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










Publication year range
1.
PLoS One ; 16(9): e0258006, 2021.
Article in English | MEDLINE | ID: mdl-34587219

ABSTRACT

Primary cultures of human corneal endothelial cells (HCECs) are an important model system for studying the pathophysiology of corneal endothelium. The purpose of this study was to identify and validate an optimal primary culture model of normal and Fuchs endothelial corneal dystrophy (FECD) endothelial cells by comparing cell morphology and marker expression under different media conditions to in vivo donor tissues. Primary and immortalized HCECs, isolated from normal and FECD donors, were cultured in proliferation media (Joyce, M4, Bartakova) alone or sequentially with maturation media (F99, Stabilization 1, M5). CD56, CD73 and CD166 expressions were quantified in confluent and matured cell lines by flow cytometry. HCECs that were allowed to proliferate in Joyce's medium followed by maturation in low-mitogen containing media yielded cells with similar morphology to corneal endothelial tissues. Elevated expression of CD56 and CD166 and low expression of CD73 correlated with regular, hexagonal-like HCEC morphology. CD56:CD73 > 2.5 was most consistent with normal HCEC morphology and mimicked corneal endothelial tissue. Immortalization of normal HCECs by hTERT transduction showed morphology and CD56:CD73 ratios similar to parental cell lines. HCECs established from FECD donors showed reduced CD56:CD73 ratios compared to normal HCECs which coincided with reduced uniformity and regularity of cell monolayers. Overall, a dual media system with Joyce's medium for proliferation and a low-mitogen media for maturation, provided normal cultures with regular, hexagonal-like cell morphologies consistent with corneal endothelial cells in vivo. CD56:CD73 expression ratio >2.5 was predictive of in vivo-like cellular morphology.


Subject(s)
Cell Culture Techniques , Endothelial Cells/pathology , Fuchs' Endothelial Dystrophy/pathology , Cell Proliferation , Culture Media , Endothelium, Corneal , Humans
2.
J Ocul Pharmacol Ther ; 37(5): 251-260, 2021 06.
Article in English | MEDLINE | ID: mdl-33784195

ABSTRACT

Purpose: To evaluate pharmacokinetic parameters and ocular hypotensive effects of cromakalim prodrug 1 (CKLP1) in normotensive large animal models. Methods: Optimal CKLP1 concentration was determined by dose response and utilized in short- (5-8 days) and long-term (60 days) evaluation in hound dogs (n = 5) and African Green Monkeys (n = 5). Blood pressure was recorded 3-5 times per week with a tail cuff. Concentrations of CKLP1 and the parent compound levcromakalim were assessed in hound dog plasma and select tissues by LC-MS/MS after bilateral ocular treatment with CKLP1 for 8 days. Pharmacokinetic parameters were calculated from days 1, 4, and 8 data. After necropsy, histology was assessed in 43 tissue samples from each animal. Results: In hound dogs and African Green monkeys, 10 mM CKLP1 (optimal concentration) significantly lowered intraocular pressure (IOP) by 18.9% ± 1.1% and 16.7% ± 6.7%, respectively, compared with control eyes (P < 0.05). During treatment, no significant change in systolic or diastolic blood pressure was observed in either species (P > 0.1). Average values for half-life of CKLP1 was 295.3 ± 140.4 min, Cmax, 10.5 ± 1.6 ng/mL, and area under the concentration vs. time curve (AUClast) 5261.4 ± 918.9 ng·min/mL. For levcromakalim, average values of half-life were 96.2 ± 27 min, Cmax 1.2 ± 0.2 ng/mL, and AUClast 281.2 ± 110.8 ng·min/mL. No significant pathology was identified. Conclusions: CKLP1 lowered IOP in hound dogs and African green monkeys with no effect on systemic blood pressure. Ocular topical treatment of CKLP1 showed excellent tolerability even after extended treatment periods.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Cromakalim/pharmacokinetics , Intraocular Pressure/drug effects , KATP Channels/drug effects , Administration, Ophthalmic , Administration, Topical , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacology , Area Under Curve , Autopsy/methods , Blood Pressure/drug effects , Chlorocebus aethiops , Cromakalim/administration & dosage , Cromakalim/pharmacology , Dogs , Dose-Response Relationship, Drug , Female , Half-Life , Models, Animal , Primates , Prodrugs
3.
Sci Rep ; 11(1): 4034, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597641

ABSTRACT

Ocular hypertension occurs due to increased resistance to aqueous humor removal through the conventional outflow pathway. Unlike the proximal region of the conventional outflow pathway, the distal region has not been well studied, mostly due to lack of model systems. Here we describe isolation and characterization of human primary vascular distal outflow pathway (VDOP) cells from the distal region of the conventional outflow pathway. Tissue from the distal region was isolated from human corneo-scleral rims, digested with collagenase type I (100 U/ml) and placed on gelatin coated plates to allow cellular growth in Dulbecco's Modified Eagle's Medium (low glucose) containing fetal bovine serum and antibiotic/antimycotic. VDOP cells showed consistent proliferation for up to 7 passages, retained endothelial-like nature of the parent tissues and showed a unique marker phenotype of Lectin+VEGFR2-CD34-NG2- that was distinct from neighboring trabecular meshwork (Lectin+VEGFR2-CD34-NG2+) and Schlemm's canal (Lectin+VEGFR2+CD34+NG2+) cells. Dexamethasone treated VDOP cells did not express myocilin and did not form cross-linked actin networks, in contrast to trabecular meshwork cells. These data show that VDOP cells are unique to the distal outflow region and can be used as a viable in vitro model system to understand the biology of the distal outflow pathway and intraocular pressure regulation.


