.K.); (S.-H.J.)
| |
Collapse
|
5
|
Balaratnasingam C, An D, Hein M, Yu P, Yu DY. Studies of the retinal microcirculation using human donor eyes and high-resolution clinical imaging: Insights gained to guide future research in diabetic retinopathy. Prog Retin Eye Res 2022; 94:101134. [PMID: 37154065 DOI: 10.1016/j.preteyeres.2022.101134] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The microcirculation plays a key role in delivering oxygen to and removing metabolic wastes from energy-intensive retinal neurons. Microvascular changes are a hallmark feature of diabetic retinopathy (DR), a major cause of irreversible vision loss globally. Early investigators have performed landmark studies characterising the pathologic manifestations of DR. Previous works have collectively informed us of the clinical stages of DR and the retinal manifestations associated with devastating vision loss. Since these reports, major advancements in histologic techniques coupled with three-dimensional image processing has facilitated a deeper understanding of the structural characteristics in the healthy and diseased retinal circulation. Furthermore, breakthroughs in high-resolution retinal imaging have facilitated clinical translation of histologic knowledge to detect and monitor progression of microcirculatory disturbances with greater precision. Isolated perfusion techniques have been applied to human donor eyes to further our understanding of the cytoarchitectural characteristics of the normal human retinal circulation as well as provide novel insights into the pathophysiology of DR. Histology has been used to validate emerging in vivo retinal imaging techniques such as optical coherence tomography angiography. This report provides an overview of our research on the human retinal microcirculation in the context of the current ophthalmic literature. We commence by proposing a standardised histologic lexicon for characterising the human retinal microcirculation and subsequently discuss the pathophysiologic mechanisms underlying key manifestations of DR, with a focus on microaneurysms and retinal ischaemia. The advantages and limitations of current retinal imaging modalities as determined using histologic validation are also presented. We conclude with an overview of the implications of our research and provide a perspective on future directions in DR research.
Collapse
Affiliation(s)
- Chandrakumar Balaratnasingam
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia; Department of Ophthalmology, Sir Charles Gairdner Hospital, Western Australia, Australia.
| | - Dong An
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Martin Hein
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Paula Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Dao-Yi Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| |
Collapse
|
6
|
Zerbini G, Maestroni S, Leocani L, Mosca A, Godi M, Paleari R, Belvedere A, Gabellini D, Tirassa P, Castoldi V, Viganò I, Galbiati S, Turco V, Lambiase A, Rama P. Topical nerve growth factor prevents neurodegenerative and vascular stages of diabetic retinopathy. Front Pharmacol 2022; 13:1015522. [PMID: 36172176 PMCID: PMC9510636 DOI: 10.3389/fphar.2022.1015522] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Specific and effective preventive treatment for diabetic retinopathy (DR) is presently unavailable, mostly because the early stages of the complication have been, until recently, poorly understood. The recent demonstration that the vascular phase of DR is preceded and possibly caused by the neurodegeneration of retinal ganglion cells suggests that DR could, at least theoretically, be prevented through an early neuroprotective approach. The aims of our study were to clarify the natural history of diabetes-driven retinal neurodegeneration and to verify the possibility to prevent DR using topical nerve growth factor (NGF). The results of the study show that retinal neurodegeneration, characterized by the loss of retinal ganglion cells represents a relatively early phenomenon of diabetes (between 5 and 16 weeks of age), which tends to be self-limiting in the long run. Neurodegeneration is followed by the development of DR-related vascular dysfunctions, as confirmed by the development of acellular capillaries and the loss of retinal pericytes. Both retinal neurodegeneration and subsequent vascular dysfunction can be successfully prevented by topical NGF administration. These findings suggest that: 1) The first stage of DR consists in a self-limiting retinal neurodegeneration 2) The demonstrated effectiveness of topical NGF in the prevention of DR could be rapidly translated into clinical practice.
Collapse
Affiliation(s)
- Gianpaolo Zerbini
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- *Correspondence: Gianpaolo Zerbini,
| | - Silvia Maestroni
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Letizia Leocani
- Experimental Neurophysiology Unit, INSPE-Institute of Experimental Neurology, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Mosca
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti and Centro per la Riferibilità Metrologica in Medicina di Laboratorio (CIRME), Università degli Studi di Milano, Milano, Italy
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche (ITB-CNR), Milano, Italy
| | - Michela Godi
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Renata Paleari
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti and Centro per la Riferibilità Metrologica in Medicina di Laboratorio (CIRME), Università degli Studi di Milano, Milano, Italy
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche (ITB-CNR), Milano, Italy
| | - Arianna Belvedere
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Daniela Gabellini
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Paola Tirassa
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Valerio Castoldi
- Experimental Neurophysiology Unit, INSPE-Institute of Experimental Neurology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ilaria Viganò
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Silvia Galbiati
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valentina Turco
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Paolo Rama
- Vita-Salute San Raffaele University, Milan, Italy
- Cornea and Ocular Surface Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
7
|
mTOR inhibition as a novel gene therapeutic strategy for diabetic retinopathy. PLoS One 2022; 17:e0269951. [PMID: 35709240 PMCID: PMC9202865 DOI: 10.1371/journal.pone.0269951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
In addition to laser photocoagulation, therapeutic interventions for diabetic retinopathy (DR) have heretofore consisted of anti-VEGF drugs, which, besides drawbacks inherent to the treatments themselves, are limited in scope and may not fully address the condition’s complex pathophysiology. This is because DR is a multifactorial condition, meaning a gene therapy focused on a target with broader effects, such as the mechanistic target of rapamycin (mTOR), may prove to be the solution in overcoming these concerns. Having previously demonstrated the potential of a mTOR-inhibiting shRNA packaged in a recombinant adeno-associated virus to address a variety of angiogenic retinal diseases, here we explore the effects of rAAV2-shmTOR-SD in a streptozotocin-induced diabetic mouse model. Delivered via intravitreal injection, the therapeutic efficacy of the virus vector upon early DR processes was examined. rAAV2-shmTOR-SD effectively transduced mouse retinas and therein downregulated mTOR expression, which was elevated in sham-treated and control shRNA-injected (rAAV2-shCon-SD) control groups. mTOR inhibition additionally led to marked reductions in pericyte loss, acellular capillary formation, vascular permeability, and retinal cell layer thinning, processes that contribute to DR progression. Immunohistochemistry showed that rAAV2-shmTOR-SD decreased ganglion cell loss and pathogenic Müller cell activation and proliferation, while also having anti-apoptotic activity, with these effects suggesting the therapeutic virus vector may be neuroprotective. Taken together, these results build upon our previous work to demonstrate the broad ability of rAAV2-shmTOR-SD to address aspects of DR pathophysiology further evidencing its potential as a human gene therapeutic strategy for DR.
