Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Adv Exp Med Biol ; 1415: 421-425, 2023.
Article in English | MEDLINE | ID: mdl-37440067

ABSTRACT

Most forms of outer retinal degenerative diseases involve the ectopic accumulation of microglia/macrophages in the subretinal space, including retinitis pigmentosa. However, their role in the loss of photoreceptor function during retinal degeneration remains unknown. Here, we examined the effect of conditional microglial depletion on photoreceptor numbers and visual function in mice with the rhodopsin P23H mutation, a dominant form of retinitis pigmentosa in humans. We found that microglial depletion led to an elevated level of rhodopsin and increased photoreceptor layer thickness. However, overall electrophysiological functions of the retina were reduced with microglial depletion. Therefore, these results identify an essential role of microglia specially in preserving visual function in outer retinal degeneration.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Humans , Mice , Animals , Rhodopsin/genetics , Retinal Degeneration/genetics , Microglia , Retinitis Pigmentosa/genetics , Retina , Disease Models, Animal
2.
Ophthalmol Sci ; 2(4): 100179, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36531586

ABSTRACT

Purpose: Rhegmatogenous retinal detachment repair by intraoperative sealing of the tear without a tamponade agent should enable faster restoration of vision and resumption of normal activities. It avoids the need for further surgery in the case of silicone oil endotamponade. This study evaluated the retinal thermofusion (RTF) retinopexy method of subretinal space dehydration before photocoagulation to create an instantaneous intraoperative retina reattachment in a preclinical model. Design: Preclinical study. Participants: Twenty Dutch Belt, pigmented rabbits that underwent RTF repair after experimental retinal detachment. Methods: This ex vivo model quantified adhesion force between the retina and underlying retinal pigment epithelium and choroid after treatment of 1 retinal edge using postmortem porcine or human retina (6 × 12 mm). We compared (1) control, (2) laser photocoagulation alone, (3) dehydration alone, and (4) dehydration followed by photocoagulation (RTF). Optimized parameters for RTF were then applied in the in vivo rabbit model of retinal detachment. Animals were followed up for 14 days. Main Outcome Measures: For this ex vivo model, we measured adhesion force and related this to tissue temperature. For the in vivo study, we assessed retinal attachment using funduscopy and histologic analysis. Results: The ex vivo model showed that RTF repair produced significantly higher adhesion force than photocoagulation alone independent of dehydration method: warm (60° C) high airflow (50-70 ml/minute) or using laser wavelengths targeting water absorption peaks (1470 or 1940 nm) with coaxial low airflow (10-20 ml/minute). The latter approach produced a smaller footprint of dehydration. Application of RTF (1940-nm laser with coaxial airflow) in an in vivo retinal detachment model in rabbit eyes resulted in immediate retinal adhesion, achieving forces similar to those in the ex vivo experiments. Retinal thermofusion repair resulted in stable reattachment of the retina over the 2-week follow-up period. Conclusions: We showed that a short preliminary dehydrating laser treatment of a retinal tear margin before traditional laser photocoagulation creates an immediate intraoperative waterproof retinopexy adhesion independent of tamponade and a wound-healing response. This approach potentially will allow rapid postoperative recovery regardless of the tear location and improved vision.

3.
Cells ; 10(9)2021 09 20.
Article in English | MEDLINE | ID: mdl-34572137

ABSTRACT

Inherited retinal degenerations (IRD) affecting either photoreceptors or pigment epithelial cells cause progressive visual loss and severe disability, up to complete blindness. Retinal organoids (ROs) technologies opened up the development of human inducible pluripotent stem cells (hiPSC) for disease modeling and replacement therapies. However, hiPSC-derived ROs applications to IRD presently display limited maturation and functionality, with most photoreceptors lacking well-developed outer segments (OS) and light responsiveness comparable to their adult retinal counterparts. In this review, we address for the first time the microenvironment where OS mature, i.e., the subretinal space (SRS), and discuss SRS role in photoreceptors metabolic reprogramming required for OS generation. We also address bioengineering issues to improve culture systems proficiency to promote OS maturation in hiPSC-derived ROs. This issue is crucial, as satisfying the demanding metabolic needs of photoreceptors may unleash hiPSC-derived ROs full potential for disease modeling, drug development, and replacement therapies.