Subject(s)
Aqueous Humor/metabolism , Intraocular Pressure/physiology , Ocular Hypertension/metabolism , Actins/metabolism , Adult , Aged , Aged, 80 and over , Aqueous Humor/physiology , Child , Child, Preschool , Cytoskeletal Proteins/metabolism , Endothelium/metabolism , Eye/pathology , Eye Proteins/metabolism , Female , Glaucoma/physiopathology , Glycoproteins/metabolism , Humans , Male , Middle Aged , Ocular Hypertension/physiopathology , Primary Cell Culture , Sclera/physiopathology , Trabecular Meshwork/metabolism
4.
PLoS One ; 15(4): e0231841, 2020.
Article in English | MEDLINE | ID: mdl-32298376

ABSTRACT

Elevated intraocular pressure is the only treatable risk factor for glaucoma, an eye disease that is the leading cause of irreversible blindness worldwide. We have identified cromakalim prodrug 1 (CKLP1), a novel water-soluble ATP-sensitive potassium channel opener, as a new ocular hypotensive agent. To evaluate the pharmacokinetic and safety profile of CKLP1 and its parent compound levcromakalim, Dutch-belted pigmented rabbits were treated intravenously (0.25 mg/kg) or topically (10 mM; 4.1 mg/ml) with CKLP1. Body fluids (blood, aqueous and vitreous humor) were collected at multiple time points and evaluated for the presence of CKLP1 and levcromakalim using a liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) based assay. Histology of tissues isolated from Dutch-belted pigmented rabbits treated once daily for 90 days was evaluated in a masked manner by a certified veterinary pathologist. The estimated plasma parameters following intravenous administration of 0.25 mg/kg of CKLP1 showed CKLP1 had a terminal half-life of 61.8 ± 55.2 min, Tmax of 19.8 ± 23.0 min and Cmax of 1968.5 ± 831.0 ng/ml. Levcromakalim had a plasma terminal half-life of 85.0 ± 37.0 min, Tmax of 61.0 ± 32.0 min and Cmax of 10.6 ± 1.2 ng/ml. Topical CKLP1 treatment in the eye showed low levels (<0.3 ng/mL) of levcromakalim in aqueous and vitreous humor, and trace amounts of CKLP1 and levcromakalim in the plasma. No observable histological changes were noted in selected tissues that were examined following topical application of CKLP1 for 90 consecutive days. These results suggest that CKPL1 is converted to levcromakalim in the eye and likely to some extent in the systemic circulation.


Subject(s)
Cromakalim/pharmacology , Cromakalim/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Administration, Intravenous , Administration, Topical , Animals , Aqueous Humor/drug effects , Aqueous Humor/metabolism , Chromatography, Liquid , Cornea/cytology , Cornea/drug effects , Cromakalim/administration & dosage , Cromakalim/blood , Eye/cytology , Eye/drug effects , Eye/metabolism , Female , Mass Spectrometry , Prodrugs/therapeutic use , Rabbits , Vitreous Body/drug effects , Vitreous Body/metabolism
5.
Invest Ophthalmol Vis Sci ; 58(13): 5731-5742, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29114841

ABSTRACT

Purpose: Cromakalim prodrug 1 (CKLP1) is a water-soluble ATP-sensitive potassium channel opener that has shown ocular hypotensive properties in ex vivo and in vivo experimental models. To determine its mechanism of action, we assessed the effect of CKLP1 on aqueous humor dynamics and in combination therapy with existing ocular hypotensive agents. Methods: Outflow facility was assessed in C57BL/6 mice by ex vivo eye perfusions and by in vivo constant flow infusion following CKLP1 treatment. Human anterior segments with no trabecular meshwork were evaluated for effect on pressure following CKLP1 treatment. CKLP1 alone and in combination with latanoprost, timolol, and Rho kinase inhibitor Y27632 were evaluated for effect on intraocular pressure in C57BL/6 mice and Dutch-belted pigmented rabbits. Results: CKLP1 lowered episcleral venous pressure (control: 8.9 ± 0.1 mm Hg versus treated: 6.2 ± 0.1 mm Hg, P < 0.0001) but had no detectable effect on outflow facility, aqueous humor flow rate, or uveoscleral outflow. Treatment with CKLP1 in human anterior segments without the trabecular meshwork resulted in a 50% ± 9% decrease in pressure, suggesting an effect on the distal portion of the conventional outflow pathway. CKLP1 worked additively with latanoprost, timolol, and Y27632 to lower IOP, presumably owing to combined effects on different aspects of aqueous humor dynamics. Conclusions: CKLP1 lowered intraocular pressure by reducing episcleral venous pressure and lowering distal outflow resistance in the conventional outflow pathway. Owing to this unique mechanism of action, CKLP1 works in an additive manner to lower intraocular pressure with latanoprost, timolol, and Rho kinase inhibitor Y27632.