Collapse
|
8
|
Kajtna J, Tsang SH, Koch SF. Late-stage rescue of visually guided behavior in the context of a significantly remodeled retinitis pigmentosa mouse model. Cell Mol Life Sci 2022; 79:148. [PMID: 35195763 PMCID: PMC8866266 DOI: 10.1007/s00018-022-04161-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
Patients with progressive neurodegenerative disorder retinitis pigmentosa (RP) are diagnosed in the midst of ongoing retinal degeneration and remodeling. Here, we used a Pde6b-deficient RP gene therapy mouse model to test whether treatment at late disease stages can halt photoreceptor degeneration and degradative remodeling, while sustaining constructive remodeling and restoring function. We demonstrated that when fewer than 13% of rods remain, our genetic rescue halts photoreceptor degeneration, electroretinography (ERG) functional decline and inner retinal remodeling. In addition, in a water maze test, the performance of mice treated at 16 weeks of age or earlier was indistinguishable from wild type. In contrast, no efficacy was apparent in mice treated at 24 weeks of age, suggesting the photoreceptors had reached a point of no return. Further, remodeling in the retinal pigment epithelium (RPE) and retinal vasculature was not halted at 16 or 24 weeks of age, although there appeared to be some slowing of blood vessel degradation. These data suggest a novel working model in which restoration of clinically significant visual function requires only modest threshold numbers of resilient photoreceptors, halting of destructive remodeling and sustained constructive remodeling. These novel findings define the potential and limitations of RP treatment and suggest possible nonphotoreceptor targets for gene therapy optimization.
Collapse
Affiliation(s)
- Jacqueline Kajtna
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
- Physiological Genomics, BioMedical Center, Ludwig-Maximilians-Universität München, Planegg/Martinsried, Germany
| | - Stephen H Tsang
- Jonas Children's Vision Care, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, 10032, USA
| | - Susanne F Koch
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
- Physiological Genomics, BioMedical Center, Ludwig-Maximilians-Universität München, Planegg/Martinsried, Germany.
| |
Collapse
|
9
|
Cheng Y, Peng L, Deng X, Li T, Guo H, Xu C, Fang T, Liu X, Sun B, Chen L. Prostaglandin F2α protects against pericyte apoptosis by inhibiting the PI3K/Akt/GSK3β/β-catenin signaling pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1021. [PMID: 34277821 PMCID: PMC8267281 DOI: 10.21037/atm-21-2717] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/17/2021] [Indexed: 11/30/2022]
Abstract
Background Diabetic retinopathy (DR) is the most common microvascular complication of diabetes and the main cause of non-traumatic blindness in adults. Pericyte loss is known to be an early pathological change of DR. Our group’s previous research indicated that prostaglandin F2α (PGF2α) acts as an eicosanoidal protector against non-proliferative DR that can regulate the mobility of pericytes in a RhoA-mediated manner. However, the effect of PGF2α on pericyte apoptosis has yet to be described. Methods Two animal models were constructed: a high-fat diet (HFD) and streptozotocin (STZ)-induced type 2 diabetes mouse model and a spontaneous type 2 diabetes db/db mouse model. We analyzed pathological changes, and performed TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling) staining and western blot to detect apoptosis in the retinas of diabetic mice. For our in vitro experiments, we selected human retinal pericytes and subjected them to high-glucose (HG), PGF2α, and AL8810 (an antagonist of the PGF2α receptor) treatment. Subsequently, apoptosis and the levels of PI3K/Akt/GSK3β/β-catenin pathway-related proteins were detected by TUNEL staining and western blot, respectively. Results The levels of apoptosis were increased in the retinas of diabetic mice in both T2DM models. In vitro, HG treatment increased apoptosis and inhibited PI3K/Akt/GSK3β/β-catenin signaling in pericytes. In contrast, PGF2α treatment inhibited pericyte apoptosis while increasing the levels of the PI3K, p-Akt/t-Akt, p-GSK3β/t-GSK3β, and β-catenin proteins; however, these PGF2α-induced effects were eliminated by ALL80. Conclusions PGF2α may make a key contribution to reducing pericyte apoptosis and protecting against DR via its inhibition of the PI3K/Akt/GSK3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Ying Cheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Liyuan Peng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaoqing Deng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Ting Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Hang Guo
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Chaofei Xu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Ting Fang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaohuan Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Fan S, Yang Z, Liu Y, Zhong J, Zhang S, Xiao Y, Liu X, Yi W, He C, Hu Y, Liu X. Extensive Sub-RPE Complement Deposition in a Nonhuman Primate Model of Early-Stage Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2021; 62:30. [PMID: 33749721 PMCID: PMC7991921 DOI: 10.1167/iovs.62.3.30] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose This study aims to reveal retinal abnormities in a spontaneous diabetic nonhuman primate model and explore the mechanism of featured injuries. Methods Twenty-eight cynomolgus monkeys were identified to suffer from spontaneous type 2 diabetes from a colony of more than eight-hundred aged monkeys, and twenty-six age-matched ones were chosen as controls. Their blood biochemistry profiles were determined and retinal changes were examined by multimodal imaging, hematoxylin and eosin staining, and immunofluorescence. Retinal pigment epithelium (RPE) cells were further investigated by RNA sequencing and computational analyses. Results These diabetic monkeys were characterized by early retinal vascular and neural damage and dyslipidemia. The typical acellular capillaries and pericyte ghost were found in the diabetic retina, which also exhibited reduced retinal nerve fiber layer thickness compared to controls (all P < 0.05). Of note, distinct sub-RPE drusenoid lesions were extensively observed in these diabetic monkeys (46.43% vs. 7.69%), and complements including C3 and C5b-9 were deposited in these lesions. RNA-seq analysis revealed complement activation, AGE/RAGE activation and inflammatory response in diabetic RPE cells. Consistently, the plasma C3 and C4 were particularly increased in the diabetic monkeys with drusenoid lesions (P = 0.028 and 0.029). Conclusions The spontaneous type 2 diabetic monkeys featured with early-stage retinopathy including not only typical vascular and neural damage but also a distinct sub-RPE deposition. The complement activation of RPE cells in response to hyperglycemia might contribute to the deposition, revealing an unrecognized role of RPE cells in the early-stage pathological process of diabetic retinopathy.
