Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
JCI Insight ; 4(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31672944

ABSTRACT

Retinopathy of prematurity (ROP) is a disorder of the developing retina of preterm infants. ROP can lead to blindness because of abnormal angiogenesis that is the result of suspended vascular development and vaso-obliteration leading to severe retinal stress and hypoxia. We tested the hypothesis that the use of the human progenitor cell combination, bone marrow-derived CD34+ cells and vascular wall-derived endothelial colony-forming cells (ECFCs), would synergistically protect the developing retinal vasculature in a mouse model of ROP, called oxygen-induced retinopathy (OIR). CD34+ cells alone, ECFCs alone, or the combination thereof were injected intravitreally at either P5 or P12 and pups were euthanized at P17. Retinas from OIR mice injected with ECFCs or the combined treatment revealed formation of the deep vascular plexus (DVP) while still in hyperoxia, with normal-appearing connections between the superficial vascular plexus (SVP) and the DVP. In addition, the combination of cells completely prevented aberrant retinal neovascularization and was more effective anatomically and functionally at rescuing the ischemia phenotype than either cell type alone. We show that the beneficial effects of the cell combination are the result of their ability to orchestrate an acceleration of vascular development and more rapid ensheathment of pericytes on the developing vessels. Lastly, our proteomic and transcriptomic data sets reveal pathways altered by the dual cell therapy, including many involved in neuroretinal maintenance, and principal component analysis (PCA) showed that cell therapy restored OIR retinas to a state that was closely associated with age-matched normal retinas. Together, these data herein support the use of dual cell therapy as a promising preventive treatment for the development of ROP in premature infants.


Subject(s)
Oxygen Inhalation Therapy/adverse effects , Retinal Neovascularization/etiology , Retinopathy of Prematurity/complications , Retinopathy of Prematurity/prevention & control , Stem Cells/cytology , Animals , Antigens, CD34/immunology , Disease Models, Animal , Mice , Retinopathy of Prematurity/pathology , Stem Cells/immunology
2.
Stem Cells Int ; 2019: 3945850, 2019.
Article in English | MEDLINE | ID: mdl-31781239

ABSTRACT

We compared the functional outcome of Isl-1+ cardiac progenitors, CD90+ bone marrow-derived progenitor cells, and the combination of the two in a rat myocardial infarction (MI) model. Isl-1+ cells were isolated from embryonic day 12.5 (E12.5) rat hearts and expanded in vitro. Thy-1+/CD90+ cells were isolated from the bone marrow of adult Sprague-Dawley rats by immunomagnetic cell sorting. Six-week-old female Sprague-Dawley rats underwent permanent left anterior descending (LAD) coronary artery ligation and received intramyocardial injection of either saline, Isl-1+ cells, CD90+ cells, or a combination of Isl-1+ and CD90+ cells, at the time of infarction. Cells were delivered transepicardially to the peri-infarct zone. Left ventricular function was assessed by transthoracic echocardiography at 1- and 4-week post-MI and by Millar catheterization (-dP/dt and +dP/dt) at 4-week post-MI. Fluorescence in situ hybridization (Isl-1+cells) and monochrystalline iron oxide nanoparticles labeling (MION; CD90+ cells) were performed to assess biodistribution of transplanted cells. Only the combination of cells demonstrated a significant improvement of cardiac function as assessed by anterior wall contractility, dP/dt (max), and dP/dt (min), compared to Isl-1+ or CD90+ cell monotherapies. In the combination cell group, viable cells were detected at week 4 when anterior wall motion was completely restored. In conclusion, the combination of Isl-1+ cardiac progenitors and adult bone marrow-derived CD90+ cells shows prolonged and robust myocardial tissue repair and provides support for the use of complementary cell populations to enhance myocardial repair.