Subject(s)
Antihypertensive Agents/therapeutic use , Aqueous Humor/physiology , Cromakalim/therapeutic use , Intraocular Pressure/drug effects , Prodrugs/therapeutic use , Amides/therapeutic use , Animals , Anterior Eye Segment/drug effects , Drug Synergism , Drug Therapy, Combination , Female , Humans , Latanoprost , Male , Mice , Mice, Inbred C57BL , Middle Aged , Ophthalmic Solutions , Prostaglandins F, Synthetic/therapeutic use , Pyridines/therapeutic use , Rabbits , Sclera/blood supply , Timolol/therapeutic use , Tonometry, Ocular , Venous Pressure/drug effects
6.
PLoS One ; 12(6): e0179345, 2017.
Article in English | MEDLINE | ID: mdl-28594895

ABSTRACT

Elevated intraocular pressure is the most prevalent and only treatable risk factor for glaucoma, a degenerative disease of the optic nerve. While treatment options to slow disease progression are available, all current therapeutic and surgical treatments have unwanted side effects or limited efficacy, resulting in the need to identify new options. Previous reports from our laboratory have established a novel ocular hypotensive effect of ATP-sensitive potassium channel (KATP) openers including diazoxide (DZ) and nicorandil (NCD). In the current study, we evaluated the role of Erk1/2 signaling pathway in KATP channel opener mediated reduction of intraocular pressure (IOP). Western blot analysis of DZ and NCD treated primary normal trabecular meshwork (NTM) cells, human TM (isolated from perfusion cultures of human anterior segments) and mouse eyes showed increased phosphorylation of Erk1/2 when compared to vehicle treated controls. DZ and NCD mediated pressure reduction (p<0.02) in human anterior segments (n = 7 for DZ, n = 4 for NCD) was abrogated by U0126 (DZ + U0126: -9.7 ± 11.5%, p = 0.11; NCD + U0126: -0.1 ± 11.5%, p = 1.0). In contrast, U0126 had no effect on latanoprostfree acid-induced pressure reduction (-52.5 ± 6.8%, n = 4, p = 0.001). In mice, DZ and NCD reduced IOP (DZ, 14.9 ± 3.8%, NCD, 16.9 ± 2.5%, n = 10, p<0.001), but the pressure reduction was inhibited by U0126 (DZ + U0126, 0.7 ± 3.0%; NCD + U0126, 0.9 ± 2.2%, n = 10, p>0.1). Histologic evaluation of transmission electron micrographs from DZ + U0126 and NCD + U0126 treated eyes revealed no observable morphological changes in the ultrastructure of the conventional outflow pathway. Taken together, the results indicate that the Erk1/2 pathway is necessary for IOP reduction by KATP channel openers DZ and NCD.


Subject(s)
Diazoxide/pharmacology , Intraocular Pressure/drug effects , Ion Channel Gating/drug effects , KATP Channels/metabolism , MAP Kinase Signaling System/drug effects , Nicorandil/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Butadienes/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Humans , Infant , Mice, Inbred C57BL , Middle Aged , Nitriles/pharmacology , Ocular Hypotension/physiopathology , Perfusion , Phosphorylation/drug effects
7.
Invest Ophthalmol Vis Sci ; 58(5): 2715-2724, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28538979

ABSTRACT

Purpose: To identify downstream signaling molecules through which intraocular pressure (IOP) is lowered following treatment with the prostaglandin analog latanoprost. Methods: Total RNA and protein isolated from primary human Schlemm's canal cells (n = 3) treated with latanoprost (free acid; 100 nM) were processed for quantitative PCR and Western blot analysis. IOP was evaluated in stanniocalcin-1 (STC-1-/-) and wild-type mice following treatment with latanoprost or Rho kinase inhibitor Y27632. Human anterior segment pairs (n = 8) were treated with recombinant STC-1 (5, 50, or 500 ng/mL) and pressure was recorded using custom-designed software. The effect of recombinant STC-1 (0.5 mg/mL) on IOP was evaluated in wild-type mice. Tissue morphology was evaluated by light and transmission electron microscopy. Results: Increased STC-1 mRNA (4.0- to 25.2-fold) and protein expression (1.9- to 5.1-fold) was observed within 12 hours following latanoprost treatment. Latanoprost reduced IOP in wild-type mice (22.0% ± 1.9%), but had no effect on STC-1-/- mice (0.5% ± 0.7%). In contrast, Y27632 reduced IOP in both wild-type (12.5% ± 1.2%) and in STC-1-/- mice (13.1% ± 2.8%). Human anterior segments treated with STC-1 (500 ng/mL) showed an increase in outflow facility (0.15 ± 0.03 to 0.27 ± 0.09 µL/min/mm Hg) while no change was observed in paired vehicle-treated controls. Recombinant STC-1 reduced IOP in wild-type mice by 15.2% ± 3.0%. No observable morphologic changes were identified between treatment groups when evaluated by microscopy. Conclusions: Latanoprost-induced reduction of IOP is mediated through the downstream signaling molecule STC-1. When used by itself, STC-1 exhibits ocular hypotensive properties.