Collapse
Affiliation(s)
- Shuxin Fan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ziqi Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiawei Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shuyao Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuhua Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wei Yi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Youjin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Jiang H, Zhang H, Jiang X, Wu S. Overexpression of D-amino acid oxidase prevents retinal neurovascular pathologies in diabetic rats. Diabetologia 2021; 64:693-706. [PMID: 33319325 DOI: 10.1007/s00125-020-05333-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/06/2020] [Indexed: 10/22/2022]
Abstract
AIMS/HYPOTHESIS Diabetic retinopathy is characterised by retinal neurodegeneration and retinal vascular abnormalities, affecting one third of diabetic patients with disease duration of more than 10 years. Accumulated evidence suggests that serine racemase (SR) and D-serine are correlated with the pathogenesis of diabetic retinopathy and the deletion of the Srr gene reverses neurovascular pathologies in diabetic mice. Since D-serine content is balanced by SR synthesis and D-amino acid oxidase (DAAO) degradation, we examined the roles of DAAO in diabetic retinopathy and further explored relevant therapy. METHODS Rats were used as a model of diabetes by i.p. injection of streptozotocin at the age of 2 months and blood glucose was monitored with a glucometer. Quantitative real-time PCR was used to examine Dao mRNA and western blotting to examine targeted proteins in the retinas. Bisulphite sequencing was used to examine the methylation of Dao mRNA promoter in the retinas. Intravitreal injection of DAAO-expressing adenovirus (AAV8-DAAO) was conducted one week before streptozotocin administration. Brain specific homeobox/POU domain protein 3a (Brn3a) immunofluorescence was conducted to indicate retinal ganglion cells at 3 months after virus injection. The permeability of the blood-retinal barrier was examined by Evans blue leakage from retinal capillaries. Periodic acid-Schiff staining and haematoxylin counterstaining were used to indicate retinal vasculature, which was further examined with double immunostaining at 7 months after virus injection. RESULTS At the age of 12 months, DAAO mRNA and protein levels in retinas from diabetic animals were reduced to 66.2% and 70.4% of those from normal (control) animals, respectively. The Dao proximal promoter contained higher levels of methylation in diabetic than in normal retinas. Consistent with the observation, DNA methyltransferase 1 was increased in diabetic retinas. Injection of DAAO-expressing virus completely prevented the loss of retinal ganglion cells and the disruption of blood-retinal barrier in diabetic rats. Diabetic retinas contained retinal ganglion cells at a density of 54 ± 4/mm2, which was restored to 68 ± 9/mm2 by DAAO overexpression, similar to the levels in normal retinas. The ratio between the number of endothelial cells and pericytes in diabetic retinas was 6.06 ± 1.93/mm2, which was reduced to 3.42 ± 0.55/mm2 by DAAO overexpression; the number of acellular capillaries in diabetic retinas was 10 ± 5/mm2, which was restored to 6 ± 2/mm2 by DAAO overexpression, similar to the levels in normal retinas. Injection of the DAAO-expressing virus increased the expression of occludin and reduced gliosis, which were examined to probe the mechanism by which the disrupted blood-retinal barrier in diabetic rats was rescued and retinal neurodegeneration was prevented. CONCLUSIONS/INTERPRETATION Altogether, overexpression of DAAO before the onset of diabetes protects against neurovascular abnormalities in retinas from diabetic rats, which suggests a novel strategy for preventing diabetic retinopathy. Graphical abstract.
Collapse
Affiliation(s)
- Haiyan Jiang
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
- State Key Laboratory of Optometry, Ophthalmology, and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - He Zhang
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
- State Key Laboratory of Optometry, Ophthalmology, and Visual Science, Wenzhou, Zhejiang, People's Republic of China
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, Zhejiang, People's Republic of China
| | - Xue Jiang
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
- State Key Laboratory of Optometry, Ophthalmology, and Visual Science, Wenzhou, Zhejiang, People's Republic of China
| | - Shengzhou Wu
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
- State Key Laboratory of Optometry, Ophthalmology, and Visual Science, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
12
|
Saghiri MA, Suscha A, Wang S, Saghiri AM, Sorenson CM, Sheibani N. Noninvasive temporal detection of early retinal vascular changes during diabetes. Sci Rep 2020; 10:17370. [PMID: 33060607 PMCID: PMC7567079 DOI: 10.1038/s41598-020-73486-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetes associated complications, including diabetic retinopathy and loss of vision, are major health concerns. Detecting early retinal vascular changes during diabetes is not well documented, and only few studies have addressed this domain. The purpose of this study was to noninvasively evaluate temporal changes in retinal vasculature at very early stages of diabetes using fundus images from preclinical models of diabetes. Non-diabetic and Akita/+ male mice with different duration of diabetes were subjected to fundus imaging using a Micron III imaging system. The images were obtained from 4 weeks- (onset of diabetes), 8 weeks-, 16 weeks-, and 24 weeks-old male Akita/+ and non-diabetic mice. In total 104 fundus images were subjected to analysis for various feature extractions. A combination of Canny Edge Detector and Angiogenesis Analyzer plug-ins in ImageJ were utilized to quantify various retinal vascular changes in fundus images. Statistical analyses were conducted to determine significant differences in the various extracted features from fundus images of diabetic and non-diabetic animals. Our novel image analysis method led to extraction of over 20 features. These results indicated that some of these features were significantly changed with a short duration of diabetes, and others remained the same but changed after longer duration of diabetes. These patterns likely distinguish acute (protective) and chronic (damaging) associated changes with diabetes. We show that with a combination of various plugging one can extract over 20 features from retinal vasculature fundus images. These features change during diabetes, thus allowing the quantification of quality of retinal vascular architecture as biomarkers for disease progression. In addition, our method was able to identify unique differences among diabetic mice with different duration of diabetes. The ability to noninvasively detect temporal retinal vascular changes during diabetes could lead to identification of specific markers important in the development and progression of diabetes mediated-microvascular changes, evaluation of therapeutic interventions, and eventual reversal of these changes in order to stop or delay disease progression.
Collapse
Affiliation(s)
- Mohammad Ali Saghiri
- Director of Biomaterial and Prosthodontic Laboratory, Department of Restorative Dentistry, Rutgers School of Dental Medicine, Rutgers Biomedical and Health Sciences, MSB C639A, 185 South Orange Avenue, Newark, NJ, 07103, USA.
- Department of Endodontics, University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA, USA.
| | - Andrew Suscha
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Shoujian Wang
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | | | - Christine M Sorenson
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Department of Biomedical Engineering, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
13
|
Mazzoli V, Zhong LH, Dang VT, Shi Y, Werstuck GH. Characterization of Retinal Microvascular Complications and the Effects of Endoplasmic Reticulum Stress in Mouse Models of Diabetic Atherosclerosis. Invest Ophthalmol Vis Sci 2020; 61:49. [PMID: 32852545 PMCID: PMC7452854 DOI: 10.1167/iovs.61.10.49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/27/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Recent evidence suggests that there is a correlation between the micro- and macrovascular complications of diabetes mellitus. The aim of this study is to investigate the molecular mechanisms by which diabetes promotes the development of microvascular disease (diabetic retinopathy [DR]) through characterization of the effects of hyperglycemia in the retina of mouse models of diabetic atherosclerosis. Methods Hyperglycemia was induced in apolipoprotein E-deficient (ApoE-/-) mice, a model of accelerated atherosclerosis, either through streptozotocin (STZ) injection or introduction of the Ins2Akita mutation (ApoE-/-Ins2+/Akita). Another subset of ApoE-/- mice was supplemented with glucosamine (GlcN). To attenuate atherosclerosis, subsets of mice from each experimental group were treated with the chemical chaperone, 4-phenylbutyric acid (4PBA). Eyes from 15-week-old mice were either trypsin digested and stained with periodic acid-Schiff (PAS) or frozen for cryostat sectioning and immunostained for endoplasmic reticulum (ER) stress markers, including C/EBP homologous protein (CHOP) and 78-kDa glucose-regulated protein (GRP78). PAS-stained retinal flatmounts were analyzed for microvessel density, acellular capillaries, and pericyte ghosts. Results Features of DR, including pericyte ghosts and reduced microvessel density, were observed in hyperglycemic and GlcN-supplemented mice. Treatment with 4PBA reduced ER stress in the retinal periphery and attenuated DR in the experimental groups. Conclusions Mouse models of diabetic atherosclerosis show characteristic pathologies of DR that correlate with atherosclerosis. The increased magnitude of these changes and responses to 4PBA in the peripheral retina suggest that future studies should be aimed at assessing regional differences in mechanisms of ER stress-related pathways in these mouse models.