3.
Invest Ophthalmol Vis Sci ; 58(12): 5043-5055, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28980000

ABSTRACT

Purpose: We investigated the relationship between inflammation, neuronal loss, and expression of indoleamine 2, 3-dioxygenase (IDO) and quinolinic acid (QUIN) in the retina of subjects with type 1 diabetes (T1D) and type 2 diabetes (T2D) and in the retina of rats with T1D. Methods: Retinas from T1D (n = 7), T2D (n = 13), and 20 age-matched nondiabetic human donors and from T1D (n = 3) and control rats (n = 3) were examined using immunohistochemistry for IDO, QUIN, cluster of differentiation 39 (CD39), ionized calcium-binding adaptor molecule (Iba-1, for macrophages and microglia), Vimentin (VIM; for Müller cells), neuronal nuclei (NeuN; for neurons), and UEA1 lectin (for blood vessels). Results: Based on morphologic criteria, CD39+/ionized calcium binding adaptor molecule 1(Iba-1+) resident microglia and CD39-/Iba-1+ bone marrow-derived macrophages were present at higher density in T1D (13% increase) and T2D (26% increase) human retinas when compared with controls. The density and brightness of IDO+ microglia were increased in both T1D and T2D human retinas. The intensity of QUIN+ expression on CD39+ microglia and VIM+ Müller cells was greatly increased in both human T1D and T2D retinas. T1D retinas showed a 63% loss of NeuN+ neurons and T2D retinas lost approximately 43% when compared with nondiabetic human retinas. Few QUIN+ microglia-like cells were seen in nondiabetic retinas, but the numbers increased 18-fold in T1D and 7-fold in T2D in the central retina. In T1D rat retinas, the density of IDO+ microglia increased 2.8-fold and brightness increased 2.1-fold when compared with controls. Conclusions: Our findings suggest that IDO and QUIN expression in the retinas of diabetic rats and humans could contribute to the neuronal degeneration that is characteristic of diabetic retinopathy.


Subject(s)
Biomarkers/metabolism , Diabetic Retinopathy/metabolism , Ependymoglial Cells/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Microglia/metabolism , Quinolinic Acid/metabolism , Retina/metabolism , Aged , Animals , Antigens, CD/metabolism , Antigens, Nuclear/metabolism , Apyrase/metabolism , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Retinopathy/pathology , Ependymoglial Cells/pathology , Female , Fluorescent Antibody Technique, Indirect , Humans , Male , Microfilament Proteins/metabolism , Microglia/pathology , Microscopy, Confocal , Middle Aged , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Retina/pathology , Vimentin/metabolism
4.
Eye Brain ; 8: 113-122, 2016.
Article in English | MEDLINE | ID: mdl-28539806

ABSTRACT

Since its first epidemic in the 1940s, retinopathy of prematurity (ROP) has been a challenging illness in neonatology. Higher than physiological oxygen levels impede the development of the immature retinal neuropil and vasculature. Current treatment regimens include cryotherapy, laser photocoagulation, and anti-VEGF agents. Unfortunately, none of these approaches can rescue the normal retinal vasculature, and each has significant safety concerns. The limitations of these approaches have led to new efforts to understand the pathological characteristics in each phase of ROP and to find a safer and more effective therapeutic approach. In the era of stem cell biology and with the need for new treatments for ROP, this review discusses the possible future use of unique populations of proangiogenic cells for therapeutic revascularization of the preterm retina.

5.
Growth Horm IGF Res ; 23(3): 45-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23578754

ABSTRACT

IGFs and their binding proteins have been shown to exhibit both protective and deleterious effects in ocular disease. Recent studies have characterized the expression patterns of different IGFBPs in retinal layers and within the vitreous. IGFBP-3 has roles in vascular protection stimulating proliferation, migration, and differentiation of vascular progenitor cells to sites of injury. IGFBP-3 increases pericyte ensheathment and shows anti-inflammatory effects by reducing microglia activation in diabetes. IGFBP-5 has recently been linked to mediating fibrosis in proliferative vitreoretinopathy but also reduces neovascularization. Thus, the regulatory balance between IGF and IGFBPs can have profound impact on target tissues. This review discusses recent findings of IGF and IGFBP expression in the eye with relevance to different retinopathies.


Subject(s)
Eye Diseases/pathology , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Eye Diseases/metabolism , Humans
6.
Diabetes ; 62(1): 273-82, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23193187

ABSTRACT

In this study, we assessed whether Per2 clock gene-mutant mice exhibit a vascular phenotype similar to diabetes. Per2 (B6.129-Per2(tm1Drw)/J) or wild-type control mice 4 and 12 months of age were used. To evaluate diabetes-like phenotype in Per2 mutant mice, retina was quantified for mRNA expression, and degree of diabetic retinopathy was evaluated. Bone marrow neuropathy was studied by staining femurs for tyrosine hydroxylase (TH) and neurofilament 200 (NF-200). The rate of proliferation and quantification of bone marrow progenitor cells (BMPCs) was performed, and a threefold decrease in proliferation and 50% reduction in nitric oxide levels were observed in Per2 mutant mice. TH-positive nerve processes and NF-200 staining were reduced in Per2 mutant mice. Both retinal protein and mRNA expression of endothelial nitric oxide synthase were decreased by twofold. Other endothelial function genes (VEGFR2, VEGFR1) were downregulated (1.5-2-fold) in Per2 mutant retinas, whereas there was an upregulation of profibrotic pathway mediated by transforming growth factor-ß1. Our studies suggest that Per2 mutant mice recapitulate key aspects of diabetes without the metabolic abnormalities, including retinal vascular damage, neuronal loss in the bone marrow, and diminished BMPC function.


