Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Transl Vis Sci Technol ; 11(4): 24, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35486039

ABSTRACT

Purpose: To improve outcomes for subretinal implantation surgery in pigs. Methods: Analysis of variables affecting the success of subretinal implantation surgery was performed on videos of 37 surgeries. Ex vivo experiments were conducted to measure intraocular pressure (IOP) and test various prototyped implanters for effectiveness at maintaining IOP. Results: A video analysis revealed a prolonged sclerotomy open time owing to a combination of uncontrolled bleeding and excessive fluid outflow often resulting in retinal prolapse. Precauterization of the choroid before full-thickness sclerotomy (n = 10) resulted in a reduced incidence of uncontrolled bleeding from 39.1% (9/23) versus 0% (0/10) (P = 0.005) and improved implantation success from 73% to 90%. An ex vivo analysis of the IOP revealed a mean decrease in the IOP from 30.2 ± 3.0 mm Hg to 5.0 ± 2.1 mm Hg after a fully penetrating sclerotomy. To address this situation, we produced a series of plugs that integrated with a custom implant insertion device to seal the sclerotomy during implantation. The use of the plugs was cumbersome, however, and so we opted instead to increase the width of the inserter tip to fill the open sclerotomy. This improved device restored and maintained IOP during implantation (27.1 ± 1.9 mm Hg). Combined with precauterization the improved inserter resulted in 100% successful implantation (n = 4). Conclusions: For subretinal implantation in pigs, a modified procedure to precauterize the choroid before sclerotomy combined with an instrument that better fills the scleral opening decreases bleeding, hypotony, and open sclerotomy time, improving the success rate. Translational Relevance: Better management of IOP and bleeding from a sclerotomy will improve implant-based therapies.


Subject(s)
Glaucoma Drainage Implants , Glaucoma, Open-Angle , Animals , Glaucoma, Open-Angle/surgery , Intraocular Pressure , Swine , Tonometry, Ocular , Treatment Outcome
2.
J Biomed Mater Res A ; 109(11): 2357-2368, 2021 11.
Article in English | MEDLINE | ID: mdl-33973708

ABSTRACT

Fibrin is a degradable biopolymer with an excellent clinical safety profile. Use of higher mechanical strength fibrin hydrogels is limited by the rapid rate of fibrin polymerization. We recently demonstrated the use of higher mechanical strength (fibrinogen concentrations >30 mg/ml) fibrin scaffolds for surgical implantation of cells. The rapid polymerization of fibrin at fibrinogen concentrations impaired our ability to scale production of these fibrin scaffolds. We serendipitously discovered that the azo dye Trypan blue (TB) slowed fibrin gelation kinetics allowing for more uniform mixing of fibrinogen and thrombin at high concentrations. A screen of closely related compounds identified similar activity for Evans blue (EB), an isomer of TB. Both TB and EB exhibited a concentration dependent increase in clot time, though EB had a larger effect. While gelation time was increased by TB or EB, overall polymerization time was unaffected. Scanning electron microscopy showed similar surface topography, but transmission electron microscopy showed a higher cross-linking density for gels formed with TB or EB versus controls. Based on these data we conclude that addition of TB or EB during thrombin mediated fibrin polymerization slows the initial gelation time permitting generation of larger more uniform fibrin hydrogels with high-mechanical strength.


Subject(s)
Azo Compounds/chemistry , Fibrin/chemistry , Hydrogels/chemistry , Models, Chemical , Kinetics
3.
PLoS One ; 15(1): e0227641, 2020.
Article in English | MEDLINE | ID: mdl-31929571

ABSTRACT

Retinal pigment epithelium (RPE) transplantation for the treatment of macular degeneration has been studied for over 30 years. Human clinical trials have demonstrated that RPE monolayers exhibit improved cellular engraftment and survival compared to single cell suspensions. The use of a scaffold facilitates implantation of a flat, wrinkle-free, precisely placed monolayer. Scaffolds currently being investigated in human clinical trials are non-degradable which results in the introduction of a chronic foreign body. To improve RPE transplant technology, a degradable scaffold would be desirable. Using human fibrin, we have generated scaffolds that support the growth of an RPE monolayer in vitro. To determine whether these scaffolds are degraded in vivo, we developed a surgical approach that delivers a fibrin hydrogel implant to the sub-retinal space of the pig eye and determined whether and how fast they degraded. Using standard ophthalmic imaging techniques, the fibrin scaffolds were completely degraded by postoperative week 8 in 5 of 6 animals. Postmortem histologic analysis confirmed the absence of the scaffold from the subretinal space at 8 weeks, and demonstrated the reattachment of the neurosensory retina and a normal RPE-photoreceptor interface. When mechanical debridement of a region of native RPE was performed during implantation surgery degradation was accelerated and scaffolds were undetectable by 4 weeks. These data represent the first in situ demonstration of a fully biodegradable scaffold for use in the implantation of RPE and other cell types for treatment of macular degeneration and other retinal degenerative diseases.