Collapse
Affiliation(s)
- Vienna Mazzoli
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Lexy H. Zhong
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Vi T. Dang
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Yuanyuan Shi
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Geoff H. Werstuck
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
14
|
Liu Y, Zheng Y, Zhou Y, Liu Y, Xie M, Meng W, An M. The expression and significance of mTORC1 in diabetic retinopathy. BMC Ophthalmol 2020; 20:297. [PMID: 32689970 PMCID: PMC7370483 DOI: 10.1186/s12886-020-01553-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 07/06/2020] [Indexed: 01/23/2023] Open
Abstract
Background To investigate the expression and significance of mechanistic target of rapamycin complex 1(mTORC1) in diabetic retinopathy (DR), and to find new targets and new methods for the treatment of DR. Methods A DR rat model was prepared by general feeding combined with intraperitoneal injection of 10% streptozotocin (60 mg/kg). The rats were randomly divided into a control group (NDM group) and a diabetes group (DM group). Three months later, the degrees of retinopathy was determined using hematoxylin and eosin staining, and the levels of p-S6, VEGF, and PEDF proteins were detected by immunohistochemistry and western blotting. Human retinal capillary endothelial cells (HRCECs) were cultured in high glucose (HG) conditions, then treated with rapamycin or transfected with siTSC1.The protein levels of p-S6 were assessed by western blotting. The 5-ethynyl-2′-deoxyuridine assay was used to detect cell proliferation, and the Transwell assay was used to detect cell migration. Results A DM rat model was successfully developed. The expressions of p-S6 and VEGF proteins were significantly increased in the DM group (p < 0.05), and the expression of PEDF protein was significantly decreased compared with the NDM group (p < 0.05). In vitro, the p-S6 protein, as well as cell proliferation and migration, in HG induced HRCECs were increased (p < 0.05) compared with the control (normal glucose) group (p < 0.05). After transfection with siTSC1 to activate mTORC1, the expression of p-S6, as well as cell proliferation and migration, were increased. In contrast, rapamycin decreased p-S6 expression, as well as proliferation and migration, in HG induced HRCECs compared to the control group (p < 0.05). Conclusion mTORC1 plays an important role in DR. After activation, mTORC1 induced expression of the p-S6 protein, regulated the expressions of VEGF and PEDF proteins, and changed the proliferation and migration of endothelial cells. The mTORC1 can therefore be used as a new target,as well as in the treatment of DR.
Collapse
Affiliation(s)
- Yanli Liu
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diesases, Guangzhou, China
| | - Yarong Zheng
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diesases, Guangzhou, China
| | - Yekai Zhou
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diesases, Guangzhou, China
| | - Yi Liu
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diesases, Guangzhou, China
| | - Mengxuan Xie
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diesases, Guangzhou, China
| | - Wenjing Meng
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diesases, Guangzhou, China
| | - Meixia An
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diesases, Guangzhou, China.
| |
Collapse
|
15
|
Zhu K, Hu X, Chen H, Li F, Yin N, Liu AL, Shan K, Qin YW, Huang X, Chang Q, Xu GZ, Wang Z. Downregulation of circRNA DMNT3B contributes to diabetic retinal vascular dysfunction through targeting miR-20b-5p and BAMBI. EBioMedicine 2019; 49:341-353. [PMID: 31636010 PMCID: PMC6945224 DOI: 10.1016/j.ebiom.2019.10.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/25/2019] [Accepted: 10/02/2019] [Indexed: 12/16/2022] Open
Abstract
Background Diabetic retinopathy, a vascular complication of diabetes mellitus, is the leading cause of visual impairment and blindness. circRNAs act as competing endogenous RNA, sponging target miRNA and thus influencing mRNA expression in vascular diseases. We investigated whether and how circDNMT3B is involved in retinal vascular dysfunction under diabetic conditions. Methods qRT-PCR was performed to detect expression of circDNMT3B, miR-20b-5p, and BAMBI in retinal microvascular endothelial cells under diabetic conditions. Western blot, Cell Counting Kit-8, Transwell, Matrigel tube formation, and retinal trypsin digestion assays were conducted to explore the roles of circDNMT3B/miR-20b-5p/BAMBI in retinal vascular dysfunction. Bioinformatics analysis and luciferase reporter, siRNA, and overexpression assays were used to reveal the mechanisms of the circDNMT3B/miR-20b-5p/BAMBI interaction. Electroretinograms were used to evaluate visual function. Findings Upregulation of miR-20b-5p under diabetic conditions promoted proliferation, migration, and tube formation of human retinal microvascular endothelial cells (HRMECs), which was mediated by downregulated BAMBI. Under diabetic conditions, circDNMT3B, which acts as a sponge of miR-20b-5p, is downregulated. circDNMT3B overexpression reduced retinal acellular capillary number and alleviated visual damage in diabetic rats. Changes in expression of circDNMT3B and miR-20b-5p were confirmed in the proliferative fibrovascular membranes of patients with diabetic retinopathy. Interpretation Downregulation of circDNMT3B contributes to vascular dysfunction in diabetic retinas through regulating miR-20b-5p and BAMBI, providing a potential treatment strategy for diabetic retinopathy. Funding National Natural Science Foundation of China, National Key Basic Research Program of China, Shanghai Municipal Science and Technology Major Project, and ZJLab.
Collapse
Affiliation(s)
- Ke Zhu
- Department of Ophthalmology and Vision Science, Eye and Ear Nose Throat Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200031, China
| | - Xin Hu
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Han Chen
- Department of Ophthalmology and Vision Science, Eye and Ear Nose Throat Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200031, China
| | - Fang Li
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Yin
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ai-Lin Liu
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Kun Shan
- Department of Ophthalmology and Vision Science, Eye and Ear Nose Throat Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200031, China
| | - Yao-Wu Qin
- Department of Ophthalmology and Vision Science, Eye and Ear Nose Throat Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200031, China
| | - Xin Huang
- Department of Ophthalmology and Vision Science, Eye and Ear Nose Throat Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200031, China
| | - Qing Chang
- Department of Ophthalmology and Vision Science, Eye and Ear Nose Throat Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200031, China
| | - Ge-Zhi Xu
- Department of Ophthalmology and Vision Science, Eye and Ear Nose Throat Hospital, Shanghai Key Laboratory of Visual Impairment and Restoration, NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200031, China.
| | - Zhongfeng Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
16
|
Toh H, Smolentsev A, Bozadjian RV, Keeley PW, Lockwood MD, Sadjadi R, Clegg DO, Blodi BA, Coffey PJ, Reese BE, Thomson JA. Vascular changes in diabetic retinopathy-a longitudinal study in the Nile rat. J Transl Med 2019; 99:1547-1560. [PMID: 31101854 PMCID: PMC6788790 DOI: 10.1038/s41374-019-0264-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/11/2019] [Accepted: 03/24/2019] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy is the most common microvascular complication of diabetes and is a major cause of blindness, but an understanding of the pathogenesis of the disease has been hampered by a lack of accurate animal models. Here, we explore the dynamics of retinal cellular changes in the Nile rat (Arvicanthis niloticus), a carbohydrate-sensitive model for type 2 diabetes. The early retinal changes in diabetic Nile rats included increased acellular capillaries and loss of pericytes that correlated linearly with the duration of diabetes. These vascular changes occurred in the presence of microglial infiltration but in the absence of retinal ganglion cell loss. After a prolonged duration of diabetes, the Nile rat also exhibits a spectrum of retinal lesions commonly seen in the human condition including vascular leakage, capillary non-perfusion, and neovascularization. Our longitudinal study documents a range and progression of retinal lesions in the diabetic Nile rat remarkably similar to those observed in human diabetic retinopathy, and suggests that this model will be valuable in identifying new therapeutic strategies.