Subject(s)
Antihypertensive Agents/pharmacology , Glycoproteins/genetics , Intraocular Pressure/drug effects , Prostaglandins F, Synthetic/pharmacology , Signal Transduction/physiology , Aged , Aged, 80 and over , Amides/pharmacology , Animals , Anterior Eye Segment/cytology , Anterior Eye Segment/drug effects , Blotting, Western , Cell Line , Gene Expression Regulation/physiology , Humans , Latanoprost , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Middle Aged , Ocular Hypotension/drug therapy , Pyridines/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tonometry, Ocular , rho-Associated Kinases/antagonists & inhibitors
9.
PLoS One ; 10(11): e0141783, 2015.
Article in English | MEDLINE | ID: mdl-26535899

ABSTRACT

Elevated intraocular pressure (IOP) is the most prevalent and only treatable risk factor for glaucoma, a leading cause of irreversible blindness worldwide. Unfortunately, all current therapeutics used to treat elevated IOP and glaucoma have significant and sometimes irreversible side effects necessitating the development of novel compounds. We evaluated the IOP lowering ability of the broad spectrum KATP channel opener cromakalim. Cultured human anterior segments when treated with 2 µM cromakalim showed a decrease in pressure (19.33 ± 2.78 mmHg at 0 hours to 13.22 ± 2.64 mmHg at 24 hours; p<0.001) when compared to vehicle treated controls (15.89 ± 5.33 mmHg at 0 h to 15.56 ± 4.88 mmHg at 24 hours; p = 0.89). In wild-type C57BL/6 mice, cromakalim reduced IOP by 18.75 ± 2.22% compared to vehicle treated contralateral eyes (17.01 ± 0.32 mmHg at 0 hours to 13.82 ± 0.37 mmHg at 24 hours; n = 10, p = 0.002). Cromakalim demonstrated an additive effect when used in conjunction with latanoprost free acid, a common ocular hypotensive drug prescribed to patients with elevated IOP. To examine KATP channel subunit specificity, Kir6.2(-/-) mice were treated with cromakalim, but unlike wild-type animals, no change in IOP was noted. Histologic analysis of treated and control eyes in cultured human anterior segments and in mice showed similar cell numbers and extracellular matrix integrity within the trabecular meshwork, with no disruptions in the inner and outer walls of Schlemm's canal. Together, these studies suggest that cromakalim is a potent ocular hypotensive agent that lowers IOP via activation of Kir6.2 containing KATP channels, its effect is additive when used in combination with the commonly used glaucoma drug latanoprost, and is not toxic to cells and tissues of the aqueous humor outflow pathway, making it a candidate for future therapeutic development.


Subject(s)
Cromakalim/pharmacology , Intraocular Pressure/drug effects , KATP Channels/agonists , Models, Biological , Administration, Topical , Adult , Aged , Aged, 80 and over , Animals , Cells, Cultured , Cromakalim/therapeutic use , Drug Therapy, Combination , Eye/cytology , Eye/pathology , Female , Glaucoma/drug therapy , Humans , KATP Channels/metabolism , Latanoprost , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prostaglandins F, Synthetic/pharmacology , Prostaglandins F, Synthetic/therapeutic use , Tissue Donors
10.
Am J Ophthalmol ; 153(6): 1206-13, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22464365

ABSTRACT

PURPOSE: To determine whether a second-generation trabecular meshwork (TM) bypass stent (iStent inject) influences outflow facility in cultured human anterior segments. DESIGN: Prospective laboratory investigation using normal human donor eyes. METHODS: Human anterior segments (n = 7) were placed in perfusion organ culture. One or 2 iStent inject stents were inserted into the TM within the nasal and/or superior quadrants using a specially designed injector. Anterior segments were returned to culture and perfused for an additional 24 hours. Morphology of the TM and Schlemm canal (SC) was assessed by scanning electron microscopy (SEM) and 3-dimensional micro-computed tomography (3D micro-CT). RESULTS: Insertion of 1 iStent inject into the nasal or superior quadrant of the TM increased outflow facility from 0.16 ± 0.05 µL/min/mm Hg to 0.38 ± 0.23 µL/min/mm Hg (P < .03, n = 7), with concurrent pressure reduction from 16.7 ± 5.4 mm Hg to 8.6 ± 4.4 mm Hg. Addition of a second iStent inject further increased outflow facility to 0.78 ± 0.66 µL/min/mm Hg (n = 2). SEM showed the iStent inject flange compressed against the uveal region of the TM, the thorax securely inserted within the TM, and the head located in the lumen of SC. Dilation of SC was noted around the iStent inject head and SC cell disruption was observed at the iStent inject insertion site. 3D micro-CT confirmed iStent inject placement. CONCLUSION: iStent inject, a second-generation bypass stent, increased outflow facility in human anterior segment culture. The iStent inject is a promising new device to lower intraocular pressure via TM bypass.