Subject(s)
Absorbable Implants , Fibrin , Retina/surgery , Tissue Scaffolds , Animals , Equipment Design , Female , Retina/cytology , Retina/diagnostic imaging , Retinal Degeneration/surgery , Sus scrofa
4.
Plast Reconstr Surg ; 143(5): 993e-1002e, 2019 05.
Article in English | MEDLINE | ID: mdl-31033820

ABSTRACT

BACKGROUND: Cell-seeded biomaterial scaffolds have been proposed as a future option for reconstruction of bone tissue. The ability to generate larger, functional volumes of bone has been a challenge that may be addressed through the use of perfusion bioreactors. In this study, the authors investigated use of a tubular perfusion bioreactor system for the growth and differentiation of bone marrow stromal (mesenchymal stem) cells seeded onto fibrin, a highly angiogenic biomaterial. METHODS: Cells were encapsulated within fibrin beads and cultured either within a tubular perfusion bioreactor system or statically for up to 14 days. Scaffolds were analyzed for osteogenic differentiation. A rodent cranial defect model (8-mm diameter) was used to assess the bone regeneration of scaffolds cultured in the bioreactor, statically, or used immediately after formation. Immunohistochemistry was used to visualize CD31 vessel density. Micro-computed tomographic imaging was used to visualize mineral formation within the defect volume. RESULTS: Tubular perfusion bioreactor system-cultured samples showed significantly greater osteodifferentiation, indicated by an increase in VEGF expression and mineral deposition, compared with statically cultured samples. Increased expression of OPN, RUNX2, VEGF, and CD90 was seen over time in both culture methods. After implantation, bioreactor samples exhibited greater bone formation and vessel density compared with all other groups. Analysis of micro-computed tomographic images showed full union formation through the greatest diameter of the defect in all bioreactor samples and the highest levels of mineralized volume after 8 weeks. CONCLUSION: Mesenchymal stem cells encapsulated in fibrin beads and cultured in the tubular perfusion bioreactor system resulted in increased vascularization and mineralized tissue formation in vivo relative to static culture.


Subject(s)
Bone Regeneration , Cell Culture Techniques/methods , Osteogenesis/physiology , Skull/injuries , Tissue Scaffolds , Animals , Bioreactors , Bone Marrow Cells/physiology , Cell Culture Techniques/instrumentation , Cell Differentiation/physiology , Cells, Cultured , Craniocerebral Trauma/surgery , Disease Models, Animal , Humans , Male , Mesenchymal Stem Cells/physiology , Orthopedic Procedures/instrumentation , Orthopedic Procedures/methods , Perfusion/methods , Rats , Rats, Sprague-Dawley , Plastic Surgery Procedures/instrumentation , Plastic Surgery Procedures/methods , Skull/surgery , Treatment Outcome
5.
Stem Cells Transl Med ; 8(6): 512-521, 2019 06.
Article in English | MEDLINE | ID: mdl-30768863

