Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Article in English | MEDLINE | ID: mdl-37487631

ABSTRACT

Cell-replacement therapies are a new class of treatments, which include induced pluripotent stem cell (iPSC)-derived tissues that aim to replace degenerated cells. iPSCs can potentially be used to generate any cell type of the body, making them a powerful tool for treating degenerative diseases. Cell replacement for retinal degenerative diseases is at the forefront of cell therapies, given the accessibility of the eye for surgical procedures and a huge unmet medical need for retinal degenerative diseases with no current treatment options. Clinical trials are ongoing in different parts of the world using stem cell-derived retinal pigment epithelium (RPE). This review focuses on scientific and regulatory considerations when developing an iPSC-derived RPE cell therapy from the development of a robust and efficient differentiation protocol to critical quality control assays for cell validation, the choice of an appropriate animal model for preclinical testing, and the regulatory aspects that dictate the final approval for proceeding to a first-in-human clinical trial.


Subject(s)
Induced Pluripotent Stem Cells , Animals , Humans , Retinal Pigment Epithelium/metabolism , Cell Differentiation , Models, Animal
2.
Exp Eye Res ; 236: 109666, 2023 11.
Article in English | MEDLINE | ID: mdl-37783334

ABSTRACT

Angiogenesis, although required during eye development, has a causative effect in many ocular diseases. Aberrant neovascularization contributes to the progression of neovascular age-related macular degeneration (nAMD), a vision-threaten disease in aging Americans. Since increased amounts of vascular endothelial growth factor (VEGF) drives neovascularization during the pathogenesis of nAMD the standard of care are anti-VEGF therapies attempt to disrupt this vicious cycle. These current anti-VEGF therapies try to maintain vascular homeostasis while abating aberrant neovascularization but regrettably don't prevent fibrosis or scar formation. In addition, some patients demonstrate an incomplete response to anti-VEGF therapy as demonstrated by progressive vision loss. Here, we show choroidal endothelial cells (ChEC) incubated with artesunate demonstrated decreased migration and inflammatory and fibrotic factor expression, which corresponded with decreased sprouting in a choroid/retinal pigment epithelium (RPE) explant sprouting angiogenesis assay. To assess the efficacy of artesunate to curtail neovascularization in vivo, we utilized laser photocoagulation-induced rupture of the Bruch's membrane to induce choroidal neovascularization (CNV). Artesunate significantly inhibited CNV and the accompanying fibrotic scar, perhaps due in part to its ability to inhibit mononuclear phagocyte (MP) recruitment. Thus, artesunate shows promise in inhibiting both CNV and fibrosis.


Subject(s)
Choroidal Neovascularization , Vascular Endothelial Growth Factor A , Humans , Animals , Mice , Vascular Endothelial Growth Factor A/metabolism , Artesunate/therapeutic use , Cicatrix/prevention & control , Cicatrix/pathology , Endothelial Cells/metabolism , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/prevention & control , Choroidal Neovascularization/etiology , Vascular Endothelial Growth Factors , Disease Models, Animal , Mice, Inbred C57BL
3.
J Vis Exp ; (197)2023 07 28.
Article in English | MEDLINE | ID: mdl-37578220

ABSTRACT

The retinal pigment epithelium (RPE) is a monolayer of hexagonal cells located at the back of the eye. It provides nourishment and support to photoreceptors and choroidal capillaries, performs phagocytosis of photoreceptor outer segments (POS), and secretes cytokines in a polarized manner for maintaining the homeostasis of the outer retina. Dysfunctional RPE, caused by mutations, aging, and environmental factors, results in the degeneration of other retinal layers and causes vision loss. A hallmark phenotypic feature of degenerating RPE is intra and sub-cellular lipid-rich deposits. These deposits are a common phenotype across different retinal degenerative diseases. To reproduce the lipid deposit phenotype of monogenic retinal degenerations in vitro, induced pluripotent stem cell-derived RPE (iRPE) was generated from patients' fibroblasts. Cell lines generated from patients with Stargardt and Late-onset retinal degeneration (L-ORD) disease were fed with POS for 7 days to replicate RPE physiological function, which caused POS phagocytosis-induced pathology in these diseases. To generate a model for age-related macular degeneration (AMD), a polygenic disease associated with alternate complement activation, iRPE was challenged with alternate complement anaphylatoxins. The intra and sub-cellular lipid deposits were characterized using Nile Red, boron-dipyrromethene (BODIPY), and apolipoprotein E (APOE). To quantify the density of lipid deposits, a machine learning-based software, LipidUNet, was developed. The software was trained on maximum-intensity projection images of iRPE on culture surfaces. In the future, it will be trained to analyze three-dimensional (3D) images and quantify the volume of lipid droplets. The LipidUNet software will be a valuable resource for discovering drugs that decrease lipid accumulation in disease models.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Humans , Retinal Pigment Epithelium , Retina , Retinal Degeneration/pathology , Lipids
4.
Pharmacol Ther ; 249: 108482, 2023 09.
Article in English | MEDLINE | ID: mdl-37385300