Subject(s)
Anterior Eye Segment/metabolism , Aqueous Humor/metabolism , Stents , Trabecular Meshwork/surgery , Adult , Aged , Aged, 80 and over , Diffusion Chambers, Culture , Female , Humans , Imaging, Three-Dimensional , Male , Microscopy, Electron, Scanning , Middle Aged , Organ Culture Techniques , Prospective Studies , Tissue Donors , Trabecular Meshwork/ultrastructure
11.
Invest Ophthalmol Vis Sci ; 52(9): 6435-42, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21743021

ABSTRACT

PURPOSE. ATP-sensitive potassium channel (K(ATP)) openers target key cellular events, many of which have been implicated in glaucoma. The authors sought to determine whether K(ATP) channel openers influence outflow facility in human anterior segment culture and intraocular pressure (IOP) in vivo. METHODS. Anterior segments from human eyes were placed in perfusion organ culture and treated with the K(ATP) channel openers diazoxide, nicorandil, and P1075 or the K(ATP) channel closer glyburide (glibenclamide). The presence, functionality, and specificity of K(ATP) channels were determined by RT-PCR, immunohistochemistry, and inside-out patch clamp in human trabecular meshwork (TM) tissue or primary cultures of normal human trabecular meshwork (NTM) cells. The effect of diazoxide on IOP in anesthetized Brown Norway rats was measured with a rebound tonometer. RESULTS. K(ATP) channel openers increased outflow facility in human anterior segments (0.14 ± 0.02 to 0.26 ± 0.09 µL/min/mm Hg; P < 0.001) compared with fellow control eyes (0.22 ± 0.11 to 0.21 ± 0.11 µL/min/mm Hg; P > 0.5). The effect was reversible, with outflow facility returning to baseline after drug removal. The addition of glyburide inhibited diazoxide from increasing outflow facility. Electrophysiology confirmed the presence and specificity of functional K(ATP) channels. K(ATP) channel subunits K(ir)6.1, K(ir)6.2, SUR2A, and SUR2B were expressed in TM and NTM cells. In vivo, diazoxide significantly lowered IOP in Brown Norway rats. CONCLUSIONS. Functional K(ATP) channels are present in the trabecular meshwork. When activated by K(ATP) channel openers, these channels increase outflow facility through the trabecular outflow pathway in human anterior segment organ culture and decrease IOP in Brown Norway rat eyes.


Subject(s)
Intraocular Pressure/drug effects , KATP Channels/metabolism , Trabecular Meshwork/drug effects , Vasodilator Agents/pharmacology , Aged , Aged, 80 and over , Animals , Cells, Cultured , Diazoxide/pharmacology , Female , Glyburide/pharmacology , Guanidines/pharmacology , Humans , Immunohistochemistry , KATP Channels/genetics , Male , Middle Aged , Nicorandil/pharmacology , Organ Culture Techniques , Patch-Clamp Techniques , Pyridines/pharmacology , Rats , Rats, Inbred BN , Reverse Transcriptase Polymerase Chain Reaction , Tonometry, Ocular , Trabecular Meshwork/metabolism
12.
Invest Ophthalmol Vis Sci ; 50(5): 2123-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19136716

ABSTRACT

PURPOSE: To determine the effects of incorporating superparamagnetic microspheres (SPMs) into cultured human corneal endothelial cells (HCECs) and to describe preliminary experiments of HCEC transplantation, facilitated by SPMs and an external magnetic field, in a human anterior segment ex vivo model. METHODS: HCECs were cultured as monolayers and incorporated with magnetite oxide SPMs (900, 300, and 100 nm) at different concentrations. Cell viability, migration toward a magnetic field, and light transmittance were measured after incorporation of the SPMs. HCEC transplantation into the eyes of human recipients was investigated by subjecting anterior segments in organ culture to an external magnetic field. Light and electron microscopy were used to assess HCEC attachment to corneal stroma. RESULTS: SPMs were incorporated into the cytoplasm of HCECs after overnight incubation. None of the SPMs affected the short-term viability of cultured HCECs (P > 0.14, n = 6) or their light transmittance (P > 0.06, n = 5), although there was a trend toward decreased transmittance with the higher concentration of 900-nm SPMs. Cell migration toward a magnetic field was higher for HCECs with incorporated SPMs than for HCECs without SPMs (P < or = 0.01, n = 6), with dose-response relationships evident for the 300- and 100-nm SPMs. SPMs facilitated the attachment of HCECs to the corneal stroma in the human anterior segment model with minimal change in intracameral (intraocular) pressure. CONCLUSIONS: SPMs facilitate migration of HCECs toward a magnetic source and attachment of cells to the corneal stroma without affecting cell viability or light transmittance. The human anterior segment model can be used to study HCEC transplantation.


Subject(s)
Anterior Eye Segment/surgery , Cell Transplantation , Endothelium, Corneal/transplantation , Models, Biological , Adolescent , Adult , Cell Count , Cell Movement/physiology , Cell Survival/physiology , Cells, Cultured , Endothelium, Corneal/metabolism , Endothelium, Corneal/ultrastructure , Ferrosoferric Oxide/metabolism , Humans , Magnetics , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microspheres , Middle Aged , Organ Culture Techniques
13.
Exp Eye Res ; 87(4): 376-84, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18674535

ABSTRACT

Although the glaucoma-associated protein myocilin has been the focus of intensive research, its biological function is still unknown. One of the limiting factors has been the lack of well-characterized antibodies, particularly monoclonal antibodies. We describe the development of six monoclonal antibodies specific to myocilin and characterize their suitability in Western blot and immunohistochemical applications. Three of the six monoclonal antibodies recognize the N-terminus of myocilin (amino acids 33-214), two antibodies recognize the middle third of the protein (amino acids 215-368), and one antibody recognizes the C-terminus (amino acids 369-504). Isotyping revealed that all antibodies are of the IgG1 kappa class except one, which is IgG2b kappa. Purified myocilin monoclonal antibodies were able to recognize myocilin in human aqueous humor separated on denatured/reduced and native gels, and human trabecular meshwork lysate by Western blot. Myocilin was also detected by immunohistochemistry in trabecular meshwork, ciliary body, iris, cornea, sclera, choroid, and retinal pigment epithelial cells.