ABSTRACT

Human fibrin hydrogels are a popular choice for use as a biomaterial within tissue engineered constructs because they are biocompatible, nonxenogenic, autologous use compatible, and biodegradable. We have recently demonstrated the ability to culture induced pluripotent stem cell (iPSC)-derived retinal pigment epithelium on fibrin hydrogels. However, iPSCs themselves have relatively few substrate options (e.g., laminin) for expansion in adherent cell culture for use in cell therapy. To address this, we investigated the potential of culturing iPSCs on fibrin hydrogels for three-dimensional applications and further examined the use of fibrinogen, the soluble precursor protein, as a coating substrate for traditional adherent cell culture. iPSCs successfully adhered to and proliferated on fibrin hydrogels. The two-dimensional culture with fibrinogen allows for immediate adaption of culture models to a nonxenogeneic model. Similarly, multiple commercially available iPSC lines adhered to and proliferated on fibrinogen coated surfaces. iPSCs cultured on fibrinogen expressed similar levels of the pluripotent stem cell markers SSea4 (98.7% ± 1.8%), Oct3/4 (97.3% ± 3.8%), TRA1-60 (92.2% ± 5.3%), and NANOG (96.0% ± 3.9%) compared with iPSCs on Geltrex. Using a trilineage differentiation assay, we found no difference in the ability of iPSCs grown on fibrinogen or Geltrex to differentiate to endoderm, mesoderm, or ectoderm. Finally, we demonstrated the ability to differentiate iPSCs to endothelial cells using only fibrinogen coated plates. On the basis of these data, we conclude that human fibrinogen provides a readily available and inexpensive alternative to laminin-based products for the growth, expansion, and differentiation of iPSCs for use in research and clinical cell therapy applications. Stem Cells Translational Medicine 2019;8:512-521.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/drug effects , Fibrin/pharmacology , Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibrin/chemistry , Fibrinogen/analysis , Fibrinogen/isolation & purification , Humans , Hydrogels/chemistry , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism
6.
Sci Rep ; 8(1): 4487, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540715

ABSTRACT

Autosomal recessive bestrophinopathy (ARB) is caused by mutations in the gene BEST1 which encodes bestrophin 1 (Best1), an anion channel expressed in retinal pigment epithelial (RPE) cells. It has been hypothesized that ARB represents the human null phenotype for BEST1 and that this occurs due to nonsense mediated decay (NMD). To test this hypothesis, we generated induced pluripotent stem cells (iPSCs) from a patient with ARB and her parents. After differentiation to retinal pigment epithelial (iPSC-RPE) cells, both BEST1 mRNA and Best1 protein expression were compared to controls. BEST1 mRNA expression levels, determined by quantitative PCR, were similar in ARB iPSC-RPE, parental cells, and genetically unrelated controls. Western blotting revealed that CRALBP and RPE65 were expressed within the range delineated by unrelated controls in iPSC-RPE from the ARB donor and her parents. Best1 protein was detected in different clones of ARB iPSC-RPE, but at reduced levels compared to all controls. When tested for the ability to phagocytose photoreceptor outer segments, ARB iPSC-RPE exhibited impaired internalization. These data suggest that impaired phagocytosis is a trait common to the bestrophinopathies. Furthermore, ARB is not universally the result of NMD and ARB, in this patient, is not due to the absence of Best1.


Subject(s)
Bestrophins/genetics , Eye Diseases, Hereditary/genetics , Gene Expression , Genes, Recessive , Induced Pluripotent Stem Cells/metabolism , Mutation , Phagocytosis/genetics , Retinal Diseases/genetics , Adolescent , Alleles , Bestrophins/metabolism , Cell Differentiation , Cell Line , Eye Diseases, Hereditary/diagnosis , Female , Fluorescein Angiography , Humans , Induced Pluripotent Stem Cells/cytology , Phenotype , Retinal Diseases/diagnosis , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism
7.
Transl Vis Sci Technol ; 7(6): 39, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30619659

ABSTRACT

PURPOSE: We measure and compare surgical devices using an ex vivo, temperature-controlled, choroidal incision model during thermal energy transfer with a high-resolution infrared camera. METHODS: Ex vivo porcine choroidal tissue specimens (n = 516) were isolated and placed on a temperature-regulated (37°C) perfusion platform. We tested the pulsed electron avalanche knife (PEAK), micropulse laser (MpL), continuous laser (CL), and bipolar cautery (BpC) at three energy settings (11 [low], 45 [medium], and 134 [high] mJ/mm). Each device was clamped to a stationary mechanical arm. Movement of tissue specimens beneath the surgical device was achieved using a stepping motor-driven x-y table. An infrared video camera measured orthogonal temperature variation in the surrounding tissue. RESULTS: Increased power resulted in greater lateral thermal spread using all modalities (P < 0.001). Mean (standard deviation) lateral thermal spread at low energy was smallest for the MpL at 0.0 (0.01) mm (P < 0.001), whereas BpC had the least collateral tissue damage at medium and high energies (0.02 [0.08] and 0.34 [0.22] mm, respectively; P < 0.001). Fluidics of the ex vivo system may limit thermal spread. The PEAK had the greatest thermal spread across all energy groups (P < 0.001), with clinically relevant variation between disposable blades. CONCLUSIONS: Our ex vivo model enabled direct comparison of threshold thermal tissue injury across four devices. MpL and BpC showed the least thermal damage. PEAK had a higher variation in energy delivery, but also has the advantage of more effective tissue cutting. TRANSLATIONAL RELEVANCE: Our ex vivo surgical device analysis provides thermal tissue injury predictions for choroidal surgery.