Subject(s)
Bioengineering/methods , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Retinal Degeneration/therapy , Retinal Pigment Epithelium/cytology , Animals , Humans , Retinal Degeneration/pathology
4.
FASEB J ; 35(7): e21689, 2021 07.
Article in English | MEDLINE | ID: mdl-34085737

ABSTRACT

Knockout of the chloride channel protein 2 (CLC-2; CLCN2) results in fast progressing blindness in mice. Retinal Pigment Epithelium (RPE) and photoreceptors undergo, in parallel, rapid, and profound morphological changes and degeneration. Immunohistochemistry and electron microscopy of the outer retina and electroretinography of the CLC-2 KO mouse demonstrated normal morphology at postnatal day 2, followed by drastic changes in RPE and photoreceptor morphology and loss of vision during the first postnatal month. To investigate whether the RPE or the photoreceptors are the primary cause of the degeneration, we injected lentiviruses carrying HA-tagged CLC-2 with an RPE-specific promotor in the subretinal space of CLC-2-KO mice at the time of eye opening. As expected, CLC-2-HA was expressed exclusively in RPE; strikingly, this procedure rescued the degeneration of both RPE and photoreceptors. Light response in transduced eyes was also recovered. Only a fraction of RPE was transduced with the lentivirus; however, the entire RPE monolayer appears healthy, even the RPE cells not expressing the CLC-2-HA. Surprisingly, in contrast with previous physiological observations that postulate that CLC-2 has a basolateral localization in RPE, our immunofluorescence experiments demonstrated CLC-2 has an apical distribution, facing the subretinal space and the photoreceptor outer segments. Our findings suggest that CLC-2 does not play the postulated role in fluid transport at the basolateral membrane. Rather, they suggest that CLC-2 performs a critical homeostatic role in the subretinal compartment involving a chloride regulatory mechanism that is critical for the survival of both RPE and photoreceptors.


Subject(s)
Chloride Channels/physiology , Photoreceptor Cells/cytology , Retina/cytology , Retinal Degeneration , Retinal Pigment Epithelium/metabolism , Animals , CLC-2 Chloride Channels , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells/metabolism , Retina/metabolism
5.
Adv Exp Med Biol ; 1296: 349-358, 2020.
Article in English | MEDLINE | ID: mdl-34185303

ABSTRACT

Retinoblastoma, an intraocular cancer primarily affecting children, interacts with surrounding intraocular and extraocular structures in the development and progression. Subretinal and vitreous seeds are characteristic features of retinoblastoma, which result from the interaction between the tumor and its environment at the levels of tissue and microenvironment. The retina and vitreous affect the disease course and responses to treatment options. Also, neighboring cells in the retina and physicochemical properties of the tumor microenvironment are related to the biological activities of retinoblastoma tumors. Researches focusing on the tumor environment of retinoblastoma will lead to the development of more effective treatment options, which can revolutionize the prognosis of patients with retinoblastoma.


Subject(s)
Retinal Neoplasms , Retinoblastoma , Child , Humans , Infant , Retina , Retinal Neoplasms/epidemiology , Retinoblastoma/diagnosis , Retinoblastoma/epidemiology , Treatment Outcome , Tumor Microenvironment
6.
Genetics ; 214(1): 121-134, 2020 01.
Article in English | MEDLINE | ID: mdl-31754016

ABSTRACT

One major aspect of the aging process is the onset of chronic, low-grade inflammation that is highly associated with age-related diseases. The molecular mechanisms that regulate these processes have not been fully elucidated. We have identified a spontaneous mutant mouse line, small with kinky tail (skt), that exhibits accelerated aging and age-related disease phenotypes including increased inflammation in the brain and retina, enhanced age-dependent retinal abnormalities including photoreceptor cell degeneration, neurodegeneration in the hippocampus, and reduced lifespan. By positional cloning, we identified a deletion in chondroitin sulfate synthase 1 (Chsy1) that is responsible for these phenotypes in skt mice. CHSY1 is a member of the chondroitin N-acetylgalactosaminyltransferase family that plays critical roles in the biosynthesis of chondroitin sulfate, a glycosaminoglycan (GAG) that is attached to the core protein to form the chondroitin sulfate proteoglycan (CSPG). Consistent with this function, the Chsy1 mutation dramatically decreases chondroitin sulfate GAGs in the retina and hippocampus. In addition, macrophage and neutrophil populations appear significantly altered in the bone marrow and spleen of skt mice, suggesting an important role for CHSY1 in the functioning of these immune cell types. Thus, our study reveals a previously unidentified impact of CHSY1 in the retina and hippocampus. Specifically, chondroitin sulfate (CS) modification of proteins by CHSY1 appears critical for proper regulation of immune cells of the myeloid lineage and for maintaining the integrity of neuronal tissues, since a defect in this gene results in increased inflammation and abnormal phenotypes associated with age-related diseases.