Subject(s)
Bone Marrow/innervation , Diabetes Mellitus/physiopathology , Diabetic Retinopathy/etiology , Period Circadian Proteins/physiology , Animals , Bone Marrow/pathology , Circadian Rhythm , Hematopoietic Stem Cells/physiology , Mice , Mutation , Nitric Oxide/analysis , Nitric Oxide Synthase Type III/analysis , Period Circadian Proteins/genetics , Permeability , Phenotype , Retina/enzymology , Transforming Growth Factor beta1/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
7.
Article in English | MEDLINE | ID: mdl-23267348

ABSTRACT

Diabetes and hyperglycemia create a proinflammatory microenvironment that progresses to microvascular complications such as nephropathy, retinopathy, and neuropathy. Diet-induced insulin resistance is a potential initiator of this change in type 2 diabetes which can increase adipokines and generate a chronic low-grade inflammatory state. Advanced glycation end-products and its receptor, glycation end-products AGE receptor axis, reactive oxygen species, and hypoxia can also interact to worsen complications. Numerous efforts have gained way to understanding the mechanisms of these modulators and attenuation of the inflammatory response, however, effective treatments have still not emerged. The complexity of inflammatory signaling may suggest a need for multi-targeted therapy. This review presents recent findings aimed at new treatment strategies.

8.
Diabetes ; 61(12): 3270-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22891211

ABSTRACT

Endothelial progenitor cells (EPCs), critical for mediating vascular repair, are dysfunctional in a hyperglycemic and/or hypercholesterolemic environment. Their dysfunction contributes to the progression of diabetic macro- and microvascular complications. Activation of "cholesterol-sensing" nuclear receptors, the liver X receptors (LXRα/LXRß), protects against atherosclerosis by transcriptional regulation of genes important in promoting cholesterol efflux and inhibiting inflammation. We hypothesized that LXR activation with a synthetic ligand would correct diabetes-induced EPC dysfunction and improve diabetic retinopathy. Studies were performed in streptozotocin (STZ)-injected DBA/2J mice fed a high-fat Western diet (DBA/STZ/WD) and treated with the LXR agonist GW3965 and in LXRα(-/-), LXRß(-/-), and LXRα/ß(-/-) mice. Retinas were evaluated for number of acellular capillaries and glial fibrillary acidic protein (GFAP) immunoreactivity. Bone marrow EPCs were analyzed for migratory function and gene expression. Compared with vehicle-treated DBA/STZ/WD mice, GW3965 treated mice showed fewer acellular capillaries and reduced GFAP expression. These mice also exhibited enhanced EPC migration and restoration of inflammatory and oxidative stress genes toward nondiabetic levels. LXRα(-/-), LXRß(-/-), and LXRα/ß(-/-) mice developed acellular capillaries and EPC dysfunction similar to the DBA/STZ/WD mice. These studies support a key role for LXR in retinal and bone marrow progenitor dysfunction associated with type 1 diabetes. LXR agonists may represent promising pharmacologic targets for correcting retinopathy and EPC dysfunction.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Orphan Nuclear Receptors/metabolism , Animals , Benzoates/pharmacology , Benzoates/therapeutic use , Benzylamines/pharmacology , Benzylamines/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Liver X Receptors , Male , Mice , Mice, Knockout , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Stem Cells/metabolism , Stem Cells/pathology
9.
PLoS One ; 7(7): e39398, 2012.
Article in English | MEDLINE | ID: mdl-22792172

ABSTRACT

Previously, we showed that insulin growth factor (IGF)-1 binding protein-3 (IGFBP-3), independent of IGF-1, reduces pathological angiogenesis in a mouse model of the oxygen-induced retinopathy (OIR). The current study evaluates novel endothelium-dependent functions of IGFBP-3 including blood retinal barrier (BRB) integrity and vasorelaxation. To evaluate vascular barrier function, either plasmid expressing IGFBP-3 under the regulation of an endothelial-specific promoter or a control plasmid was injected into the vitreous humor of mouse pups (P1) and compared to the non-injected eyes of the same pups undergoing standard OIR protocol. Prior to sacrifice, the mice were given an injection of horseradish peroxidase (HRP). IGFBP-3 plasmid-injected eyes displayed near-normal vessel morphology and enhanced vascular barrier function. Further, in vitro IGFBP-3 protects retinal endothelial cells from VEGF-induced loss of junctional integrity by antagonizing the dissociation of the junctional complexes. To assess the vasodilatory effects of IGFBP-3, rat posterior cerebral arteries were examined in vitro. Intraluminal IGFBP-3 decreased both pressure- and serotonin-induced constrictions by stimulating nitric oxide (NO) release that were blocked by L-NAME or scavenger receptor-B1 neutralizing antibody (SRB1-Ab). Both wild-type and IGF-1-nonbinding mutant IGFBP-3 (IGFBP-3NB) stimulated eNOS activity/NO release to a similar extent in human microvascular endothelial cells (HMVECs). NO release was neither associated with an increase in intracellular calcium nor decreased by Ca(2+)/calmodulin-dependent protein kinase II (CamKII) blockade; however, dephosphorylation of eNOS-Thr(495) was observed. Phosphatidylinositol 3-kinase (PI3K) activity and Akt-Ser(473) phosphorylation were both increased by IGFBP-3 and selectively blocked by the SRB1-Ab or PI3K blocker LY294002. In conclusion, IGFBP-3 mediates protective effects on BRB integrity and mediates robust NO release to stimulate vasorelaxation via activation of SRB1. This response is IGF-1- and calcium-independent, but requires PI3K/Akt activation, suggesting that IGFBP-3 has novel protective effects on retinal and systemic vasculature and may be a therapeutic candidate for ocular complications such as diabetic retinopathy.