8.
Acta Biomater ; 67: 134-146, 2018 02.
Article in English | MEDLINE | ID: mdl-29233750

ABSTRACT

Recent phase 1 trials of embryonic stem cell and induced pluripotent stem cell (iPSCs) derived RPE transplants for the treatment of macular degeneration have demonstrated the relative safety of this process. However, there is concern over clumping, thickening, folding, and wrinkling of the transplanted RPE. To deliver a flat RPE monolayer, current phase 1 trials are testing synthetic substrates for RPE transplantation. These substrates, however, cause localized inflammation and fibrosis in animal models due to long degradation times. Here we describe the use of thin fibrin hydrogels as a support material for the transplantation of RPE. Fibrin was formed into a mechanically rigid support that allow for easy manipulation with standard surgical instruments. Using fibrinolytic enzymes, fibrin hydrogels were degraded on the scale of hours. The rate of degradation could be controlled by varying the fibrinolytic enzyme concentration used. RPE cells degraded fibrin spontaneously. To preserve the fibrin support during differentiation of iPSCs to RPE, media was supplemented with the protease inhibitor aprotinin. iPSC-RPE on fibrin gels remained viable, generated monolayers with characteristic cobblestone appearance and dark pigmentation, and expressed mRNA and protein markers characteristic of RPE in the eye. Following differentiation of the cells, addition of fibrinolytic enzymes fully and rapidly degraded the fibrin support leaving behind an intact, viable iPSC-RPE monolayer. In conclusion, human fibrin hydrogels provide a xeno-free support on which iPSCs can be differentiated to RPE cells for transplant which can be rapidly degraded under controlled conditions using fibrinolytic enzymes without adverse effects to the cells. STATEMENT OF SIGNIFICANCE: Stem cell-derived retinal pigment epithelial (RPE) cell transplantation is currently in phase 1 clinical trials for macular degeneration (MD). A major obstacle in these studies is delivering the RPE as a living, flat sheets without leaving behind foreign materials in the retina. Here we investigate the suitability of using hydrogels made from human blood-derived proteins for RPE transplant. Our data shows that these fibrin hydrogels are rigid enough for use in surgery, support growth of stem cell-derived RPE, and are easily degraded within hours without damage to the RPE sheet. These fibrin hydrogels offer a promising solution to transplant RPE for patients with MD.


Subject(s)
Fibrin/chemistry , Hydrogels/chemistry , Retinal Pigment Epithelium/transplantation , Aprotinin/pharmacology , Cells, Cultured , Female , Gels/chemistry , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Kinetics , Phenotype , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Young Adult
9.
J Ocul Pharmacol Ther ; 33(8): 574-581, 2017 10.
Article in English | MEDLINE | ID: mdl-28686538