Subject(s)
Chondroitin Sulfates/metabolism , Glucuronosyltransferase/metabolism , Inflammation/metabolism , Multifunctional Enzymes/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Neurodegenerative Diseases/metabolism , Protein Processing, Post-Translational , Proteins/genetics , Retinal Degeneration/metabolism , Age Factors , Animals , Apoptosis/physiology , Female , Glucuronosyltransferase/genetics , Inflammation/genetics , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multifunctional Enzymes/genetics , Mutation , N-Acetylgalactosaminyltransferases/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Proteins/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/pathology
7.
Adv Exp Med Biol ; 1185: 181-186, 2019.
Article in English | MEDLINE | ID: mdl-31884609

ABSTRACT

As the resident macrophages of central nervous system, microglia reside in the plexiform and nerve fiber layers of the retina. In degenerative diseases, monocyte-derived macrophages can be recruited to the retina, and histopathology shows abnormal accumulation of macrophages subretinally. However, due to lack of known markers, recruited cells and resident microglia are phenotypically indistinguishable, leaving a major knowledge gap about their potentially independent roles. Here, we used single cell RNA-seq and analyzed over 10,000 immune cells of mouse retinas from normal control and light damage-induced retinal degeneration. We observed ten major macrophage clusters. Moreover, combining trajectory analysis and in situ validation allowed us to pinpoint that subretinal phagocytes are microglia-derived and express high levels of Gal3, Cd68, and Lpl but not P2ry12. Hence, we have identified novel subretinal macrophage markers indicative of their origin and phenotype, which may be useful in other degeneration models and human specimens.


Subject(s)
Microglia/classification , Retinal Degeneration/pathology , Animals , Disease Models, Animal , Humans , Macrophages/classification , Macrophages/cytology , Mice , Microglia/cytology , RNA-Seq , Retina/cytology
8.
Exp Eye Res ; 180: 1-7, 2019 03.
Article in English | MEDLINE | ID: mdl-30468719

ABSTRACT

PURPOSE: It has been proposed that changes in the permeability of Bruch's membrane play a role in the pathogenesis of age-related macular degeneration (AMD). This paper investigates, in an in vivo porcine model, the migration of fluorescent latex beads across the Bruch's membrane after subretinal injection. METHODS: Forty-one healthy eyes of 33 three-month-old domestic pigs received a subretinal injection of 0.5, 1.0, 2.0, or 4.0 µm fluorescent latex beads. Between three hours and five weeks after injection evaluations were performed with fundus photographs and histology. Fluorescent beads were identified in unstained histologic sections using the rhodamine filter with the light microscope. RESULTS: The fluorescent latex beads relocated from the subretinal space. Intact beads up to 2.0 µm were found in the choroid, sclera, and extrascleral space. The smaller beads were also found inside choroidal and extrascleral blood vessels. In contrast, the larger beads of 4.0 µm did not pass the Bruch's membrane. CONCLUSION: Subretinally implanted beads up to 2.0 µm pass the Bruch's membrane intact and cross the blood-ocular barrier. The intact beads are found in the choroid, sclera and inside blood vessels. The results give reason to consider the role of subretinal clearance and passage of Bruch's membrane in the development of AMD.


Subject(s)
Bruch Membrane/metabolism , Choroid/metabolism , Latex , Microspheres , Models, Animal , Sclera/metabolism , Animals , Biological Transport , Female , Fluorescent Dyes/metabolism , Injections, Intraocular , Intracellular Space , Particle Size , Permeability , Rhodamines/metabolism , Sus scrofa
9.
Transl Vis Sci Technol ; 7(2): 19, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29651361