Subject(s)
Antibodies, Monoclonal/immunology , Cytoskeletal Proteins/immunology , Eye Proteins/immunology , Glycoproteins/immunology , Aged, 80 and over , Animals , Antibodies, Monoclonal/biosynthesis , Antibody Specificity , Aqueous Humor/metabolism , Binding Sites, Antibody , Blotting, Western/methods , Culture Media, Conditioned , Cytoskeletal Proteins/isolation & purification , Eye/metabolism , Eye Proteins/isolation & purification , Female , Glycoproteins/isolation & purification , Humans , Hybridomas , Male , Mice , Mice, Inbred BALB C , Recombinant Proteins/isolation & purification , Trabecular Meshwork/cytology , Trabecular Meshwork/metabolism
14.
Am J Ophthalmol ; 145(1): 114-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17988642

ABSTRACT

PURPOSE: To determine the effect of latanoprost free acid and prostaglandin E1 (PGE1) on outflow facility in cultured human anterior segments. Clinical studies find prostaglandin treatment increases uveoscleral outflow, but do not agree whether trabecular outflow increases. Cultured anterior segments eliminate the uveoscleral pathway and enable a direct assessment of trabecular outflow. DESIGN: Laboratory investigation. METHODS: One anterior segment of an eye pair was placed in perfusion organ culture and received a continuous infusion of drug while the fellow anterior segment received vehicle. Histologic changes were assessed. Zymography and Western blots were used to analyze matrix metalloprotease (MMP) activity. Scleral hydraulic conductivities were measured. RESULTS: Latanoprost significantly increased outflow facility (67% +/- 11% vs control 6% +/- 10%, P < .001). Facility changes occurred within one hour of receiving drug, reaching a new baseline by 24 hours. Facility changes were reversible, requiring about 48 hours to return to pre-drug values. PGE1 caused less facility change (13% +/- 17% vs control 1% +/- 11%, P = .02). Prostaglandin treated anterior segments had regions of focal detachment and loss of Schlemm canal endothelial cells, with loss of extracellular matrix underlying some areas. MMPs were not consistently increased in Western blots, zymography, or immunohistochemistry. Scleral hydraulic conductivity increased, but not enough to account for total facility increase. CONCLUSIONS: Prostaglandins increase outflow facility in perfusion organ culture of human anterior segments. MMP activity was not consistently increased, and scleral hydraulic conductivity was not increased sufficiently to account for total facility increase. The histologic changes suggest a direct trabecular meshwork effect.


Subject(s)
Alprostadil/pharmacology , Antihypertensive Agents/pharmacology , Aqueous Humor/metabolism , Prostaglandins F, Synthetic/pharmacology , Trabecular Meshwork/drug effects , Aged , Aged, 80 and over , Anterior Eye Segment/drug effects , Anterior Eye Segment/enzymology , Blotting, Western , Fluorescent Antibody Technique, Indirect , Humans , Latanoprost , Matrix Metalloproteinases/metabolism , Organ Culture Techniques , Sclera/physiology , Trabecular Meshwork/enzymology
15.
Invest Ophthalmol Vis Sci ; 47(1): 213-21, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16384965

ABSTRACT

PURPOSE: A previous study by the authors has shown that recombinant myocilin purified from a prokaryotic expression system increases outflow resistance in cultured human anterior segments. The present study was performed to determine whether full-length myocilin purified from a human trabecular meshwork cell expression system alters outflow resistance after infusion into human anterior segments. METHODS: A feline immunodeficiency virus vector encoding both full-length myocilin (amino acids 1-503 fused to C-terminal V5 and six-histidine epitopes) and puromycin resistance was used to transduce a transformed trabecular meshwork cell line (TM5). Stably expressing cells were selected with puromycin. Recombinant myocilin was purified from the media using nickel ion affinity chromatography. Control purifications were performed on media from parental TM5 cells. Anterior segments of human eyes were placed in organ culture and perfused with either Dulbecco's modified Eagle's medium (DMEM) or DMEM supplemented with 50% porcine aqueous humor. One eye received an anterior chamber exchange with recombinant myocilin (2 microg/mL), whereas the fellow eye received an equal volume of control. Immunohistochemistry was performed with anti-myocilin and anti-V5 antibodies. Native polyacrylamide gel electrophoresis was used to analyze myocilin complex formation in porcine aqueous humor. RESULTS: Recombinant myocilin in porcine aqueous humor increased outflow resistance in cultured human anterior segments (91% +/- 68% [mean +/- SD] versus 18% +/- 31% in fellow control eye; n = 9, P = 0.004). Maximum outflow resistance was obtained 5 to 17 hours after infusion and remained above baseline for >3 days. Recombinant myocilin also increased outflow resistance in eyes incubated in DMEM, but only if myocilin was preincubated with porcine aqueous humor (78% +/- 77% when preincubated in DMEM containing porcine aqueous humor versus 13% +/- 15% when preincubated with DMEM alone, n = 6, P = 0.03). Recombinant myocilin appears to form a complex in porcine aqueous humor with a heat-labile protein(s). Immunohistochemistry revealed the presence of myocilin in the juxtacanalicular region of the trabecular meshwork. CONCLUSIONS: Myocilin purified from human trabecular meshwork cells increased outflow resistance in cultured human anterior segments, but only after incubation with porcine aqueous humor. Recombinant myocilin appears to form a complex in porcine aqueous humor that enables it to bind specifically within the trabecular meshwork.