ABSTRACT

PURPOSE: To validate the increase in intraocular pressure (IOP) caused by soluble adenylyl cyclase (sAC) inhibitors and determine reasons behind variation in IOP measurements performed by tonometry. METHODS: C57BL/6J mice were administered DMSO solubilized sAC inhibitors (KH7 or LRE-1) by intraperitoneal injection. Two hours post-treatment, mice were anesthetized with avertin or ketamine/xylazine/acepromazine (KXA). IOP was measured by a rebound tonometer or direct cannulation of the anterior chamber. Spectral-domain optical coherence tomography was used to measure anterior chamber depth and corneal thickness in live mice. Outflow facility was measured in perfused, enucleated mouse eyes. RESULTS: Compared with DMSO controls, KH7 treatment caused an increased IOP in avertin- and KXA-anesthetized mice when measured by direct cannulation [avertin: 14.4 ± 2.1 mmHg vs. 11.1 ± 1.0 mmHg (P = 0.003); KXA: 14.4 ± 1.0 mmHg vs. 11.3 ± 0.8 mmHg (P < 0.001)] and tonometry [avertin: 10.8 ± 1.4 mmHg vs. 7.4 ± 0.6 mmHg (P < 0.001); KXA: 11.9 ± 0.9 mmHg vs. 10.3 ± 1.7 mmHg (P = 0.283)]. However, treatment with KH7 in nonanesthetized mice showed a significant decrease in IOP measured by tonometry and compared with DMSO-treated animals [13.1 ± 2.6 mmHg vs. 15.6 ± 0.5 mmHg (P = 0.003)]. Both KH7- and DMSO-treated groups anesthetized with avertin showed increased corneal thickness, whereas KH7-treated mice anesthetized with KXA exhibited a shallower anterior chamber compared with untreated mice. KH7 decreased outflow facility by 85.1% in nonanesthetized, enucleated eyes (P < 0.003). CONCLUSIONS: Systemically administered DMSO and anesthesia have significant effects on anterior chamber characteristics, resulting in altered IOP readings measured by tonometry. In the presence of DMSO and anesthesia, tonometry IOP readings should be confirmed with direct cannulation.


Subject(s)
Adenylyl Cyclase Inhibitors/pharmacology , Anesthetics/administration & dosage , Intraocular Pressure/drug effects , Tonometry, Ocular/methods , Acepromazine/administration & dosage , Acepromazine/pharmacology , Anesthetics/pharmacology , Animals , Anterior Chamber/metabolism , Catheterization , Ethanol/administration & dosage , Ethanol/analogs & derivatives , Ethanol/pharmacology , Female , Humans , Injections, Intraperitoneal , Ketamine/administration & dosage , Ketamine/pharmacology , Mice , Mice, Inbred C57BL , Tomography, Optical Coherence , Xylazine/administration & dosage , Xylazine/pharmacology
10.
Sensors (Basel) ; 15(9): 23886-902, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26393602

ABSTRACT

Enhanced vascularization at sensor interfaces can improve long-term function. Fibrin, a natural polymer, has shown promise as a biomaterial for sensor coating due to its ability to sustain endothelial cell growth and promote local vascularization. However, the culture of cells, particularly endothelial cells (EC), within 3D scaffolds for more than a few days is challenging due to rapid loss of EC viability. In this manuscript, a robust method for developing fibrin microbead scaffolds for long-term culture of encapsulated ECs is described. Fibrin microbeads are formed using sodium alginate as a structural template. The size, swelling and structural properties of the microbeads were varied with needle gauge and composition and concentration of the pre-gel solution. Endothelial colony-forming cells (ECFCs) were suspended in the fibrin beads and cultured within a perfusion bioreactor system. The perfusion bioreactor enhanced ECFCs viability and genome stability in fibrin beads relative to static culture. Perfusion bioreactors enable 3D culture of ECs within fibrin beads for potential application as a sensor coating.


Subject(s)
Biosensing Techniques , Colony-Forming Units Assay , Fibrin/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Microspheres , Neovascularization, Physiologic/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Comet Assay , DNA Damage , Humans , Staining and Labeling
11.
Ophthalmic Genet ; 35(2): 112-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23521024

ABSTRACT

BACKGROUND: Familial exudative vitreoretinopathy (FEVR) is a genetic disease caused by abnormal retinal vascular development. New additional genetic loci for FEVR have recently been identified. Microduplication of 22q11.2 has been reported with a heterogeneous phenotype and microdeletion of 22q11.2 has been associated with FEVR. We describe a case of a girl with microduplication of 22q11.2 and falciform macular folds. MATERIALS AND METHODS: The infant and first-degree relatives were examined. A dilated fundus examination was performed. Genetic screening was done by chromosomal microarray analysis and confirmed by fluorescent in situ hybridization (FISH). RESULTS: Bilateral macular folds were found with temporal fibrosis in the proband. A chromosomal microarray revealed a 2.21 Mb microduplication of the 22q11.2 region. CONCLUSION: This is the first report to associate microduplication of 22q11.2 with macular folds, supporting the potential for a FEVR locus on chromosome 22q11.2. We encourage full ophthalmological examination for patients with microduplication of 22q11.2 to identify ocular associations.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Duplication/genetics , DiGeorge Syndrome/genetics , Genetic Diseases, X-Linked/genetics , Retinal Diseases/genetics , Vitreoretinopathy, Proliferative/genetics , Abnormalities, Multiple/diagnosis , Chromosomes, Human, Pair 22/genetics , DiGeorge Syndrome/diagnosis , Exudates and Transudates , Familial Exudative Vitreoretinopathies , Female , Genetic Diseases, X-Linked/diagnosis , Humans , Infant , Microarray Analysis , Microcephaly/diagnosis , Microcephaly/genetics , Retinal Diseases/diagnosis , Vitreoretinopathy, Proliferative/diagnosis
12.
Ther Deliv ; 4(3): 327-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23442080