ABSTRACT

PURPOSE: We advance studies of subretinal treatments by developing a microscope-integrated optical coherence tomography (MIOCT) image-based method for measuring the volume of therapeutics delivered into the subretinal space. METHODS: A MIOCT image-based volume measurement method was developed and assessed for accuracy and reproducibility by imaging an object of known size in model eyes. This method then was applied to subretinal blebs created by injection of diluted triamcinolone. Bleb volumes obtained from MIOCT were compared to the intended injection volume and the surgeon's estimation of leakage. RESULTS: Validation of the image-based volume measurement method showed accuracy to ±1.0 µL (6.0% of measured volume) with no statistically significant variation under different imaging settings. When this method was applied to subretinal blebs, four of 11 blebs without surgeon-observed leakage yielded a mean volume of 32 ± 12.5 µL, in contrast to the intended 50 µL volume injected from the delivery device. This constituted a mean difference of -18 µL (mean percent error, 36 ± 25%). For all 11 blebs, the surgeon's estimations of leakage were significantly different from and showed no correlation with the volume loss based on image-based volume measurements (P < 0.001, paired t-test; intraclass correlation = 0). CONCLUSIONS: We validated an accurate and reproducible method for measuring subretinal volumes using MIOCT. Use of this method revealed that the intended volume might not be delivered into the subretinal space. MIOCT can allow for accurate assessment of subretinal dose delivered, which may have therapeutic implications in evaluating the efficacy and toxicity of subretinal therapies. TRANSLATIONAL RELEVANCE: Use of MIOCT can provide feedback on the accuracy of subretinal injection volumes delivered.

10.
Transl Vis Sci Technol ; 7(1): 3, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29359109

ABSTRACT

PURPOSE: To investigate the molecular composition of subretinal fluid (SRF) in central serous chorioretinopathy (CSCR) and rhegmatogenous retinal detachment (RRD) using proteomics and metabolomics. METHODS: SRF was obtained from one patient with severe nonresolving bullous CSCR requiring surgical subretinal fibrin removal, and two patients with long-standing RRD. Proteins were trypsin-digested, labeled with Tandem-Mass-Tag and fractionated according to their isoelectric point for identification and quantification by tandem mass spectrometry. Independently, metabolites were extracted on cold methanol/ethanol, and identified by untargeted ultra-high performance liquid chromatography and high-resolution mass spectrometry. Bioinformatics analyses were conducted. RESULTS: In total, 291 proteins and 651 metabolites were identified in SRF samples. Compared with RRD, 128 proteins (77 downregulated; 51 upregulated) and 76 metabolites (43 downregulated; 33 upregulated) differed in the SRF from CSCR. Protein and metabolites notably deregulated in CSCR were related to glycolysis/gluconeogenesis, inflammation (including serum amyloid P component, versican), alternative complement pathway (complement factor H and complement factor H-related protein), cellular adhesion, biliary acid metabolism (farnesoid X receptor/retinoid X receptor), and gluco- and mineralocorticoid systems (aldosterone, angiotensin, and corticosteroid-binding globulin). CONCLUSIONS: Proteomics and metabolomics can be performed on SRF. A unique SRF sample from CSCR exhibited a distinct molecular profile compared with RRD. TRANSLATIONAL RELEVANCE: This first comparative multiomics analysis of SRF improved the understanding of CSCR and RRD pathophysiology. It identified pathways potentially involved in the better photoreceptor preservation in CSCR, suggesting neuroprotective targets that will require additional confirmation.

11.
Graefes Arch Clin Exp Ophthalmol ; 255(9): 1749-1756, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28608271

ABSTRACT

PURPOSE: Subretinal perfluorocarbon liquid (PFCL) is a serious complication that can occur after retinal detachment repair. It is possible to remove the PFCL surgically, but retinal damage related to the procedure is unknown. Also, increasing interest in subretinal treatment makes it relevant to examine the functional and morphological consequences of repeated subretinal manipulation. We hypothesized that PFCL in a porcine model can be injected in the subretinal space and removed with minimal effect on retinal structure and function. METHODS: The left eyes of ten healthy three-month-old female domestic pigs were included. Multifocal electroretinograms (mfERG) were recorded before surgery. Following vitrectomy, a PFCL bleb (decalin) was injected subretinally using a 41G cannula. After 14 days the decalin was removed through a 41G cannula in combination with a 2 ml syringe and an intermediate flexible tube. Two weeks after removal, a control mfERG was recorded, the pigs were enucleated and sacrificed and eyes were examined histologically. All statistics were carried out with a paired t-test in SAS Enterprise Guide 7.1® (SAS Institute Inc., Cary, NC, USA). RESULTS: There was no significant difference in mfERG amplitude ratio (left/right eye) between baseline and recordings two weeks after removal of decalin (P1 (M = 0.26, SD = 0.80, p = 0.39), second order kernel (M = -0.18, SD = 0.86, p = 0.57), Direct Response (M = 0.39, SD = 0.61, p = 0.12) or Induced Component (M = -0.03, SD = 0.40, p = 0.80)). Histologically, the photoreceptor outer segments were minimally affected. Otherwise the retina was normal 14 days after removal of decalin. In four pigs the subretinal decalin displaced inferiorly and was no longer accessible for removal. CONCLUSION: Subretinal decalin can be removed within 14 days without lasting retinal damage. Decalin is a heavy liquid where the risk of displacement is high. Future studies using PFCLs to control duration of an experimental retinal separation should focus on PFCLs that are isodense to the vitreus body.