Collapse
Affiliation(s)
- Huishi Toh
- Center for Stem Cell Biology and Engineering, University of California at Santa Barbara, Santa Barbara, CA, USA. .,Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, CA, USA.
| | - Alexander Smolentsev
- Center for Stem Cell Biology and Engineering, University of California at Santa Barbara, Santa Barbara, California, USA,Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA
| | - Rachel V. Bozadjian
- Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA
| | - Patrick W. Keeley
- Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA
| | - Madison D. Lockwood
- Center for Stem Cell Biology and Engineering, University of California at Santa Barbara, Santa Barbara, California, USA,Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA
| | - Ryan Sadjadi
- Center for Stem Cell Biology and Engineering, University of California at Santa Barbara, Santa Barbara, California, USA,Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA
| | - Dennis O. Clegg
- Center for Stem Cell Biology and Engineering, University of California at Santa Barbara, Santa Barbara, California, USA,Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, USA
| | - Barbara A. Blodi
- University of Wisconsin Fundus Photograph Reading Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Peter J. Coffey
- Center for Stem Cell Biology and Engineering, University of California at Santa Barbara, Santa Barbara, California, USA,Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA,NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust, UCL Institute of Ophthalmology, London, UK,The London Project to Cure Blindness, ORBIT, Institute of Ophthalmology, University College London (UCL), London, UK
| | - Benjamin E. Reese
- Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA,Department of Psychological and Brain Sciences, University of California at Santa Barbara, Santa Barbara, California, USA
| | - James A. Thomson
- Center for Stem Cell Biology and Engineering, University of California at Santa Barbara, Santa Barbara, California, USA,Neuroscience Research Institute, University of California at Santa Barbara, Santa Barbara, California, USA,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, USA,Morgridge Institute for Research, Madison, Wisconsin, USA,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
17
|
Cheng Y, Yu X, Zhang J, Chang Y, Xue M, Li X, Lu Y, Li T, Meng Z, Su L, Sun B, Chen L. Pancreatic kallikrein protects against diabetic retinopathy in KK Cg-A y/J and high-fat diet/streptozotocin-induced mouse models of type 2 diabetes. Diabetologia 2019; 62:1074-1086. [PMID: 30838453 PMCID: PMC6509079 DOI: 10.1007/s00125-019-4838-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/14/2019] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS Many studies have shown that tissue kallikrein has effects on diabetic vascular complications such as nephropathy, cardiomyopathy and neuropathy, but its effects on diabetic retinopathy are not fully understood. Here, we investigated the retinoprotective role of exogenous pancreatic kallikrein and studied potential mechanisms of action. METHODS We used KK Cg-Ay/J (KKAy) mice (a mouse model of spontaneous type 2 diabetes) and mice with high-fat diet/streptozotocin (STZ)-induced type 2 diabetes as our models. After the onset of diabetes, both types of mice were injected intraperitoneally with either pancreatic kallikrein (KKAy + pancreatic kallikrein and STZ + pancreatic kallikrein groups) or saline (KKAy + saline and STZ + saline groups) for 12 weeks. C57BL/6J mice were used as non-diabetic controls for both models. We analysed pathological changes in the retina; evaluated the effects of pancreatic kallikrein on retinal oxidative stress, inflammation and apoptosis; and measured the levels of bradykinin and B1 and B2 receptors in both models. RESULTS In both models, pancreatic kallikrein improved pathological structural features of the retina, increasing the thickness of retinal layers, and attenuated retinal acellular capillary formation and vascular leakage (p < 0.05). Furthermore, pancreatic kallikrein ameliorated retinal oxidative stress, inflammation and apoptosis in both models (p < 0.05). We also found that the levels of bradykinin and B1 and B2 receptors were increased after pancreatic kallikrein in both models (p < 0.05). CONCLUSIONS/INTERPRETATION Pancreatic kallikrein can protect against diabetic retinopathy by activating B1 and B2 receptors and inhibiting oxidative stress, inflammation and apoptosis. Thus, pancreatic kallikrein may represent a new therapeutic agent for diabetic retinopathy.
Collapse
Affiliation(s)
- Ying Cheng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaochen Yu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Jie Zhang
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yunpeng Chang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Mei Xue
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaoyu Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Yunhong Lu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Ting Li
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Ziyu Meng
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China
| | - Long Su
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China.
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Metabolic Diseases Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
18
|
Dreisig K, Blixt FW, Warfvinge K. Retinal Cryo-sections, Whole-Mounts, and Hypotonic Isolated Vasculature Preparations for Immunohistochemical Visualization of Microvascular Pericytes. J Vis Exp 2018. [PMID: 30346386 DOI: 10.3791/57733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Retinal pericytes play an important role in many diseases of the eye. Immunohistochemical staining techniques of retinal vessels and microvascular pericytes are central to ophthalmological research. It is vital to choose an appropriate method of visualizing the microvascular pericytes. We describe retinal microvascular pericyte immunohistochemical staining in cryo-sections, whole-mounts, and hypotonic isolated vasculature using antibodies for platelet-derived growth factor receptor β (PDGFRβ) and nerve/glial antigen 2 (NG2). This allows us to highlight advantages and shortcomings of each of the three tissue preparations for the visualization of the retinal microvascular pericytes. Cryo-sections provide transsectional visualization of all retinal layers but contain only a few occasional transverse cuts of the microvasculature. Whole-mount provides an overview of the entire retinal vasculature, but visualization of the microvasculature can be troublesome. Hypotonic isolation provides a method to visualize the entire retinal vasculature by the removal of neuronal cells, but this makes the tissue very fragile.