Subject(s)
Blood-Retinal Barrier/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Animals , Blood-Retinal Barrier/pathology , Cadherins/genetics , Cadherins/metabolism , Calcium/metabolism , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Claudin-5/genetics , Claudin-5/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fura-2 , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/metabolism , Intercellular Junctions/drug effects , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Male , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nitric Oxide , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Scavenger Receptors, Class B/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/pharmacology
10.
Prog Retin Eye Res ; 31(5): 481-94, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22609081

ABSTRACT

Diabetic retinopathy is the fourth most common cause of blindness in adults. Current therapies, including anti-VEGF therapy, have partial efficacy in arresting the progression of proliferative diabetic retinopathy and diabetic macular edema. This review provides an overview of a novel, innovative approach to viewing diabetic retinopathy as the result of an inflammatory cycle that affects the bone marrow (BM) and the central and sympathetic nervous systems. Diabetes associated inflammation may be the result of BM neuropathy which skews haematopoiesis towards generation of increased inflammatory cells but also reduces production of endothelial progenitor cells responsible for maintaining healthy endothelial function and renewal. The resulting systemic inflammation further impacts the hypothalamus, promoting insulin resistance and diabetes, and initiates an inflammatory cascade that adversely impacts both macrovascular and microvascular complications, including diabetic retinopathy (DR). This review examines the idea of using anti-inflammatory agents that cross not only the blood-retinal barrier to enter the retina but also have the capability to target the central nervous system and cross the blood-brain barrier to reduce neuroinflammation. This neuroinflammation in key sympathetic centers serves to not only perpetuate BM pathology but promote insulin resistance which is characteristic of type 2 diabetic patients (T2D) but is also seen in T1D. A case series of morbidly obese T2D patients with retinopathy and neuropathy treated with minocycline, a well-tolerated antibiotic that crosses both the blood-retina and blood-brain barrier is presented. Our results indicates that minocycine shows promise for improving visual acuity, reducing pain from peripheral neuropathy, promoting weight loss and improving blood pressure control and we postulate that these observed beneficial effects are due to a reduction of chronic inflammation.


Subject(s)
Bone Marrow/innervation , Diabetic Neuropathies/complications , Diabetic Retinopathy/etiology , Animals , Anti-Inflammatory Agents/therapeutic use , Blood Pressure/drug effects , Blood-Retinal Barrier/physiology , Central Nervous System/physiology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Diabetic Retinopathy/drug therapy , Humans , Sympathetic Nervous System/physiology , Visual Acuity/drug effects , Weight Loss/drug effects
11.
Invest Ophthalmol Vis Sci ; 52(11): 8278-86, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21931131