ABSTRACT

Stargardt maculopathy, caused predominantly by mutations in the ABCA4 gene, is characterized by an accumulation of non-degradable visual pigment derivative, lipofuscin, in the retinal pigment epithelium (RPE) - resulting in RPE atrophy. RPE is a monolayer tissue located adjacent to retinal photoreceptors and regulates their health and functioning; RPE atrophy triggers photoreceptor cell death and vision loss in Stargardt patients. Previously, ABCA4 mutations in photoreceptors were thought to be the major contributor to lipid homeostasis defects in the eye. Recently, we demonstrated that ABCA4 loss of function in the RPE leads to cell-autonomous lipid homeostasis defects. Our work underscores that an incomplete understanding of lipid metabolism and lipid-mediated signaling in the retina and RPE are potential causes for lacking treatments for this disease. Here we report altered lipidomic in mouse and human Stargardt models. This work provides the basis for therapeutics that aim to restore lipid homeostasis in the retina and the RPE.


Subject(s)
Macular Degeneration , Retinal Degeneration , Humans , Mice , Animals , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retina/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Lipofuscin/genetics , Lipofuscin/metabolism , Atrophy/metabolism , Atrophy/pathology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism
5.
JCI Insight ; 8(11)2023 06 08.
Article in English | MEDLINE | ID: mdl-37288665

ABSTRACT

Over 30 million people worldwide suffer from untreatable vision loss and blindness associated with childhood-onset and age-related eye diseases caused by photoreceptor (PR), retinal pigment epithelium (RPE), and choriocapillaris (CC) degeneration. Recent work suggests that RPE-based cell therapy may slow down vision loss in late stages of age-related macular degeneration (AMD), a polygenic disease induced by RPE atrophy. However, accelerated development of effective cell therapies is hampered by the lack of large-animal models that allow testing safety and efficacy of clinical doses covering the human macula (20 mm2). We developed a versatile pig model to mimic different types and stages of retinal degeneration. Using an adjustable power micropulse laser, we generated varying degrees of RPE, PR, and CC damage and confirmed the damage by longitudinal analysis of clinically relevant outcomes, including analyses by adaptive optics and optical coherence tomography/angiography, along with automated image analysis. By imparting a tunable yet targeted damage to the porcine CC and visual streak - with a structure similar to the human macula - this model is optimal for testing cell and gene therapies for outer retinal diseases including AMD, retinitis pigmentosa, Stargardt, and choroideremia. The amenability of this model to clinically relevant imaging outcomes will facilitate faster translation to patients.


Subject(s)
Macular Degeneration , Retinal Degeneration , Humans , Animals , Swine , Child , Retina/diagnostic imaging , Retinal Degeneration/etiology , Retinal Degeneration/therapy , Retinal Pigment Epithelium , Macular Degeneration/therapy
6.
Stem Cell Reports ; 17(11): 2438-2450, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36306781

ABSTRACT

Stargardt retinopathy is an inherited form of macular degeneration caused by mutations in gene ABCA4 and characterized by the accumulation of lipid-rich deposits in the retinal pigment epithelium (RPE), RPE atrophy, and photoreceptor cell death. Inadequate mechanistic insights into pathophysiological changes occurring in Stargardt RPE have hindered disease treatments. Here, we show that ABCA4 knockout and induced pluripotent stem cell-derived RPE (STGD1-iRPE) from patients with Stargardt differentiate normally but display intracellular lipid and ceramide deposits reminiscent of the disease phenotype. STGD1-iRPE also shows defective photoreceptor outer segment (POS) processing and reduced cathepsin B activity-indicating higher lysosomal pH. Lipid deposits in STGD1-iRPE are lowered by increasing the activity of ABCA1, a lipid transporter, and ABCA4 ortholog. Our work suggests that ABCA4 is involved in POS and lipid handling in RPE cells and provides guidance for ongoing gene therapy approaches to target both RPE and photoreceptor cells for an effective treatment.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Pigment Epithelium , Humans , Induced Pluripotent Stem Cells/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Stargardt Disease , Lipids
7.
Cells ; 11(6)2022 03 11.
Article in English | MEDLINE | ID: mdl-35326420