Collapse
Affiliation(s)
- Karin Dreisig
- Department of Clinical Experimental Research, Glostrup Research Institute
| | | | - Karin Warfvinge
- Department of Clinical Experimental Research, Glostrup Research Institute; Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University;
| |
Collapse
|
19
|
Fouda AY, Xu Z, Shosha E, Lemtalsi T, Chen J, Toque HA, Tritz R, Cui X, Stansfield BK, Huo Y, Rodriguez PC, Smith SB, Caldwell RW, Narayanan SP, Caldwell RB. Arginase 1 promotes retinal neurovascular protection from ischemia through suppression of macrophage inflammatory responses. Cell Death Dis 2018; 9:1001. [PMID: 30254218 PMCID: PMC6156564 DOI: 10.1038/s41419-018-1051-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/24/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
Abstract
The lack of effective therapies to limit neurovascular injury in ischemic retinopathy is a major clinical problem. This study aimed to examine the role of ureohydrolase enzyme, arginase 1 (A1), in retinal ischemia-reperfusion (IR) injury. A1 competes with nitric oxide synthase (NOS) for their common substrate l-arginine. A1-mediated l-arginine depletion reduces nitric oxide (NO) formation by NOS leading to vascular dysfunction when endothelial NOS is involved but prevents inflammatory injury when inducible NOS is involved. Studies were performed using wild-type (WT) mice, global A1+/− knockout (KO), endothelial-specific A1 KO, and myeloid-specific A1 KO mice subjected to retinal IR injury. Global as well as myeloid-specific A1 KO mice showed worsened IR-induced neuronal loss and retinal thinning. Deletion of A1 in endothelial cells had no effect, while treatment with PEGylated (PEG) A1 improved neuronal survival in WT mice. In addition, A1+/− KO mice showed worsened vascular injury manifested by increased acellular capillaries. Western blotting analysis of retinal tissue showed increased inflammatory and necroptotic markers with A1 deletion. In vitro experiments showed that macrophages lacking A1 exhibit increased inflammatory response upon LPS stimulation. PEG-A1 treatment dampened this inflammatory response and decreased the LPS-induced metabolic reprogramming. Moreover, intravitreal injection of A1 KO macrophages or systemic macrophage depletion with clodronate liposomes increased neuronal loss after IR injury. These results demonstrate that A1 reduces IR injury-induced retinal neurovascular degeneration via dampening macrophage inflammatory responses. Increasing A1 offers a novel strategy for limiting neurovascular injury and promoting macrophage-mediated repair.
Collapse
Affiliation(s)
- Abdelrahman Y Fouda
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Zhimin Xu
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Esraa Shosha
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Tahira Lemtalsi
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Haroldo A Toque
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Rebekah Tritz
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Xuezhi Cui
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Brian K Stansfield
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Yuqing Huo
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | | | - Sylvia B Smith
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA.,Department of Ophthalmology, Augusta University, Augusta, GA, USA
| | - R William Caldwell
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - S Priya Narayanan
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, USA
| | - Ruth B Caldwell
- Charlie Norwood VA Medical Center, Augusta, GA, USA. .,Vascular Biology Center, Augusta University, Augusta, GA, USA. .,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA. .,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA. .,Department of Ophthalmology, Augusta University, Augusta, GA, USA.
| |
Collapse
|
20
|
Pi J, Cheng Y, Sun H, Chen X, Zhuang T, Liu J, Li Y, Chang H, Zhang L, Zhang Y, Tao T. Apln-CreERT:mT/mG reporter mice as a tool for sprouting angiogenesis study. BMC Ophthalmol 2017; 17:163. [PMID: 28865439 PMCID: PMC5581477 DOI: 10.1186/s12886-017-0556-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/24/2017] [Indexed: 01/23/2023] Open
Abstract
Background Angiogenesis is defined as a new blood vessel sprouting from pre-existing vessels, and the sprouting angiogenesis is the start phase of angiogenesis, which is critical for both physiological and pathological processes, such as embryonic development, organ growth, wound healing, tumor growth, diabetic retinopathy and age-related macular degeneration. Better understanding of the mechanisms of sprout angiogenesis will provide a rationale for the treatments of these angiogenesis related diseases. Methods mT/mG tool mice are crossed with Apln-CreERT mice to generate Apln-CreERT: mT/mG mice, then we used neonatal retinal angiogenesis model to observe the angiogenic pattern of Apln-CreERT:mT/mG mice compared with Cdh5-CreERT:mT/mG mice. FACS analysis was used to sort eGFP and tdTomato endothelial cells (ECs) for measuring Apelin and Cdh5 expression. Retinal sprouting angiogenesis pattern was also observed at different neonatal time when induced by tamoxifen and at hypoxia condition, as well as in vivo tumor in real-time angiogenesis in a dorsal skinfold window chamber in Apln-CreERT:mT/mG mice. Results Apln-CreERT:mT/mG mice exhibited eGFP signal only in the sprouting angiogenesis, with less eGFP expression in the retinal “optic nerve” area than in that of Cdh5-CreERT: mT/mG mice, which might be due to relative mature vessels in the “optic nerve” area. The ECs sorted by FACS confirmed that the Apelin expression level was higher in eGFP ECs than tdTomato ECs of “optic nerve” area. Further we found that GFP-labeled sprouting angiogenesis decreased gradually following tamoxifen administration from P5-P7, but increased significantly during hypoxia in Apln-CreERT:mT/mG mice. At last, using Apln-CreERT:mT/mG mice we found tumor sprouting angiogenesis in dorsal skinfold, but not in the normal skinfold tissue. Conclusions Apln-CreERT:mT/mG mouse line is a useful tool to differentiate sprouting angiogenesis from whole blood vessels in the investigation of retinal and tumor sprouting angiogenesis in vivo.
Collapse
Affiliation(s)
- Jingjiang Pi
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Cheng
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, 197 Ruijin Er Rd, Huangpu District, Shanghai, 200025, China
| | - Huimin Sun
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yixi Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Dalian Medical University, Liaoning, 116044, China
| | - Huan Chang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Dalian Medical University, Liaoning, 116044, China
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - YuZhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ting Tao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, 197 Ruijin Er Rd, Huangpu District, Shanghai, 200025, China.
| |
Collapse
|
21
|
Promsote W, Powell FL, Veean S, Thounaojam M, Markand S, Saul A, Gutsaeva D, Bartoli M, Smith SB, Ganapathy V, Martin PM. Oral Monomethyl Fumarate Therapy Ameliorates Retinopathy in a Humanized Mouse Model of Sickle Cell Disease. Antioxid Redox Signal 2016; 25:921-935. [PMID: 27393735 PMCID: PMC5144884 DOI: 10.1089/ars.2016.6638] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS Sickle retinopathy (SR) is a major cause of blindness in sickle cell disease (SCD). The genetic mutation responsible for SCD is known, however; oxidative stress and inflammation also figure prominently in the development and progression of pathology. Development of therapies for SR is hampered by the lack of (a) animal models that accurately recapitulate human SR and (b) strategies for noninvasive yet effective retinal drug delivery. This study addressed both issues by validating the Townes humanized SCD mouse as a model of SR and demonstrating the efficacy of oral administration of the antioxidant fumaric acid ester monomethyl fumarate (MMF) in the disease. RESULTS In vivo ophthalmic imaging, electroretinography, and postmortem histological RNA and protein analyses were used to monitor retinal health and function in normal (HbAA) and sickle (HbSS) hemoglobin-producing mice over a one-year period and in additional HbAA and HbSS mice treated with MMF (15 mg/ml) for 5 months. Functional and morphological abnormalities and molecular hallmarks of oxidative stress/inflammation were evident early in HbSS retinas and increased in number and severity with age. Treatment with MMF, a known inducer of Nrf2, induced γ-globin expression and fetal hemoglobin production, improved hematological profiles, and ameliorated SR-related pathology. Innovation and Conclusion: United States Food and Drug Administration-approved formulations in which MMF is the primary bioactive ingredient are currently available to treat multiple sclerosis; such drugs may be effective for treatment of ocular and systemic complications of SCD, and given the pleiotropic effects, other nonsickle-related diseases in which oxidative stress, inflammation, and retinal vascular pathology figure prominently. Antioxid. Redox Signal. 25, 921-935.