ABSTRACT

PURPOSE: To examine the effect of free insulin-like growth factor (IGF) binding protein-3 (IGFBP-3), independent of the effect of insulin-like growth factors, in modulating retinal vascular permeability. METHODS: We assessed the ability of a form of IGFBP-3 that does not bind IGF-1 (IGFBP-3NB), to regulate the blood retinal barrier (BRB) using two distinct experimental mouse models, laser-induced retinal vessel injury and vascular endothelial growth factor (VEGF)-induced retinal vascular permeability. Additionally, in vitro studies were conducted. In the animal models, BRB permeability was quantified by intravenous injection of fluorescein labeled serum albumin followed by digital confocal image analysis of retinal flat-mounts. Claudin-5 and vascular endothelial-cadherin (VE-cadherin) localization at interendothelial junctions was studied by immunofluorescence. In vitro changes in transendothelial electrical resistance (TEER) and flux of fluorescent dextran in bovine retinal endothelial monolayers (BREC) were measured after IGFBP-3NB treatment. Acid (ASMase) and neutral (NSMase) sphingomyelinase mRNA levels and activity were measured in mouse retinas. RESULTS: Four days postinjury, laser-injured mouse retinas injected with IGFBP-3NB plasmid demonstrated reduced vascular permeability compared with retinas of laser-injured mouse retinas injected with control plasmid. IGFBP-3NB administration resulted in a significant decrease in laser injury-associated increases in ASMase and NSMase mRNA and activity when compared with laser alone treated mice. In vivo, intravitreal injection of IGFBP-3NB reduced vascular leakage associated with intravitreal VEGF injection. IGFBP-3NB partially restored VEGF-induced in vivo permeability and dissociation of claudin-5 and VE-cadherin at junctional complexes. When IGFBP-3NB was applied basally to bovine retinal endothelial cells (BREC) in vitro, TEER increased and macromolecular flux decreased. CONCLUSIONS: Intravitreal administration of IGFBP-3NB preserves junctional integrity in the presence of VEGF or laser injury by reducing BRB permeability in part by modulating sphingomyelinase levels.


Subject(s)
Blood-Retinal Barrier/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Retina/injuries , Retina/metabolism , Retinal Vessels/drug effects , Sphingomyelin Phosphodiesterase/metabolism , Adherens Junctions/metabolism , Animals , Blood-Retinal Barrier/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cattle , Disease Models, Animal , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Lasers/adverse effects , Light Coagulation , Mice , Mice, Inbred C57BL , Plasmids/pharmacology , Recombinant Proteins/pharmacology , Retina/drug effects , Retinal Vessels/metabolism , Sphingomyelin Phosphodiesterase/genetics , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
12.
Curr Diab Rep ; 11(4): 265-74, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21611766

ABSTRACT

Autologous endothelial progenitor cell (EPC) populations represent a novel treatment for therapeutic revascularization and vascular repair for diabetic patients with complications including diabetic retinopathy. Current therapies are applicable to late-stage disease and carry significant side effects, whereas cell-based therapy may provide an alternative by repairing areas of vasodegeneration and reversing ischemia. However, EPCs from diabetic patients with vascular complications are dysfunctional. Moreover, the diabetic environment poses its own challenges and complicates the use of autologous EPCs. Before EPCs become the ideal "cell therapy," the optimal EPC must be determined, any functional dysfunction must be corrected prior to use, and the diabetic milieu will require modification to accept the EPCs. This review describes the rationale for harnessing the vascular reparative properties of EPCs with emphasis on the molecular and phenotypic nature of healthy EPCs, how diabetes alters them, and novel strategies to improve dysfunctional EPCs.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Diabetic Retinopathy/therapy , Animals , Diabetic Retinopathy/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Renin-Angiotensin System/physiology
13.
Am J Pathol ; 178(4): 1517-28, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435441

ABSTRACT

This study was conducted to determine the perivascular cell responses to increased endothelial cell expression of insulin-like growth factor binding protein-3 (IGFBP-3) in mouse retina. The contribution of bone marrow cells in the IGFBP-3-mediated response was examined using green fluorescent protein-positive (GFP(+)) adult chimeric mice subjected to laser-induced retinal vessel occlusion injury. Intravitreal injection of an endothelial-specific IGFBP-3-expressing plasmid resulted in increased differentiation of GFP(+) hematopoietic stem cells (HSCs) into pericytes and astrocytes as determined by immunohistochemical analysis. Administration of IGFBP-3 plasmid to mouse pups that underwent the oxygen-induced retinopathy model resulted in increased pericyte ensheathment and reduced pericyte apoptosis in the developing retina. Increased IGFBP-3 expression reduced the number of activated microglial cells and decreased apoptosis of neuronal cells in the oxygen-induced retinopathy model. In summary, IGFBP-3 increased differentiation of GFP(+) HSCs into pericytes and astrocytes while increasing vascular ensheathment of pericytes and decreasing apoptosis of pericytes and retinal neurons. All of these cytoprotective effects exhibited by IGFBP-3 overexpression can result in a more stable retinal vascular bed. Thus, endothelial expression of IGFBP-3 may represent a physiologic response to injury and may represent a therapeutic strategy for the treatment of ischemic vascular eye diseases, such as diabetic retinopathy and retinopathy of prematurity.