ABSTRACT

Neovascular or wet age-related macular degeneration (nAMD) causes vision loss due to inflammatory and vascular endothelial growth factor (VEGF)-driven neovascularization processes in the choroid. Due to the excess in VEGF levels associated with nAMD, anti-VEGF therapies are utilized for treatment. Unfortunately, not all patients have a sufficient response to such therapies, leaving few if any other treatment options for these patients. Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator found in endothelial cells that participates in modulating barrier function, angiogenesis, and inflammation. S1P, through its receptor (S1PR1) in endothelial cells, prevents illegitimate sprouting angiogenesis during vascular development. In the present paper, we show that, in choroidal endothelial cells, S1PR1 is the most abundantly expressed S1P receptor and agonism of S1PR1-prevented choroidal endothelial cell capillary morphogenesis in culture. Given that nAMD pathogenesis draws from enhanced inflammation and angiogenesis as well as a loss of barrier function, we assessed the impact of S1PR agonism on choroidal neovascularization in vivo. Using laser photocoagulation rupture of Bruch's membrane to induce choroidal neovascularization, we show that S1PR non-selective (FTY720) and S1PR1 selective (CYM5442) agonists significantly inhibit choroidal neovascularization in this model. Thus, utilizing S1PR agonists to temper choroidal neovascularization presents an additional novel use for these agonists presently in clinical use for multiple sclerosis as well as other inflammatory diseases.


Subject(s)
Choroidal Neovascularization , Fingolimod Hydrochloride , Choroid/metabolism , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/metabolism , Endothelial Cells/metabolism , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , Inflammation/pathology , Phosphates , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factors
8.
Commun Biol ; 4(1): 1360, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34887495

ABSTRACT

Late-onset retinal degeneration (L-ORD) is an autosomal dominant disorder caused by a missense substitution in CTRP5. Distinctive clinical features include sub-retinal pigment epithelium (RPE) deposits, choroidal neovascularization, and RPE atrophy. In induced pluripotent stem cells-derived RPE from L-ORD patients (L-ORD-iRPE), we show that the dominant pathogenic CTRP5 variant leads to reduced CTRP5 secretion. In silico modeling suggests lower binding of mutant CTRP5 to adiponectin receptor 1 (ADIPOR1). Downstream of ADIPOR1 sustained activation of AMPK renders it insensitive to changes in AMP/ATP ratio resulting in defective lipid metabolism, reduced Neuroprotectin D1(NPD1) secretion, lower mitochondrial respiration, and reduced ATP production. These metabolic defects result in accumulation of sub-RPE deposits and leave L-ORD-iRPE susceptible to dedifferentiation. Gene augmentation of L-ORD-iRPE with WT CTRP5 or modulation of AMPK, by metformin, re-sensitize L-ORD-iRPE to changes in cellular energy status alleviating the disease cellular phenotypes. Our data suggests a mechanism for the dominant behavior of CTRP5 mutation and provides potential treatment strategies for L-ORD patients.


Subject(s)
AMP-Activated Protein Kinases/genetics , Retinal Degeneration/genetics , AMP-Activated Protein Kinases/metabolism , Female , Humans , Male , Middle Aged , Phenotype
9.
Stem Cells Transl Med ; 10(2): 198-208, 2021 02.
Article in English | MEDLINE | ID: mdl-32946199

ABSTRACT

Induced pluripotent stem cells (iPSC)-based therapies have been hailed as the future of regenerative medicine because of their potential to provide treatment options for most degenerative diseases. A key promise of iPSC-based therapies is the possibility of an autologous transplant that may engraft better in the longer-term due to its compatibility with the patient's immune system. Despite over a decade of research, clinical translation of autologous iPSC-based therapies has been slow-partly due to a lacking pre-defined regulatory path. Here, we outline regulatory considerations for developing an autologous iPSC-based product and challenges associated with the clinical manufacturing of autologous iPSCs and their derivatives. These challenges include donor tissue source, reprogramming methods, heterogeneity of differentiated cells, controls for the manufacturing process, and preclinical considerations. A robust manufacturing process with appropriate quality controls and well-informed, prospectively designed preclinical studies provide a path toward successful approval of autologous iPSC-based therapies.