Collapse
Affiliation(s)
- Wanwisa Promsote
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Folami Lamoke Powell
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Satyam Veean
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Menaka Thounaojam
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Shanu Markand
- 3 Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Alan Saul
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Diana Gutsaeva
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Manuela Bartoli
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Sylvia B Smith
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,3 Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Vadivel Ganapathy
- 5 Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Pamela M Martin
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| |
Collapse
|
22
|
Arginase 2 promotes neurovascular degeneration during ischemia/reperfusion injury. Cell Death Dis 2016; 7:e2483. [PMID: 27882947 PMCID: PMC5260867 DOI: 10.1038/cddis.2016.295] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/18/2016] [Accepted: 08/17/2016] [Indexed: 02/07/2023]
Abstract
Retinal ischemia is a major cause of visual impairment and blindness and is involved in various disorders including diabetic retinopathy, glaucoma, optic neuropathies and retinopathy of prematurity. Neurovascular degeneration is a common feature of these pathologies. Our lab has previously reported that the ureahydrolase arginase 2 (A2) is involved in ischemic retinopathies. Here, we are introducing A2 as a therapeutic target to prevent neurovascular injury after retinal ischemia/reperfusion (I/R) insult. Studies were performed with mice lacking both copies of A2 (A2−/−) and wild-type (WT) controls (C57BL6J). I/R insult was conducted on the right eye and the left eye was used as control. Retinas were collected for analysis at different times (3 h–4 week after injury). Neuronal and microvascular degeneration were evaluated using NeuN staining and vascular digests, respectively. Glial activation was evaluated by glial fibrillary acidic protein expression. Necrotic cell death was studied by propidium iodide labeling and western blot for RIP-3. Arginase expression was determined by western blot and quantitative RT-PCR. Retinal function was determined by electroretinography (ERG). A2 mRNA and protein levels were increased in WT I/R. A2 deletion significantly reduced ganglion cell loss and microvascular degeneration and preserved retinal morphology after I/R. Glial activation, reactive oxygen species formation and cell death by necroptosis were significantly reduced by A2 deletion. ERG showed improved positive scotopic threshold response with A2 deletion. This study shows for the first time that neurovascular injury after retinal I/R is mediated through increased expression of A2. Deletion of A2 was found to be beneficial in reducing neurovascular degeneration after I/R.
Collapse
|
23
|
Shi L, Kim AJ, Chang RCA, Chang JYA, Ying W, Ko ML, Zhou B, Ko GYP. Deletion of miR-150 Exacerbates Retinal Vascular Overgrowth in High-Fat-Diet Induced Diabetic Mice. PLoS One 2016; 11:e0157543. [PMID: 27304911 PMCID: PMC4909316 DOI: 10.1371/journal.pone.0157543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness among American adults above 40 years old. The vascular complication in DR is a major cause of visual impairment, making finding therapeutic targets to block pathological angiogenesis a primary goal for developing DR treatments. MicroRNAs (miRs) have been proposed as diagnostic biomarkers and potential therapeutic targets for various ocular diseases including DR. In diabetic animals, the expression levels of several miRs, including miR-150, are altered. The expression of miR-150 is significantly suppressed in pathological neovascularization in mice with hyperoxia-induced retinopathy. The purpose of this study was to investigate the functional role of miR-150 in the development of retinal microvasculature complications in high-fat-diet (HFD) induced type 2 diabetic mice. Wild type (WT) and miR-150 null mutant (miR-150-/-) male mice were given a HFD (59% fat calories) or normal chow diet. Chronic HFD caused a decrease of serum miR-150 in WT mice. Mice on HFD for 7 months (both WT and miR-150-/-) had significant decreases in retinal light responses measured by electroretinograms (ERGs). The retinal neovascularization in miR-150-/--HFD mice was significantly higher compared to their age matched WT-HFD mice, which indicates that miR-150 null mutation exacerbates chronic HFD-induced neovascularization in the retina. Overexpression of miR-150 in cultured endothelial cells caused a significant reduction of vascular endothelial growth factor receptor 2 (VEGFR2) protein levels. Hence, deletion of miR-150 significantly increased the retinal pathological angiogenesis in HFD induced type 2 diabetic mice, which was in part through VEGFR2.
Collapse
Affiliation(s)
- Liheng Shi
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Andy Jeesu Kim
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Richard Cheng-An Chang
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Janet Ya-An Chang
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Wei Ying
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Michael L. Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Beiyan Zhou
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
- Department of Immunology, University of Connecticut Health Center School of Medicine, Farmington, Connecticut, United States of America
| | - Gladys Yi-Ping Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute of Neuroscience, Texas A&M University, College Station, Texas 77843–4458, United States of America
| |
Collapse
|
24
|
Ghanian Z, Staniszewski K, Jamali N, Sepehr R, Wang S, Sorenson CM, Sheibani N, Ranji M. Quantitative Assessment of Retinopathy Using Multi-parameter Image Analysis. JOURNAL OF MEDICAL SIGNALS AND SENSORS 2016; 6:71-80. [PMID: 27186534 PMCID: PMC4855887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A multi-parameter quantification method was implemented to quantify retinal vascular injuries in microscopic images of clinically relevant eye diseases. This method was applied to wholemount retinal trypsin digest images of diabetic Akita/+, and bcl-2 knocked out mice models. Five unique features of retinal vasculature were extracted to monitor early structural changes and retinopathy, as well as quantifying the disease progression. Our approach was validated through simulations of retinal images. Results showed fewer number of cells (P = 5.1205e-05), greater population ratios of endothelial cells to pericytes (PCs) (P = 5.1772e-04; an indicator of PC loss), higher fractal dimension (P = 8.2202e-05), smaller vessel coverage (P = 1.4214e-05), and greater number of acellular capillaries (P = 7.0414e-04) for diabetic retina as compared to normal retina. Quantification using the present method would be helpful in evaluating physiological and pathological retinopathy in a high-throughput and reproducible manner.
Collapse
Affiliation(s)
- Zahra Ghanian
- Biophotonics Laboratory, University of Wisconsin Milwaukee, Department of Electrical Engineering and Computer Science, 3200 N Cramer St., Milwaukee, WI 53211-3029, USA
| | - Kevin Staniszewski
- Biophotonics Laboratory, University of Wisconsin Milwaukee, Department of Electrical Engineering and Computer Science, 3200 N Cramer St., Milwaukee, WI 53211-3029, USA
| | - Nasim Jamali
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA
| | - Reyhaneh Sepehr
- Biophotonics Laboratory, University of Wisconsin Milwaukee, Department of Electrical Engineering and Computer Science, 3200 N Cramer St., Milwaukee, WI 53211-3029, USA
| | - Shoujian Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA
| | - Christine M. Sorenson
- Department of Pediatircs, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA,McPherson Eye research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA,McPherson Eye research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA,Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA
| | - Mahsa Ranji
- Biophotonics Laboratory, University of Wisconsin Milwaukee, Department of Electrical Engineering and Computer Science, 3200 N Cramer St., Milwaukee, WI 53211-3029, USA,McPherson Eye research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705-2275, USA,Address for correspondence: Prof. Mahsa Ranji, University of Wisconsin-Milwaukee, Biophotonics Lab, EE Department, 3200 N Cramer St, EMS room 1195, Milwaukee, WI 53211, Tel: (414) 229-5889, Fax: (414) 229-6958, USA. E-mail:
| |
Collapse
|
25
|
Inner retinal oxygen metabolism in the 50/10 oxygen-induced retinopathy model. Sci Rep 2015; 5:16752. [PMID: 26576731 PMCID: PMC4649746 DOI: 10.1038/srep16752] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023] Open
Abstract
Retinopathy of prematurity (ROP) represents a major cause of childhood vision loss worldwide. The 50/10 oxygen-induced retinopathy (OIR) model mimics the findings of ROP, including peripheral vascular attenuation and neovascularization. The oxygen metabolism of the inner retina has not been previously explored in this model. Using visible-light optical coherence tomography (vis-OCT), we measured the oxygen saturation of hemoglobin and blood flow within inner retinal vessels, enabling us to compute the inner retinal oxygen delivery (irDO2) and metabolic rate of oxygen (irMRO2). We compared these measurements between age-matched room-air controls and rats with 50/10 OIR on postnatal day 18. To account for a 61% decrease in the irDO2 in the OIR group, we found an overall statistically significant decrease in retinal vascular density affecting the superficial and deep retinal vascular capillary networks in rats with OIR compared to controls. Furthermore, matching the reduced irDO2, we found a 59% decrease in irMRO2, which we correlated with a statistically significant reduction in retinal thickness in the OIR group, suggesting that the decreased irMRO2 was due to decreased neuronal oxygen utilization. By exploring these biological and metabolic changes in great detail, our study provides an improved understanding of the pathophysiology of OIR model.
Collapse
|
26
|
Chou JC, Rollins SD, Ye M, Batlle D, Fawzi AA. Endothelin receptor-A antagonist attenuates retinal vascular and neuroretinal pathology in diabetic mice. Invest Ophthalmol Vis Sci 2014; 55:2516-25. [PMID: 24644048 DOI: 10.1167/iovs.13-13676] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE We sought to determine the effects of atrasentan, a selective endothelin-A receptor antagonist, on the retinal vascular and structural integrity in a db/db mouse, an animal model of type 2 diabetes and diabetic retinopathy. METHODS Diabetic mice, 23 weeks old, were given either atrasentan or vehicle treatment in drinking water for 8 weeks. At the end of the treatment period, eyes underwent trypsin digest to assess the retinal vascular pathology focusing on capillary degeneration, endothelial cell, and pericyte loss. Paraffin-embedded retinal cross sections were used to evaluate retinal sublayer thickness both near the optic nerve and in the retinal periphery. Immunohistochemistry and TUNEL assay were done to evaluate retinal cellular and vascular apoptosis. RESULTS Compared with untreated db/db mice, atrasentan treatment was able to ameliorate the retinal vascular pathology by reducing pericyte loss (29.2% ± 0.4% vs. 44.4% ± 2.0%, respectively, P < 0.05) and capillary degeneration as determined by the percentage of acellular capillaries (8.6% ± 0.3% vs. 3.3% ± 0.41%, respectively, P < 0.05). A reduction in inner retinal thinning both at the optic nerve and at the periphery in treated diabetic mice was also observed in db/db mice treated with atrasentan as compared with untreated db/db mice (P < 0.05). TUNEL assay suggested that atrasentan may decrease enhanced apoptosis in neuroretinal layers and vascular pericytes in the db/db mice. CONCLUSIONS Endothelin-A receptor blockade using atrasentan significantly reduces the vascular and neuroretinal complications in diabetic mice. Endothelin-A receptor blockade is a promising therapeutic target in diabetic retinopathy.
Collapse
Affiliation(s)
- Jonathan C Chou
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | | | | | | | | |
Collapse
|
27
|
Fawzi AA, Chou JC, Kim GA, Rollins SD, Taylor JM, Farrow KN. Sildenafil attenuates vaso-obliteration and neovascularization in a mouse model of retinopathy of prematurity. Invest Ophthalmol Vis Sci 2014; 55:1493-501. [PMID: 24519428 DOI: 10.1167/iovs.13-13207] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE We sought to determine the effect of sildenafil on retinal vascular changes in a mouse model of oxygen-induced retinopathy (OIR). METHODS Vascular defects in OIR mice were quantified by measuring vaso-obliteration at postnatal days 12 and 17 (P12 and P17) and neovascularization at P17 to compare sildenafil-treated to dextrose-treated OIR mice. Retinal HIF1α protein expression was quantified by Western blotting and normalized to that of β-actin. Right ventricular hypertrophy was measured by Fulton's index as a surrogate for hyperoxia-induced pulmonary hypertension. RESULTS At P12, OIR mice treated with sildenafil demonstrated a 24% reduction in vaso-obliteration (P < 0.05), whereas at P17, treated animals showed a 50% reduction in neovascularization (P < 0.05) compared to dextrose-treated controls. Sildenafil-treated OIR mice had stabilization of retinal HIF1α at P12, immediately after hyperoxia. At P17, sildenafil-treated OIR mice had decreased HIF1α relative to untreated mice. OIR mice developed right ventricle hypertrophy that was significant compared to that in room air controls, which was abrogated by sildenafil. CONCLUSIONS Sildenafil treatment significantly decreased retinal vaso-obliteration and neovascularization in a mouse OIR model. These effects are likely due to sildenafil-induced HIF1α stabilization during hyperoxia exposure. Furthermore, we confirm disease overlap by showing that OIR mice also develop hyperoxia-induced right ventricular hypertrophy, which is prevented by sildenafil. This study is a first step toward delineating a potential therapeutic role for sildenafil in OIR and further suggests that there may be common pathophysiologic mechanisms underlying hyperoxia-induced retinal and pulmonary vascular disease.
Collapse
Affiliation(s)
- Amani A Fawzi
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | | | | |
Collapse
|
28
|
Chou J, Rollins S, Fawzi AA. Role of endothelial cell and pericyte dysfunction in diabetic retinopathy: review of techniques in rodent models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:669-75. [PMID: 24664757 PMCID: PMC4324463 DOI: 10.1007/978-1-4614-3209-8_84] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Diabetic Retinopathy is one of the hallmark microvascular diseases secondary to diabetes. Endothelial cells and pericytes are key players in the pathogenesis. Interaction between the two cell types is important in the regulation of vascular function and the maintenance of the retinal homeostatic environment. There are currently several approaches to analyze changes in morphology and function of the two cell types. Morphologic approaches include trypsin digest, while functional approaches include studying blood flow. This review explores the advantages and limitations of various methods and summarizes recent experimental studies of EC and pericyte dysfunction in rodent models of DR. An improved understanding of the role played by EC and pericyte dysfunction can lead to enhanced insights into retinal vascular regulation in DR and open new avenues for future treatments that reverse their dysfunction.
Collapse
Affiliation(s)
- Jonathan Chou
- Department of Ophthalmology, Northwestern Feinberg School of Medicine, 645 N Michigan Ave., 60611, Chicago, IL, USA,
| | | | | |
Collapse
|