ABSTRACT

Therapeutic stimulation of vessel growth to improve tissue perfusion has shown promise in many regenerative medicine and tissue engineering applications. Alginate-based biomaterial systems have been investigated for growth factor and/or cell delivery as tools for modulating vessel assembly. Growth factor encapsulation allows for a sustained release of protein and protection from degradation. Implantation of growth factor-loaded alginate constructs typically shows an increase in capillary density but without vascular stabilization. Delivery of multiple factors may improve these outcomes. Cell delivery approaches focus on stimulating vascularization either via cell release of soluble factors, cell proliferation and incorporation into new vessels or alginate prevascularization prior to implantation. These methods have shown some promise but routine clinical application has not been achieved. In this review, current research on the application of alginate for therapeutic neovascularization is presented, shortcomings are addressed and the future direction of these systems discussed.


Subject(s)
Alginates/administration & dosage , Drug Delivery Systems , Neovascularization, Physiologic/drug effects , Alginates/chemistry , Animals , Fibroblast Growth Factor 1/administration & dosage , Fibroblast Growth Factor 2/administration & dosage , Glucuronic Acid/administration & dosage , Glucuronic Acid/chemistry , Hepatocyte Growth Factor/administration & dosage , Hexuronic Acids/administration & dosage , Hexuronic Acids/chemistry , Humans , Tissue Engineering , Vascular Endothelial Growth Factor A/administration & dosage
13.
Invest Ophthalmol Vis Sci ; 52(3): 1336-44, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21212173

ABSTRACT

PURPOSE: Blood-retinal barrier [BRB] breakdown, characteristic of diabetic retinopathy (DR), is believed to depend on inflammation and apoptosis. Retinal inflammation is almost completely suppressed in the absence of TNFα, which is also associated with apoptosis. This study was conducted to determine the role of TNFα in these diabetic complications. METHODS: Diabetes was induced with streptozotocin in Tnfa knockout (KO) mice, to provide a chemical model of diabetes, and Tnfa (KO) mice were crossed with Ins2(Akita) mice to generate a genetic model, with both models being devoid of TNFα. The BRB was assessed at 1, 1.5, 3, and 6 months. Leukostasis was assessed using FITC-conjugated ConA to label leukocytes. Apoptosis was assessed with TUNEL and activated caspase-3 staining. PECAM1 identified endothelial cells, and SMA identified pericytes. RESULTS: At 1 month of diabetes, the absence of TNFα had no effect on DR-associated BRB breakdown, even though it prevented retinal leukostasis, demonstrating that neither TNFα nor inflammation is essential for early BRB breakdown in DR in either model of diabetes. At 3 months of diabetes, BRB breakdown was significantly suppressed and at 6 months, it was completely prevented in the absence of TNFα in both models, showing that TNFα is essential for progressive BRB breakdown. DR-mediated apoptosis in the retina, which appears to involve endothelial cells, pericytes, and neurons, was inhibited in the absence of TNFα in both models. CONCLUSIONS: Although neither TNFα nor inflammation is necessary for early BRB breakdown in DR, TNFα is critical for later complications and would be a good therapeutic target for the prevention of the progressive BRB breakdown, retinal leukostasis, and apoptosis associated with DR.


Subject(s)
Apoptosis , Blood-Retinal Barrier/metabolism , Diabetic Retinopathy/metabolism , Leukostasis/prevention & control , Retinal Neurons/pathology , Retinal Vessels/pathology , Tumor Necrosis Factor-alpha/physiology , Actins/metabolism , Animals , Capillary Permeability/physiology , Caspase 3/metabolism , Cell Survival , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/pathology , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique, Indirect , Gene Silencing/physiology , Genotype , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...