Subject(s)
Induced Pluripotent Stem Cells , Investigational New Drug Application , Regenerative Medicine/legislation & jurisprudence , Transplantation, Autologous/legislation & jurisprudence , Cell Differentiation , Humans
10.
J Ophthalmic Vis Res ; 13(4): 470-486, 2018.
Article in English | MEDLINE | ID: mdl-30479719

ABSTRACT

Angiogenesis, the formation of new blood vessels from pre-existing capillaries, is very tightly regulated and normally does not occur except during developmental and reparative processes. This tight regulation is maintained by a balanced production of positive and negative regulators, and alterations under pathological conditions such as retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration can lead to growth of new and abnormal blood vessels. Although the role of proangiogenic factors such as vascular endothelial growth factor has been extensively studied, little is known about the roles of negative regulators of angiogenesis in the pathogenesis of these diseases. Here, we will discuss the role of thrombospondin-1 (TSP1), one of the first known endogenous inhibitors of angiogenesis, in ocular vascular homeostasis, and how its alterations may contribute to the pathogenesis of age-related macular degeneration and choroidal neovascularization. We will also discuss its potential utility as a therapeutic target for treatment of ocular diseases with a neovascular component.

11.
Am J Physiol Cell Physiol ; 314(4): C456-C472, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351407

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of vision loss among the elderly population, and is associated with severe macular degeneration and choroidal neovascularization (CNV). Although the pathogenesis of AMD is associated with choroidal dysfunction and CNV, the detailed underlying mechanisms remain unresolved. Altered production of pigment epithelium-derived factor (PEDF), a neuroprotective and antiangiogenic factor, contributes to CNV. Furthermore, exogenous PEDF mitigates angiogenesis in preclinical CNV models. How PEDF expression affects choroidal endothelial cell (ChEC) function is unknown. Here we isolated ChECs from PEDF+/+ and PEDF-deficient (PEDF-/-) mice and determined the impact of PEDF expression on the proangiogenic and pro-inflammatory properties of ChECs. We showed that PEDF expression significantly affects the proliferation, migration, adhesion, and oxidative and inflammatory state of ChECs. The PEDF-/- ChECs were, however, more sensitive to H2O2 challenge and exhibited increased rate of apoptosis and oxidative stress. We also observed a significant increase in production of cytokines with a primary role in inflammation and angiogenesis including vascular endothelial growth factor (VEGF) and osteopontin, and a reprograming of chemokines and cytokines expression profiles in PEDF-/- ChECs. Collectively, our results indicate that PEDF expression has a significant impact on oxidative and inflammatory properties of ChECs, whose alteration could contribute to pathogenesis of chronic inflammatory diseases including exudative AMD.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Eye Proteins/metabolism , Inflammation Mediators/metabolism , Macular Degeneration/metabolism , Nerve Growth Factors/metabolism , Oxidative Stress , Serpins/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Endothelial Cells/pathology , Eye Proteins/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Osteopontin/metabolism , Serpins/deficiency , Serpins/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Article in English | MEDLINE | ID: mdl-29104816

ABSTRACT

The physiological relevance of Matrigel as a cell-culture substrate and in angiogenesis assays is often called into question. Here, we describe an array-based method for the identification of synthetic hydrogels that promote the formation of robust in vitro vascular networks for the detection of putative vascular disruptors, and that support human embryonic stem cell expansion and pluripotency. We identified hydrogel substrates that promoted endothelial-network formation by primary human umbilical vein endothelial cells and by endothelial cells derived from human induced pluripotent stem cells, and used the hydrogels with endothelial networks to identify angiogenesis inhibitors. The synthetic hydrogels show superior sensitivity and reproducibility over Matrigel when evaluating known inhibitors, as well as in a blinded screen of a subset of 38 chemicals, selected according to predicted vascular disruption potential, from the Toxicity ForeCaster library of the US Environmental Protection Agency. The identified synthetic hydrogels should be suitable alternatives to Matrigel for common cell-culture applications.