Subject(s)
Aqueous Humor/physiology , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glycoproteins/metabolism , Histidine/metabolism , Trabecular Meshwork/metabolism , Aged , Aged, 80 and over , Animals , Cell Line , Female , Gene Expression , Genetic Vectors , Humans , Male , Middle Aged , Organ Culture Techniques , Recombinant Fusion Proteins/metabolism , Swine , Trabecular Meshwork/cytology , Transfection
16.
Invest Ophthalmol Vis Sci ; 46(1): 1-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15623746

ABSTRACT

PURPOSE: To determine whether segmental labeling by the tracer molecule cationic ferritin (CF) is indicative of preferential patterns of fluid flow in the trabecular meshwork or of differences in cell and extracellular matrix properties. Nonlabeled regions could indicate no fluid entering that area, insufficient perfusion time, or that the cells and extracellular matrix differ in that region and cannot bind CF. METHODS: Six whole eyes (three normal and three with pseudoexfoliation [PEX]) syndrome were perfused with CF for 30 minutes to 4 hours. Wedges of trabecular meshwork were dissected and some wedges immediately fixed. Adjacent wedges were placed in a CF bath before fixation. Transmission electron microscopy was used to analyze CF labeling. RESULTS: CF increased in the trabecular meshwork with increasing perfusion time. At 30 minutes, CF labeled mainly the uveal and corneoscleral regions. By 4 hours, CF was found diffusely through the meshwork, although a few isolated nonlabeled areas were still present. Wedges immersed in the CF bath showed fewer nonlabeled regions at all time points. Clumps of PEX material labeled more heavily in the periphery than the center, suggesting the clumps were less permeable than surrounding regions. PEX eyes otherwise had similar labeling patterns. CONCLUSIONS: Segmental labeling with CF implies regions of preferential flow exist in the meshwork. With increasing perfusion time, there were fewer nonlabeled regions. CF labeling of most regions of bath-immersed tissue suggests that nonlabeled regions do not differ in the characteristics of the cells, but rather that CF does not reach these regions.


Subject(s)
Ferritins/metabolism , Trabecular Meshwork/metabolism , Aged , Aged, 80 and over , Biological Transport, Active , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Exfoliation Syndrome/metabolism , Exfoliation Syndrome/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Humans , Ligands , Microscopy, Electron, Transmission , Perfusion , Staining and Labeling , Trabecular Meshwork/ultrastructure
17.
Invest Ophthalmol Vis Sci ; 45(9): 3091-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15326125

ABSTRACT

PURPOSE: To address a problem impeding research into glaucoma-associated genetic mutations and glaucoma gene therapy and achieve permanent, targeted transgene expression in the trabecular meshwork (TM). Lentiviral vectors are known to transduce human donor eye TM ex vivo, but efficacy in vivo has not been shown. More generally in the field of gene therapy, the authors hypothesized that distinctive properties of the intraocular aqueous circulation could facilitate solving problems of accessibility, targeting, and scale that have hindered realization of gene therapy in other settings. METHODS: A domestic cat model was developed in which long-term in vivo studies were performed. After dose-response studies in primary human TM cells, 19 cats received anterior chamber (AC) injections of stepped doses (10(6)-10(8) transduction units) of lentiviral vectors encoding different marker transgenes (beta-galactosidase, Aequorea victoria green fluorescent protein [GFP], or Renilla reniformis GFP). Animals were monitored serially for transgene expression and IOP. RESULTS: High-grade, stable transgene expression in the TM was achieved and monitored noninvasively over time in living animals. Extensive expression resulted after a single transcorneal injection, persisted for at least 10 months (time of death in the present studies), and was targeted to the TM. The initial IOP did not differ significantly from the IOP at the end of the study (P = 0.4). Aequorea GFP was superior to Renilla GFP. Vectors were effective enough to cause GFP-specific overexpression cytotoxicity at the highest dose, which was solved by dose reduction. CONCLUSIONS: High-grade transgene expression in this large-animal model persisted stably for at least 10 months after a single transcorneal lentiviral vector injection, was highly targeted, and could be monitored serially and noninvasively in living animals. These studies provide a basis for developing realistic disease models and administering glaucoma gene therapy.


Subject(s)
Aqueous Humor/physiology , Gene Expression , Gene Targeting , Animals , Animals, Genetically Modified , Cats , Cells, Cultured , Genetic Vectors , Green Fluorescent Proteins , Humans , Indicators and Reagents/metabolism , Intraocular Pressure , Lentivirus/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Time Factors , Trabecular Meshwork/cytology , Trabecular Meshwork/physiology , Transduction, Genetic , Transgenes , beta-Galactosidase/genetics
18.
Invest Ophthalmol Vis Sci ; 45(9): 3137-43, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15326132

ABSTRACT

PURPOSE: To determine why variations in intraocular pressure (IOP) occur in cultured human anterior segments despite a constant rate of infusion of culture medium. Two types of variations occur: an initial elevation of IOP and small changes in baseline IOP. METHODS: Anterior segments from human eyes were placed in perfusion organ culture. In cultures with initially high IOP, eyes were fixed at the high IOP level and histologic examination performed. In other cultures with high initial IOP, effluent medium was collected and subsequently reinfused after IOP had decreased to baseline. In cultures with stable baseline IOP, cell fragments from monolayer-cultured cells, or human genomic DNA, were infused at concentrations equivalent to 30,000 to 300,000 cells. RESULTS: Electron microscopy of initially high-pressure cultures revealed scattered cell debris throughout the meshwork in greater amounts than found in eyes without initially high IOP. Reinfusion of effluent media from cultures with high initial pressures caused elevation of IOP. Centrifugation of effluent media lessened this elevation of IOP. In cultures with stable baseline IOP, infusion of cell fragments or genomic DNA raised IOP in a dose-dependent manner, with elevation of IOP for a minimum of 24 hours. CONCLUSIONS: Cell debris can elevate IOP during the initial culture period, and after baseline pressures are established. Cell fragments and DNA increase IOP in a dose-dependent manner. The variations in baseline IOP seen during culture are probably caused by cell fragments and debris from dying cells in the meshwork, ciliary body, and other anterior segment tissues.


Subject(s)
Anterior Eye Segment/physiopathology , Intraocular Pressure , Aged , Aged, 80 and over , Anterior Eye Segment/ultrastructure , Cadaver , DNA/pharmacology , Genome, Human , Humans , Intraocular Pressure/drug effects , Microscopy, Electron , Middle Aged , Organ Culture Techniques , Perfusion , Time Factors , Trabecular Meshwork/ultrastructure
19.
Invest Ophthalmol Vis Sci ; 45(7): 2246-54, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15223802

ABSTRACT

PURPOSE: To determine the effect of disruption of Schlemm's canal cells on outflow facility. Pharmacologic agents that weaken the cytoskeleton or interfere with integrin binding may allow targeted disruption of the cells lining Schlemm's canal because of the transmural pressure gradient the cells face as aqueous passes into the canal. METHODS: Anterior segments of human eyes were placed in perfusion organ culture, and either single or sequential doses of H-7 or RGD peptide were added. Fellow eyes received vehicle or RGE peptide. Eyes were fixed and examined by light and electron microscopy. RESULTS: Both agents caused a partial loss of the endothelial lining of Schlemm's canal cells without disruption of trabecular cells in other regions. H-7 significantly increased outflow facility after single or sequential doses, with moderate cell loss of both the inner and outer wall canal cells: 20.0% +/- 10.5% of the width of the canal versus 5.2% +/- 3.7% in control meshworks (P = 0.05). No significant correlation between the amount of canal cell loss and outflow facility was found. RGD was associated with a variable loss of canal cells but did not change outflow facility. CONCLUSIONS: Pharmacologic disruption of Schlemm's canal cells appears possible. H-7 increased outflow facility, causing a partial loss of the endothelial lining of Schlemm's canal. A simple relationship between canal cells and outflow facility was not found; canal cells probably interact with the extracellular matrix in influencing outflow facility.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Aqueous Humor/metabolism , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Oligopeptides/pharmacology , Trabecular Meshwork/drug effects , Adult , Aged , Aged, 80 and over , Cell Survival , Endothelial Cells/ultrastructure , Humans , Intraocular Pressure/drug effects , Middle Aged , Organ Culture Techniques , Trabecular Meshwork/ultrastructure
20.
Am J Ophthalmol ; 138(6): 988-94, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15629290

ABSTRACT

PURPOSE: To determine the effect on intraocular pressure (IOP) of bypassing the trabecular meshwork in cultured human anterior segments. DESIGN: Prospective laboratory investigation using normal human eyes obtained at autopsy. METHODS: Anterior segments from 21 eyes were placed in perfusion culture, and trabecular bypass stents were inserted through the trabecular meshwork, with the lumen of the tube opening into Schlemm's canal. Eyes received from one to four stents, placed equidistant apart. In eyes receiving one or two stents, additional stents were later added to a maximum of four per eye. RESULTS: Intraocular pressure was lowered after placement of a single stent, from 21.4 +/- 3.8 mm Hg to 12.4 +/- 4.2 (P < .001). This corresponded to an 84% increase in facility of outflow. Eyes receiving more than one stent had final IOP of 11.9 +/- 3.7 mm Hg. Nine eyes had sequential addition of stents, and seven of these had a further decrease of IOP (13.6 +/- 4.1 to 10.0 +/- 4.3; P = .02). Excision of the entire meshwork, between stents, dropped IOP to 6.3 +/- 3.2 mm Hg, indicating some residual meshwork or canal resistance remained even after placement of three stents. CONCLUSIONS: Bypass of the trabecular meshwork lowers IOP in cultured human anterior segments. One stent produced the greatest change in pressure. The sequential addition of more stents further lowered pressure in seven of nine eyes. This technique holds promise as a new clinical surgery for glaucoma.


Subject(s)
Aqueous Humor/metabolism , Intraocular Pressure/physiology , Prosthesis Implantation/methods , Stents , Trabecular Meshwork/surgery , Anterior Eye Segment/physiology , Glaucoma/surgery , Humans , Organ Culture Techniques , Perfusion , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...