Subject(s)
Apoptosis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Ischemia/pathology , Microglia/metabolism , Neurons/metabolism , Pericytes/metabolism , Retina/injuries , Animals , Astrocytes/cytology , Cell Death , Cell Differentiation , Eye Diseases/pathology , Female , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Pericytes/cytology , Retina/metabolism
14.
Microvasc Res ; 79(3): 207-16, 2010 May.
Article in English | MEDLINE | ID: mdl-20188747

ABSTRACT

Bone-marrow-derived endothelial progenitor cells (EPCs) contribute to angiogenesis-mediated pathological neovascularization, and recent studies have begun to recognize the biological significance of this contribution. This review will discuss the ability of EPCs to contribute to neovascularization in both physiological and pathological conditions. Circulating EPCs were originally identified in 1997 by Asahara as CD34(+) VEGFR2(+) mononuclear cells. These cells differentiated into an endothelial phenotype, expressed endothelial markers, and incorporated into neovessels at sites of ischemia (Asahara et al., 1997). EPCs provide both instructive (release of pro-angiogenic cytokines) and structural (vessel incorporation and stabilization) functions that contribute to the initiation of neo-angiogenesis. EPC populations can be characterized based on surface markers of freshly isolated cells, or they can be described by their in vitro characteristics once placed in culture. However, a major stumbling block to progress in the field has been the lack of consensus among investigators as to the optimal characterization of EPCs. This review intends to address the role of both EPC classes and evaluate how they interact in the setting of pathological angiogenesis. Since the EPCs may be responsible for turning on the "angiogenic switch," strategies have been employed to keep this switch in the "off" position for diseases like cancer, retinopathy, and wet AMD. The expectation is that EPCs will evolve into clinically useful prognostic and predictive tools in cancer and in ocular diseases associated with pathological neovascularization and that targeting this cell type is a key to successful management of patients suffering from diseases associated with pathological neovascularization.


Subject(s)
Endothelial Cells/pathology , Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Stem Cells/pathology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Endothelial Cells/drug effects , Humans , Neoplasms/drug therapy , Neovascularization, Pathologic/physiopathology , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic , Phenotype , Retinal Neovascularization/pathology , Retinal Neovascularization/physiopathology , Stem Cell Niche , Stem Cells/drug effects
15.
EPMA J ; 1(1): 88-100, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-21494317

ABSTRACT

Progressive obliteration of the retinal microvessels is a characteristic of diabetic retinopathy and the resultant retinal ischemia can lead to sight-threatening macular edema, macular ischemia and ultimately preretinal neovascularization. Bone marrow derived endothelial progenitor cells (EPCs) play a critical role in vascular maintenance and repair. There is still great debate about the most appropriate markers that define an EPC. EPCs can be isolated using cell sorting by surface phenotype selection or in vitro cell culture. For freshly isolated cells, EPC cell sorting is heavily dependent on the surface markers used; EPCs can also be isolated by in vitro propagation of heterogeneous mixtures of cells in culture using adhesion to specific substrates and cell growth characteristics. in vitro isolation enables consistent reproducibility and using this approach at least two distinct types of EPCs with different angiogenic properties have been identified from adult peripheral and umbilical cord blood; early EPCs (eEPCs) and late outgrowth endothelial progenitor cells (OECs). Emerging studies demonstrate the potential of these cells in revascularization of ischemic/injured retina in animal models of retinal disease. Since ischemic retinopathies are leading causes of blindness, they are a potential disease target for EPC-based therapy. In this chapter, we summarize the current knowledge about EPCs and discuss the possibility of cellular therapy for treatment of diabetic macular ischemia and the vasodegenerative phase of diabetic retinopathy. We also report current pharmacological options that can be utilized to correct diabetes associated defects in EPCs so as to enhance the therapeutic utility of these cells.

16.
Expert Rev Endocrinol Metab ; 5(1): 29-37, 2010 Jan.
Article in English | MEDLINE | ID: mdl-23678364

ABSTRACT

Progressive obliteration of the retinal microvessels is a characteristic of diabetic retinopathy. The resultant retinal ischemia leads to sight-threatening neovascularization and macular edema. Bone marrow-derived endothelial progenitor cells play a critical role in vascular maintenance and repair and forms the basis of cellular therapy for revascularization of ischemic myocardium and ischemic limbs. Emerging studies show potential of these cells in revascularization of ischemic retina and this review summarizes this possibility. We also report current pharmacological options to correct diabetes-associated defects in endothelial progenitor cells for their therapeutic transfer.

17.
Invest Ophthalmol Vis Sci ; 51(5): 2697-704, 2010 May.
Article in English | MEDLINE | ID: mdl-20007826

ABSTRACT

PURPOSE: Modulators of angiogenesis typically work in an orchestrated manner. The authors examined the interaction between insulinlike growth factor (IGF)-1, vascular endothelial growth factor (VEGF), and stromal derived factor (SDF)-1 in vivo and in vitro using angiogenesis models. METHODS: The angiogenic effect of SDF-1, alone or in combination with IGF-1 and VEGF, was assessed in human lung microvascular endothelial cells using capillary tube formation and thymidine incorporation. Immunohistochemical analysis for CD31, SDF-1, and CXCR4 was performed on mouse eyes 2 weeks after the initiation of laser rupture of Bruch's membrane, a choroidal neovascularization (CNV) model. CXCR4 antagonist and CXCR4 blocking antibody were tested on inhibition of CNV lesion size in this model. Real-time PCR was used to determine mRNA levels for SDF-1, VEGF, IGF-1, and their cognate receptors in the retinal pigment epithelium/choroid complex of mice that underwent this CNV model. RESULTS: IGF-1 and VEGF demonstrated an additive effect on SDF-1-induced in vitro angiogenesis. CXCR4 immunoreactivity was present in both normal and laser-injured mice at the laser burn site and at the ganglion cell layer, the anterior portion of the inner nuclear layer, photoreceptors, and choroidal stroma. SDF-1 was observed in identical locations but was not seen in photoreceptors. mRNA levels for SDF-1, VEGF, and IGF-1 and their receptors were increased after laser injury. CXCR4-neutralizing antibody reduced neovascularization when injected subretinally but not intraperitoneally or intravitreally. CONCLUSIONS: The potent proangiogenic factors IGF-1 and VEGF both stimulate SDF-1-induced angiogenesis. Local inhibition of CXCR4 is required for an antiangiogenic effect in CNV lesions.


Subject(s)
Angiogenic Proteins/pharmacology , Choroidal Neovascularization/metabolism , Endothelium, Vascular/drug effects , Insulin-Like Growth Factor I/pharmacology , Paracrine Communication/physiology , Receptors, CXCR4/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Chemokine CXCL12/genetics , Chemokine CXCL12/pharmacology , Choroidal Neovascularization/pathology , Disease Models, Animal , Endothelium, Vascular/pathology , Humans , Injections , Injections, Intraperitoneal , Insulin-Like Growth Factor I/genetics , Laser Coagulation , Lung/cytology , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/metabolism , Receptors, CXCR4/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vitreous Body
18.
Circ Res ; 105(9): 897-905, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19762684

ABSTRACT

RATIONALE: Insulin-like growth factor binding protein (IGFBP)-3 modulates vascular development by regulating endothelial progenitor cell (EPC) behavior, specifically stimulating EPC cell migration. This study was undertaken to investigate the mechanism of IGFBP-3 effects on EPC function and how IGFBP-3 mediates cytoprotection following vascular injury. OBJECTIVE: To examine the mechanism of IGFBP-3-mediated repair following vascular injury. METHODS AND RESULTS: We used 2 complementary vascular injury models: laser occlusion of retinal vessels in adult green fluorescent protein (GFP) chimeric mice and oxygen-induced retinopathy in mouse pups. Intravitreal injection of IGFBP-3-expressing plasmid into lasered GFP chimeric mice stimulated homing of EPCs, whereas reversing ischemia induced increases in macrophage infiltration. IGFBP-3 also reduced the retinal ceramide/sphingomyelin ratio that was increased following laser injury. In the OIR model, IGFBP-3 prevented cell death of resident vascular endothelial cells and EPCs, while simultaneously increasing astrocytic ensheathment of vessels. For EPCs to orchestrate repair, these cells must migrate into ischemic tissue. This migratory ability is mediated, in part, by endogenous NO generation. Thus, we asked whether the migratory effects of IGFBP-3 were attributable to stimulation of NO generation. IGFBP-3 increased endothelial NO synthase expression in human EPCs leading to NO generation. IGFBP-3 exposure also led to the redistribution of vasodilator-stimulated phosphoprotein, an NO regulated protein critical for cell migration. IGFBP-3-mediated NO generation required high-density lipoprotein receptor activation and stimulation of phosphatidylinositol 3-kinase/Akt pathway. CONCLUSION: These studies support consideration of IGFBP-3 as a novel agent to restore the function of injured vasculature and restore NO generation.