13.
Clin Sci (Lond) ; 131(15): 1763-1780, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28679845

ABSTRACT

Angiogenesis contributes to the pathogenesis of many diseases including exudative age-related macular degeneration (AMD). It is normally kept in check by a tightly balanced production of pro- and anti-angiogenic factors. The up-regulation of the pro-angiogenic factor, vascular endothelial growth factor (VEGF), is intimately linked to the pathogenesis of exudative AMD, and its antagonism has been effectively targeted for treatment. However, very little is known about potential changes in expression of anti-angiogenic factors and the role they play in choroidal vascular homeostasis and neovascularization associated with AMD. Here, we will discuss the important role of thrombospondins and pigment epithelium-derived factor, two major endogenous inhibitors of angiogenesis, in retinal and choroidal vascular homeostasis and their potential alterations during AMD and choroidal neovascularization (CNV). We will review the cell autonomous function of these proteins in retinal and choroidal vascular cells. We will also discuss the potential targeting of these molecules and use of their mimetic peptides for therapeutic development for exudative AMD.


Subject(s)
Angiogenesis Inhibitors/physiology , Choroidal Neovascularization/physiopathology , Eye Proteins/physiology , Macular Degeneration/physiopathology , Nerve Growth Factors/physiology , Serpins/physiology , Thrombospondins/physiology , Angiogenesis Inhibitors/therapeutic use , Angiostatins/therapeutic use , Choroidal Neovascularization/drug therapy , Endostatins/therapeutic use , Humans , Macular Degeneration/drug therapy , Molecular Targeted Therapy/methods
14.
Invest Ophthalmol Vis Sci ; 58(1): 299-308, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28114591

ABSTRACT

Purpose: The role of ß-adrenergic receptor (AR) signaling in neovascular ocular diseases has recently emerged. We have previously reported that intraperitoneal propranolol inhibits choroidal neovascularization (CNV) in vivo and ß2-AR blockade reduces vascular endothelial growth factor (VEGF) expression in mouse retinal pigment epithelium and choroidal endothelial cells in culture. Here we tested the hypothesis that the ß2-AR regulates CNV through modulation of VEGF and inflammatory cytokine expression. Methods: Mice were subjected to laser burns, inducing CNV, and were treated with an intravitreal ß2-AR antagonist. After 3 and 5 days, total eye interleukin-6 (IL-6) and VEGF protein levels were measured, respectively. After 14 days, CNV was measured on choroidal-scleral flatmounts. The effects of ß-AR signaling on VEGF and IL-6 expression were investigated in various mouse retinal and human RPE cells by using specific ß-AR agonists and antagonists. Results: ß2-Adrenergic receptor signaling increased Vegf mRNA expression by approximately 3- to 4-fold in mouse retinal microglia and pericytes in culture. ß2-Adrenergic receptor signaling upregulated IL-6 mRNA expression between 10- and 60-fold in mouse retinal microglia, pericytes, RPE, and choroidal endothelial cells in culture. Intravitreal injection of ß2-AR antagonist ICI 118,551 reduced CNV by 35% and decreased IL-6 protein levels by approximately 50%. In primary human RPE cells, ß2-AR activation also stimulated VEGF and IL-6 mRNA expression by 2- and 10-fold, respectively. Conclusions: Anti-VEGF therapy for CNV is highly effective; however, some patients are resistant to therapy while others undergo repeated, frequent treatments. ß2-Adrenergic receptor signaling is a potential therapeutic target because of its angiogenic and inflammatory properties.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Choroidal Neovascularization/drug therapy , Gene Expression Regulation/drug effects , Interleukin-6/genetics , RNA, Messenger/genetics , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Cells, Cultured , Choroid/drug effects , Choroid/metabolism , Choroid/pathology , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , Receptors, Adrenergic, beta/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/biosynthesis
15.
Am J Physiol Cell Physiol ; 311(3): C418-36, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27440660