Subject(s)
Endotamponade/methods , Naphthalenes/administration & dosage , Retina/surgery , Retinal Detachment/surgery , Robotic Surgical Procedures/methods , Vitrectomy/methods , Animals , Disease Models, Animal , Electroretinography , Female , Follow-Up Studies , Injections, Intraocular , Reoperation , Retina/drug effects , Retina/pathology , Retinal Detachment/diagnosis , Retinal Detachment/physiopathology , Subretinal Fluid , Swine
12.
Surv Ophthalmol ; 62(6): 723-769, 2017.
Article in English | MEDLINE | ID: mdl-28506603

ABSTRACT

Exudative retinal detachment develops when fluid collects in the subretinal space. The subretinal space between the photoreceptors and the retinal pigment epithelium is the remnant of the embryonic optic vesicle. In the developed eye the subretinal space is of minimal size, but it can reopen under pathological conditions that disrupt the integrity of blood-retinal barrier. Inflammatory, infectious, infiltrative, neoplastic, vascular, and degenerative conditions may be associated with blood-retinal barrier breakdown and the sequential development of exudative retinal detachment. We elaborate on the pathogenesis and the differential diagnosis of exudative retinal detachment and specifically discuss the spectrum of diseases associated with exudative retinal detachment in uveitis clinics.


Subject(s)
Retinal Detachment , Angiogenesis Inhibitors/therapeutic use , Choroid Diseases/complications , Diagnosis, Differential , Humans , Immunosuppressive Agents/therapeutic use , Retinal Detachment/diagnosis , Retinal Detachment/pathology , Retinal Detachment/therapy , Retinal Pigment Epithelium/pathology , Risk Factors
13.
Methods Mol Biol ; 1565: 215-227, 2017.
Article in English | MEDLINE | ID: mdl-28364246

ABSTRACT

Neonatal in vivo electroporations and retinal explant electroporations have been widely employed in understanding the effects of loss or gain of function of protein-coding genes in retinal development. Here, we describe a rapid and efficient delivery of morpholinos to add another tool to perturb gene expression during mouse retinal development.


Subject(s)
Electroporation , Gene Expression Regulation , Morpholinos/genetics , Retina/metabolism , Animals , Electroporation/methods , Gene Expression , Gene Transfer Techniques , Genes, Reporter , Mice , Microscopy, Fluorescence , Morpholinos/administration & dosage
14.
Elife ; 52016 07 20.
Article in English | MEDLINE | ID: mdl-27438413

ABSTRACT

Photo-transduction in cone segments (CS) is crucial for high acuity daytime vision. For ill-defined reasons, CS degenerate in retinitis pigmentosa (RP) and in the transitional zone (TZ) of atrophic zones (AZ), which characterize geographic atrophy (GA). Our experiments confirm the loss of cone segments (CS) in the TZ of patients with GA and show their association with subretinal CD14(+)mononuclear phagocyte (MP) infiltration that is also reported in RP. Using human and mouse MPs in vitro and inflammation-prone Cx3cr1(GFP/GFP) mice in vivo, we demonstrate that MP-derived IL-1ß leads to severe CS degeneration. Our results strongly suggest that subretinal MP accumulation participates in the observed pathological photoreceptor changes in these diseases. Inhibiting subretinal MP accumulation or Il-1ß might protect the CS and help preserve high acuity daytime vision in conditions characterized by subretinal inflammation, such as AMD and RP.


Subject(s)
Geographic Atrophy/pathology , Geographic Atrophy/physiopathology , Interleukin-1beta/metabolism , Phagocytes/immunology , Retina/pathology , Retinal Cone Photoreceptor Cells/physiology , Animals , Humans , Mice
15.
Adv Exp Med Biol ; 854: 315-21, 2016.
Article in English | MEDLINE | ID: mdl-26427427

ABSTRACT

We present an optimized surgical technique for feline retinal detachment which allows for natural re-attachment, reduces retinal scarring and vitreal bands, and allows central placement of the detachment in close proximity to the optic nerve. This enables imaging via Optical Coherence Tomography (OCT) and multifocal electroretinography (mfERG) analysis. Ideal detachment conditions involve a lensectomy followed by a three-port pars plana vitrectomy. A 16-20 % retinal detachment is induced by injecting 8 % C3F8 gas into the subretinal space in the central retina with a 42G cannula. The retinal detachment resolves approximately 6 weeks post-surgery. Imaging is enhanced by using a 7.5 and 20 diopter lens for OCT and mfERG fundus imaging, respectively, to compensate for the removed lens.


Subject(s)
Cat Diseases/surgery , Retina/surgery , Retinal Detachment/surgery , Vitrectomy/methods , Animals , Cat Diseases/diagnosis , Cat Diseases/physiopathology , Cats , Electroretinography , Fundus Oculi , Retina/pathology , Retinal Detachment/diagnosis , Retinal Detachment/physiopathology , Time Factors , Tomography, Optical Coherence , Treatment Outcome
16.
Prog Retin Eye Res ; 51: 90-106, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26344734

ABSTRACT

Rod cell oxidative stress is a major pathogenic factor in retinal disease, such as diabetic retinopathy (DR) and retinitis pigmentosa (RP). Personalized, non-destructive, and targeted treatment for these diseases remains elusive since current imaging methods cannot analytically measure treatment efficacy against rod cell compartment-specific oxidative stress in vivo. Over the last decade, novel MRI-based approaches that address this technology gap have been developed. This review summarizes progress in the development of MRI since 2006 that enables earlier evaluation of the impact of disease on rod cell compartment-specific function and the efficacy of anti-oxidant treatment than is currently possible with other methods. Most of the new assays of rod cell compartment-specific function are based on endogenous contrast mechanisms, and this is expected to facilitate their translation into patients with DR and RP, and other oxidative stress-based retinal diseases.


Subject(s)
Antioxidants/therapeutic use , Diabetic Retinopathy/physiopathology , Magnetic Resonance Imaging , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/physiopathology , Animals , Calcium Channels, L-Type/metabolism , Diabetic Retinopathy/drug therapy , Disease Models, Animal , Humans , Oxidative Stress , Retinitis Pigmentosa/drug therapy
17.
Invest Ophthalmol Vis Sci ; 56(3): 1916-23, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25736794

ABSTRACT

PURPOSE: Calcium regulates many functions of the RPE. Its concentration in the subretinal space and RPE cytoplasm is closely regulated. Transient receptor potential (TRP) channels are a superfamily of ion channels that are moderately calcium-selective. This study investigates the subcellular localization and potential functions of TRP channels in a first-passage culture model of human fetal RPE (hfRPE). METHODS: The RPE isolated from 15- to 16-week gestation fetuses were maintained in serum-free media. Cultures were treated with barium chloride (BaCl2) in the absence and presence of TRP channel inhibitors and monitored by the transepithelial electrical resistance (TER). The expression of TRP channels was determined using quantitative RT-PCR, immunoblotting, and immunofluorescence confocal microscopy. RESULTS: Barium chloride substantially decreased TER and disrupted cell-cell contacts when added to the apical surface of RPE, but not when added to the basolateral surface. The effect could be partially blocked by the general TRP inhibitor, lanthanum chloride (LaCl3, ~75%), or an inhibitor of calpain (~25%). Family member-specific inhibitors, ML204 (TRPC4) and HC-067047 (TRPV4), had no effect on basal channel activity. Expression of TRPC4, TRPM1, TRPM3, TRPM7, and TRPV4 was detected by RT-PCR and immunoblotting. The TRPM3 localized to the base of the primary cilium, and TRPC4 and TRPM3 localized to apical tight junctions. The TRPV4 localized to apical microvilli in a small subset of cells. CONCLUSIONS: The TRP channels localized to subdomains of the apical membrane, and BaCl2 was only able to dissociate tight junctions when presented to the apical membrane. The data suggest a potential role for TRP channels as sensors of [Ca(2+)] in the subretinal space.


Subject(s)
Cell Membrane/metabolism , Fetus/metabolism , Gene Expression Regulation , RNA/genetics , Retinal Pigment Epithelium/metabolism , Transient Receptor Potential Channels/genetics , Biological Transport , Cells, Cultured , Fetus/cytology , Humans , Immunoblotting , Immunohistochemistry , Membrane Potentials , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/embryology , Tight Junctions , Transient Receptor Potential Channels/biosynthesis
18.
Neurosci Lett ; 558: 8-13, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24076256

ABSTRACT

The retinal ischemia-reperfusion injury (RIR) is a common pathological process that leads to progressive visual loss and blindness in many retinal diseases such as retinal vascular occlusion disease, diabetic retinopathy, and acute glaucoma. Currently, there has been no effective therapy. The purpose of this study was to investigate the effects of transplantation of retinal progenitor cells (RPCs) into the subretinal space (SRS) and the superior colliculus (SC) in a rat model of RIR injury. We used cultured postnatal day 1 rat RPCs transfected with adeno-associated virus containing the cDNA encoding enhanced green fluorescence protein (EGFP) for transplantation. RIR injury was induced by increases in the intraocular pressure to 110 mmHg for 60 min. The effects of transplantation were evaluated by immunohistochemistry, electroretinography (ERG), and visual evoked potentials (VEP). We found that in rats with RIR injury, RPCs transplanted into the SRS and the SC survived for at least 8 weeks, migrated into surrounding tissues, and improved the ERG and VEP responses. Cells transplanted into the SC improved the VEP response more than those transplanted into the SRS. Our data suggest that transplantation of RPCs into the SRS and the SC may be a possible method for cell replacement therapy for retinal diseases.


Subject(s)
Neural Stem Cells/transplantation , Reperfusion Injury/therapy , Retinal Diseases/therapy , Retinal Neurons/transplantation , Animals , Animals, Newborn , Cells, Cultured , Electroretinography , Evoked Potentials, Visual , Male , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Retina/pathology , Retinal Diseases/physiopathology , Superior Colliculi
19.
Int J Ophthalmol ; 3(3): 216-9, 2010.
Article in English | MEDLINE | ID: mdl-22553557

ABSTRACT

UNLABELLED: CORRESPONDENCE TO: Fang-Tian Dong. Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China. d_fangtian@sina.com AIM: To access the differentiation of rat mesenchymal stem cell (MSC) in the microenvironment of retinal degeneration induced by the administration of sodium iodate. METHODS: In-vitro cultured Lewis rat MSC were injected into the sub-retinal space of NaIO(3) induced retinal degeneration rat eyes (30g/L NaIO(3) 100mg/kg). To observe the trace and differentiation of MSC by immuno-fluorescent method successively in 5 weeks after the surgery. RESULTS: The majority of the transplanted cells stay in retinal pigment epithelium layer and cones & rods layer. From the 2(nd) week after transplantation, the engrafted MSC express PCK and rhodopsin under fluorescent microscope. CONCLUSION: MSC can survive mainly in the outer layer of retina in the microenvironment of retinal degeneration and differentiate forward the RPE cell and photoreceptor.

20.
International Eye Science ; (12): 1851-1854, 2010.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-641417

ABSTRACT

AIM: To assess the differentiation of rat mesenchymal stem cell (MSC) in the microenvironment of retinitis pigmentosa(RP) induced by the administration of sodium iodate. METHODS: In vitro cultured Lewis rat MSC were injected into the subretinal space of NaIO3 induced RP rat eyes (30g/L NaIO3 100mg/kg). To observe the trace and differentiation of MSC by immuno-fluorescent method successively in 5 weeks after the surgery.RESULTS: The majority of the transplanted cells stay in retinal pigment epithelium(RPE) layer and cones and rods layer. From the 2nd week after transplantation, the engrafted MSC expressed PCK and rhodopsin under fluorescent microscope.CONCLUSION: MSC can survive mainly in the outer layer of retina in the microenvironment of RP and differentiate forward the RPE cell and photoreceptor.

SELECTION OF CITATIONS
SEARCH DETAIL