Subject(s)
Cell Movement , Endothelial Cells/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Nitric Oxide/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Cell Adhesion Molecules/metabolism , Cell Death , Cell Proliferation , Cells, Cultured , Ceramides/metabolism , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Cytoprotection , Disease Models, Animal , Endothelial Cells/pathology , Female , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Humans , Infant, Newborn , Insulin-Like Growth Factor Binding Protein 3/genetics , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/pathology , Retinal Neovascularization/physiopathology , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/physiopathology , Scavenger Receptors, Class B/metabolism , Signal Transduction , Sphingomyelins/metabolism , Stem Cells/pathology , Vasodilation
19.
Invest Ophthalmol Vis Sci ; 49(11): 5094-102, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18599562

ABSTRACT

PURPOSE: To define the molecular pharmacology underlying the antiangiogenic effects of nonpeptide imidazolidine-2,4-dione somatostatin receptor agonists (NISAs) and evaluate the efficacy of NISA in ocular versus systemic delivery routes in ocular disease models. METHODS: Functional inhibitory effects of the NISAs and the somatostatin peptide analogue octreotide were evaluated in vitro by chemotaxis, proliferation, and tube-formation assays. The oxygen-induced retinopathy (OIR) model and the laser model of choroidal neovascularization (CNV) were used to test the in vivo efficacy of NISAs. Transscleral permeability of a candidate NISA was also measured. RESULTS: NISAs inhibited growth factor-induced HREC proliferation, migration and tube formation with submicromolar potencies (IC(50), 0.1-1.0 microM) comparable to octreotide. In the OIR model, systemic administration of the NISAs RFE-007 and RFE-011 inhibited retinal neovascularization in a dose-dependent manner, comparable to octreotide. In the CNV model, intravitreal RFE-011 resulted in a 56% reduction (P < 0.01) in CNV lesion area, whereas systemic administration resulted in a 35% reduction (P < 0.05) in lesion area. RFE-011 demonstrated transscleral penetration. CONCLUSIONS: Micromolar concentrations of octreotide and NISAs are necessary for antiangiogenic effects, whereas nanomolar concentrations are effective for endocrine inhibition. This suggests that the antiangiogenic activity of NISAs and octreotide is mediated by an overall much less efficient downstream coupling mechanism than is growth hormone release. As a result, the intravitreal or transscleral route of administration should be seriously considered for future clinical studies of SSTR2 agonists used for treatment of ocular neovascularization to ensure efficacious concentrations in the target retinal and choroidal tissue.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Gene Expression/drug effects , Imidazolidines/agonists , RNA/genetics , Receptors, Somatostatin/genetics , Retinal Neovascularization/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/therapeutic use , Autoradiography , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Humans , Injections , Mice , Mice, Inbred C57BL , Octreotide/administration & dosage , Octreotide/pharmacokinetics , Octreotide/therapeutic use , Ophthalmic Solutions , Polymerase Chain Reaction , Rabbits , Receptors, Somatostatin/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Sclera , Treatment Outcome , Vitreous Body
20.
J Pharmacol Exp Ther ; 324(3): 894-901, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18083913

ABSTRACT

The alpha(V) integrins are key receptors involved in mediating cell migration and angiogenesis. In age-related macular degeneration (AMD) and diabetic retinopathy, angiogenesis plays a critical role in the loss of vision. These ocular vasculopathies might be treatable with a suitable alpha(V) antagonist, and an oral drug would offer a distinct advantage over current therapies. (3,S,beta,S)-1,2,3,4-Tetrahydro-beta-[[1-[1-oxo-3-(1,5,6,7-tetrahydro-1,8-naphthyridin-2-yl)propyl]-4-piperidinyl]methyl]-3-quinolinepropanoic acid (JNJ-26076713) is a potent, orally bioavailable, nonpeptide alpha(V) antagonist derived from the arginine-glycine-asparagine binding motif in the matrix protein ligands (e.g., vitronectin). This compound inhibits alpha(V)beta(3) and alpha(V)beta(5) binding to vitronectin in the low nanomolar range, it has excellent selectivity over integrins alpha(IIb)beta(3) and alpha(5)beta(1), and it prevents adhesion to human, rat, and mouse endothelial cells. JNJ-26076713 blocks cell migration induced by vascular endothelial growth factor, fibroblast growth factor (FGF), and serum, and angiogenesis induced by FGF in the chick chorioallantoic membrane model. JNJ-26076713 is the first alpha(V) antagonist reported to inhibit retinal neovascularization in an oxygen-induced model of retinopathy of prematurity after oral administration. In diabetic rats, orally administered JNJ-26076713 markedly inhibits retinal vascular permeability, a key early event in diabetic macular edema and AMD. Given this profile, JNJ-26076713 represents a potential therapeutic candidate for the treatment of age-related macular degeneration, macular edema, and proliferative diabetic retinopathy.


Subject(s)
Capillary Permeability/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Integrin alphaV/metabolism , Naphthyridines/administration & dosage , Naphthyridines/pharmacokinetics , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Retinal Neovascularization/metabolism , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Animals , Biological Availability , Capillary Permeability/drug effects , Cell Line , Chick Embryo , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/drug therapy , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Naphthyridines/chemistry , Pregnancy , Quinolines/chemistry , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Retinal Neovascularization/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...