ABSTRACT

Defects in the outer blood-retinal barrier have significant impact on the pathogenesis of diabetic retinopathy and macular edema. However, the detailed mechanisms involved remain largely unknown. This is, in part, attributed to the lack of suitable animal and cell culture models, including those of mouse origin. We recently reported a method for the culture of retinal pigment epithelial (RPE) cells from wild-type and transgenic mice. The RPE cells are responsible for maintaining the integrity of the outer blood-retinal barrier whose dysfunction during diabetes has a significant impact on vision. Here we determined the impact of high glucose on the function of RPE cells. We showed that high glucose conditions resulted in enhanced migration and increased the level of oxidative stress in RPE cells, but minimally impacted their rate of proliferation and apoptosis. High glucose also minimally affected the cell-matrix and cell-cell interactions of RPE cells. However, the expression of integrins and extracellular matrix proteins including pigment epithelium-derived factor (PEDF) were altered under high glucose conditions. Incubation of RPE cells with the antioxidant N-acetylcysteine under high glucose conditions restored normal migration and PEDF expression. These cells also exhibited increased nuclear localization of the antioxidant transcription factor Nrf2 and ZO-1, reduced levels of ß-catenin and phagocytic activity, and minimal effect on production of vascular endothelial growth factor, inflammatory cytokines, and Akt, MAPK, and Src signaling pathways. Thus high glucose conditions promote RPE cell migration through increased oxidative stress and expression of PEDF without a significant effect on the rate of proliferation and apoptosis.


Subject(s)
Cell Movement/physiology , Eye Proteins/metabolism , Glucose/metabolism , Nerve Growth Factors/metabolism , Oxidative Stress/physiology , Serpins/metabolism , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Apoptosis/physiology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/physiology , Integrins/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiology , Retinal Pigments/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism
16.
Physiol Rep ; 3(1)2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25602019

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of vision loss among elderly. Although the pathogenesis of AMD is associated with retinal pigmented epithelium (RPE) dysfunction and abnormal neovascularization the detailed mechanisms remain unresolved. RPE is a specialized monolayer of epithelial cells with important functions in ocular homeostasis. Pathological RPE damage contributes to major ocular conditions including retinal degeneration and irreversible loss of vision in AMD. RPE cells also assist in the maintenance of the ocular angiogenic balance by production of positive and negative regulatory factors including vascular endothelial growth factor (VEGF), thrombospondin-1 (TSP1), and pigment epithelium-derived factor (PEDF). The altered production of PEDF and TSP1, as endogenous inhibitors of angiogenesis and inflammation, by RPE cells have been linked to pathogenesis of AMD and choroidal and retinal neovascularization. However, lack of simple methods for isolation and culture of mouse RPE cells has resulted in limited knowledge regarding the cell autonomous role of TSP1 and PEDF in RPE cell function. Here, we describe a method for routine isolation and propagation of RPE cells from wild-type, TSP1, and PEDF-deficient mice, and have investigated their impact on RPE cell function. We showed that expression of TSP1 and PEDF significantly impacted RPE cell proliferation, migration, adhesion, oxidative state, and phagocytic activity with minimal effect on their basal rate of apoptosis. Together, our results indicated that the expression of PEDF and TSP1 by RPE cells play crucial roles not only in regulation of ocular vascular homeostasis but also have significant impact on their cellular function.

17.
PLoS One ; 9(12): e116423, 2014.
Article in English | MEDLINE | ID: mdl-25548916

ABSTRACT

The choroidal circulation plays a central role in maintaining the health of outer retina and photoreceptor function. Alterations in this circulation contribute to pathogenesis of many eye diseases including exudative age-related macular degeneration. Unfortunately, very little is known about the choroidal circulation and its molecular and cellular regulation. This has been further hampered by the lack of methods for routine culturing of choroidal endothelial cells (ChEC), especially from wild type and transgenic mice. Here we describe a method for isolation and culturing of mouse ChEC. We show that expression of thrombospondin-1 (TSP1), an endogenous inhibitor of angiogenesis and inflammation, has a significant impact on phenotype of ChEC. ChEC from TSP1-deficient (TSP1-/-) mice were less proliferative and more apoptotic, less migratory and less adherent, and failed to undergo capillary morphogenesis in Matrigel. However, re-expression of TSP1 was sufficient to restore TSP1-/- ChEC migration and capillary morphogenesis. TSP1-/- ChEC expressed increased levels of TSP2, phosphorylated endothelial nitric oxide synthase (NOS) and inducible NOS (iNOS), a marker of inflammation, which was associated with significantly higher level of NO and oxidative stress in these cells. Wild type and TSP1-/- ChEC produced similar levels of VEGF, although TSP1-/- ChEC exhibited increased levels of VEGF-R1 and pSTAT3. Other signaling pathways including Src, Akt, and MAPKs were not dramatically affected by the lack of TSP1. Together our results demonstrate an important autocrine role for TSP1 in regulation of ChEC phenotype.


Subject(s)
Choroid/metabolism , Endothelial Cells/metabolism , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Animals , Cell Adhesion , Cell Proliferation , Cells, Cultured , Choroid/cytology , Choroid/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidative Stress , Signal Transduction , Thrombospondins/metabolism
18.
Molecules ; 19(11): 17578-603, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25361423

ABSTRACT

The objective of this study was to determine whether resveratrol or a defined, reconstituted grape powder can attenuate the formation of new blood vessels in a mouse model of choroidal neovascularization (CNV). To accomplish this objective, C57BL/6J mice were randomized into control or treatment groups which received either resveratrol or grape powder by daily oral gavage, resveratrol or grape powder delivered ad libitum through the drinking water, or resveratrol by slow release via implanted osmotic pumps. A laser was used to rupture Bruch's membrane to induce CNV which was then detected in sclerochoroidal eyecups stained with antibodies against intercellular adhesion molecule-2. CNV area was measured using fluorescence microscopy and Image J software. Ad libitum delivery of both resveratrol and grape powder was shown to significantly reduce the extent of CNV by 68% and 57%, respectively. Parallel experiments conducted in vitro demonstrated that resveratrol activates p53 and inactivates Akt/protein kinase B in choroidal endothelial cells, contributing to its anti-proliferative and anti-migratory properties. In addition resveratrol was shown to inhibit the formation of endothelial cell networks, augmenting its overall anti-angiogenic effects. The non-toxic nature of resveratrol makes it an especially attractive candidate for the prevention and/or treatment of CNV.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Choroidal Neovascularization/drug therapy , Delayed-Action Preparations/administration & dosage , Neovascularization, Pathologic/drug therapy , Powders/administration & dosage , Stilbenes/administration & dosage , Vitis/chemistry , Animals , Bruch Membrane/drug effects , Bruch Membrane/metabolism , Cell Adhesion Molecules/metabolism , Choroidal Neovascularization/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Lasers , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Resveratrol , Tumor Suppressor Protein p53/metabolism
19.
Fungal Genet Biol ; 49(11): 857-65, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23022264

ABSTRACT

Aspergillus fumigatus is an increasingly serious pathogen of immunocompromised patients, causing the often fatal disease invasive aspergillosis (IA). One A. fumigatus virulence determinant of IA is LaeA, a conserved virulence factor in pathogenic fungi. To further understand the role of LaeA in IA, the expression profile of ΔlaeA was compared to wild type, and several transcription factors were found significantly misregulated by LaeA loss. One of the transcription factors up-regulated over 4-fold in the ΔlaeA strain was Afu4g09710, similar in sequence to Aspergillus nidulans NosA, which is involved in sexual development. Here we assessed loss of nosA (ΔnosA) and overexpression of nosA (OE::nosA) on A. fumigatus in both a wild type and ΔlaeA background. Based on the multiple alterations of physiological development of single and double mutants, we suggest that NosA mediates the decreased radial growth and delayed conidial germination observed in ΔlaeA strains, the former in a light dependent manner. The ΔnosA mutant showed increased virulence in the Galleria mellonella larvae model of disseminated aspergillosis, potentially due to its increased growth and germination rate. Furthermore, the A. fumigatus nosA allele was able to partially remediate sexual development in an A. nidulans ΔnosA background. Likewise, the A. nidulans nosA allele was able to restore the menadione sensitivity defect of the A. fumigatus ΔnosA strain, suggesting conservation of function of the NosA protein in these two species.


Subject(s)
Aspergillosis/microbiology , Aspergillus fumigatus/metabolism , Fungal Proteins/metabolism , Gene Deletion , Spores, Fungal/growth & development , Transcription Factors/metabolism , Animals , Aspergillus fumigatus/genetics , Aspergillus fumigatus/growth & development , Aspergillus fumigatus/pathogenicity , Fungal Proteins/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Fungal , Humans , Moths , Spores, Fungal/genetics , Spores, Fungal/metabolism , Transcription Factors/genetics , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL