Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Biochem Biophys Res Commun ; 496(3): 814-819, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29395082

ABSTRACT

Channelrhodopsin-2 (ChR2), a light-activated cation-selective ion channel, has been widely used as a tool in optogenetic research. ChR2 is specifically sensitive to wavelengths less than 550 nm. One of the methods to expand the sensitivity of a channelrhodopsin to a wider range of wavelengths is to express another channelrhodopsin in the cells by the transduction of an additional gene. Here, we report the characteristic features of cells expressing two types of channelrhodopsins, each having different wavelength sensitivities. In HEK293 cells stably expressing ChR2, photocurrents were elicited at stimuli of 400-550 nm, and the wavelength sensitivity range was expanded by the additional transduction of the modified Volvox channelrhodopsin-1 (mVChR1) gene, which has broad wavelength sensitivities, ranging from 400 to 600 nm. However, the photocurrent at 550 nm was lower than that of the mVChR1-expressing cell; moreover, the turning-on and turning-off constants were delayed, and the deactivation rates were decreased. Meanwhile, the response to lower light intensity was improved by the additional gene. Thus, the transduction of an additional gene is a useful method to improve the light and wavelength sensitivities, as well as photocurrent kinetic profiles, of channelrhodopsins.


Subject(s)
Channelrhodopsins/physiology , Channelrhodopsins/radiation effects , Ion Channel Gating/physiology , Ion Channel Gating/radiation effects , Light Signal Transduction/physiology , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Dose-Response Relationship, Radiation , HEK293 Cells , Humans , Kinetics , Light , Radiation Dosage
2.
Pak J Med Sci ; 33(2): 276-279, 2017.
Article in English | MEDLINE | ID: mdl-28523021

ABSTRACT

OBJECTIVE: To evaluate the single incision laparoscopic appendectomy (SILA) using existing instruments, the 10-mm laparoscope, and glove port technique. METHODS: SILA was performed on 16 patients (8 male cases, 8 female cases) between June 2012 and September 2015. A 20-mm incision was made in the umbilicus and a wound retractor was placed. A 10-mm trocar for the laparoscope and two 5-mm trocars were fixed to the three fingers of the latex gloves and it was attached to the wound retractor. Another thin forceps were inserted from right low abdomen. RESULTS: Average age of patients was 32.6 ± 17.7 years. Preoperative average white blood cell was 13,325 ± 4,584 /mm3, and average CRP was 1.81 ± 3.70 mg/dL. Preoperative body temperature was 36.8 ± 0.5°C. The mean appendix size was 9.6 ± 2.3 mm and none of the patients had an abscess on preoperative CT. The CT also revealed a fecal pellet in 5/16 (31%) of patients. Mean operation time was 66.4 ± 25.4 minutes, and minimal intraoperative bleeding was observed in all patients. Average hospital stay was 5.3 ± 1.9 days and none of the patients had complications. CONCLUSION: SILA using the 10-mm laparoscope and glove port technique may be a safe and feasible operation for mild to moderate appendicitis.

3.
Sci Rep ; 7: 41210, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28112267

ABSTRACT

Optogenetic technologies are expected to be applicable for clinical use in restoring vision. However, the degree of recovered visual function is highly dependent on the function of the chosen optogenetic gene. To investigate the effect on visual function of dual expression of genes with different wavelength sensitivities, we transduced a modified Volvox-derived channelrhodopsin gene (mVChR1) via an adeno-associated virus vector into transgenic rats harbouring the ChR2 gene in retinal ganglion cells. These transgenic rats were given an intraperitoneal injection of N-methyl-N-nitrosourea to induce the degeneration of native photoreceptor cells prior to transduction of mVChR1. Optical coherence tomography images indicated the degeneration of the native photoreceptor cells after the N-methyl-N-nitrosourea injection. Complete loss of function of the native photoreceptor cells was confirmed using electroretinograms. In the ChR2 transgenic rats, visually evoked potentials were clearly detectable in spite of native photoreceptor function abolishment; however the responses were limited to within blue wavelengths. In contrast, the limited wavelength sensitivities were improved by the additional transduction of mVChR1, which exhibited sensitivities to green and red. Thus, the transductions of dual genes encoding channelrhodopsins that exhibit different wavelength sensitivities represents a promising candidate method to expand and to enhance rescued wavelength sensitivities in blind subjects.


Subject(s)
Channelrhodopsins/genetics , Optogenetics , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/physiology , Animals , Electroretinography , Evoked Potentials, Visual , Genetic Vectors , Methylnitrosourea/administration & dosage , Photic Stimulation , Photoreceptor Cells, Vertebrate/drug effects , Rats , Rats, Transgenic , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Tomography, Optical Coherence , Volvox/genetics
4.
Mol Ther ; 22(8): 1434-1440, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24821344

ABSTRACT

We previously showed that blind rats whose vision was restored by gene transfer of Chlamydomonas channelrhodopsin-2 (ChR2) could only detect wavelengths less than 540 nm because of the action spectrum of the transgene product. Volvox-derived channelrhodopsin-1, VChR1, has a broader spectrum than ChR2. However, the VChR1 protein was mainly localized in the cytoplasm and showed weak ion channel properties when the VChR1 gene was transfected into HEK293 cells. We generated modified Volvox channelrhodopsin-1 (mVChR1), which is a chimera of Volvox channelrhodopsin-1 and Chlamydomonas channelrhodopsin-1 and demonstrated increased plasma membrane integration and dramatic improvement in its channel properties. Under whole-cell patch clamp, mVChR1-expressing cells showed a photo-induced current upon stimulation at 468-640 nm. The evoked currents in mVChR1-expressing cells were ~30 times larger than those in VChR1-expressing cells. Genetically, blind rats expressing mVChR1 via an adeno-associated virus vector regained their visual responses to light with wavelengths between 468 and 640 nm and their recovered visual responses were maintained for a year. Thus, mVChR1 is a candidate gene for gene therapy for restoring vision, and gene delivery of mVChR1 may provide blind patients access to the majority of the visible light spectrum.


Subject(s)
Blindness/therapy , Genetic Therapy/methods , Retina/physiopathology , Rhodopsin/metabolism , Volvox/genetics , Animals , Blindness/metabolism , Chlamydomonas/genetics , Dependovirus/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , HEK293 Cells , Humans , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhodopsin/genetics
5.
J Ophthalmol ; 2013: 185825, 2013.
Article in English | MEDLINE | ID: mdl-24319591

ABSTRACT

The retina is constantly subjected to oxidative stress, which is countered by potent antioxidative systems present in retinal pigment epithelial (RPE) cells. Disruption of these systems leads to the development of age-related macular degeneration. Thioredoxin 2 (Trx2) is a potent antioxidant, which acts directly on mitochondria. In the present study, oxidative stress was induced in the human RPE cell line (ARPE-19) using 4-hydroxynonenal (4-HNE) or C2-ceramide. The protective effect of Trx2 against oxidative stress was investigated by assessing cell viability, the kinetics of cell death, mitochondrial metabolic activity, and expression of heat shock proteins (Hsps) in Trx2-overexpressing cell lines generated by transfecting ARPE cells with an adeno-associated virus vector encoding Trx2. We show that overexpression of Trx2 reduced cell death induced by both agents when they were present in low concentrations. Moreover, early after the induction of oxidative stress Trx2 played a key role in the maintenance of the cell viability through upregulation of mitochondrial metabolic activity and inhibition of Hsp70 expression.

6.
BMC Ophthalmol ; 13: 19, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23683117

ABSTRACT

BACKGROUND: Numerous rodent models of photoreceptor degeneration have been developed for the study of visual function. However, no viable model has been established in a species that is more closely related to Homo sapiens. Here, we present a rabbit model of monocular photoreceptor degeneration. METHODS: We tested 2 chemicals, verteporfin and sodium nitroprusside (SNP), for developing a 1-eye limited photoreceptor degeneration model in pigmented rabbits. After the intravenous injection of verteporfin, the retina was exposed to light from a halogen lamp for 0, 10, 30, or 60 min. Alternately, 100 µL of various concentrations of sodium nitroprusside (0.1 mM, 0.5 mM, and 1 mM) were intravitreously injected into the rabbit eye. Retinal degeneration was evaluated by fundus photography, electroretinogram (ERG), and histological examinations. RESULTS: Fundus photographs of animals in the verteporfin- or SNP-treated groups showed evidence of retinal degeneration. The severity of this degradation depended on the duration of light exposure and the concentration of SNP administered. The degeneration was clearly limited to the light-exposed areas in the verteporfin-treated groups. Extensive retinal atrophy was observed in the SNP-treated groups. The a- and b-wave amplitudes were dramatically decreased on the ERGs from SNP-treated groups. Histological examination revealed that either verteporfin or SNP induced severe photoreceptor degeneration. High-dose SNP treatment (1 mM) was also associated with inner retinal layer degeneration. CONCLUSIONS: Both SNP and verteporfin clearly caused photoreceptor degeneration without any effect on the contralateral eye. These compounds therefore represent valuable tools for the empirical investigation of visual function recovery. The findings will inform guidelines for clinical applications such as retinal prostheses, cell-based therapy, and gene therapy.


Subject(s)
Nitroprusside , Porphyrins , Retinal Degeneration/chemically induced , Animals , Disease Models, Animal , Electroretinography , Injections, Intravenous , Light , Photography , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/radiation effects , Photosensitizing Agents/pharmacology , Rabbits , Retinal Degeneration/pathology , Vasodilator Agents/pharmacology , Verteporfin
7.
Cell Struct Funct ; 38(1): 81-8, 2013.
Article in English | MEDLINE | ID: mdl-23485938

ABSTRACT

Age-related macular degeneration (AMD) affects the retina and is the most common cause of blindness in elderly persons in developed countries. The retina is constantly subjected to oxidative stress; to avoid the effects of oxidative stress, retinal pigment epithelial (RPE) cells possess potent anti-oxidant systems. Disruption of these systems leads to dysfunction of RPE cells, which then accelerates the development of AMD. Here, we investigated the role of thioredoxins (TRXs), scavengers of intracellular reactive oxygen species, by assessing the effect of TRX overexpression on cell viability, morphology, NF-κB expression, and mitochondrial membrane potential, in RPE cells. TRX-overexpressing cell lines were generated by infection of an established human RPE cell line (ARPE) with adeno-associated virus vectors encoding either TRX1 or TRX2. We showed that overexpression of TRXs reduced cell death caused by 4-hydroxynonenal (4-HNE)-induced oxidative stress; TRX2 was more effective than TRX1 in promoting cell survival. 4-HNE caused perinuclear NF-κB accumulation, which was absent in TRX-overexpressing cells. Moreover, overexpression of TRXs prevented depolarization of mitochondrial membranes; again, TRX2 was more effective than TRX1 in maintaining the membrane potential. The difference in the protective effects of these TRXs against oxidative stress may be due to their expression profile. TRX2 was expressed in the mitochondria, while TRX1 was expressed in the cytoplasm. Thus, TRX2 may directly protect mitochondria by preventing depolarization. These results demonstrate that TRXs are potent antioxidant proteins in RPE cells and their direct effect on mitochondria may be a key to prevent oxidative stress.


Subject(s)
Antioxidants , Thioredoxins , Aldehydes/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Humans , Macular Degeneration , Membrane Potential, Mitochondrial/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Retina/cytology , Retina/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism
8.
J Mol Neurosci ; 46(2): 393-400, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21792608

ABSTRACT

The objective of this study is to investigate age-related differences in recovered visual function in Royal College of Surgeons (RCS) rats transduced with the Channelrhodopsin-2 (ChR2) gene. An adeno-associated virus vector that contained ChR2 was injected intravitreously into young or aged RCS rats. After 4 months, visual evoked potentials were recorded. To estimate the transduction efficiencies, ChR2V-expressing cells and retrograde labeled retinal ganglion cells (RGCs) were counted. After photoreceptor degradation, immunohistochemistry was used to detect glial fibrillary acidic protein (GFAP) in the retinas. The amplitudes and latencies from young RCS rats were higher and shorter, respectively, than those from aged RCS rats. ChR2V was expressed in the RGCs of both groups of rats; there was no significant difference in the transduction efficiency of either group. However, the number of RGCs in aged RCS rats was significantly less than that in young RCS rats. In addition, strong GFAP immunoreactivity was observed after photoreceptor degeneration, whereas it was weaker in ChR2V-expressing RGCs. ChR2 transduction produced photosensitive RGCs in both young and aged rats. However, the degree of recovery depended on the age at the time of transduction.


Subject(s)
Carrier Proteins/physiology , Genetic Therapy , Genetic Vectors/therapeutic use , Nerve Degeneration/pathology , Retinal Dystrophies/pathology , Retinal Ganglion Cells/pathology , Transduction, Genetic , Age Factors , Animals , Carrier Proteins/genetics , Dependovirus/genetics , Evoked Potentials, Visual , Eye Proteins/analysis , Glial Fibrillary Acidic Protein/analysis , Models, Animal , Nerve Degeneration/prevention & control , Peptide Fragments/genetics , Peptide Fragments/physiology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Rats , Rats, Mutant Strains , Reaction Time , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Retinal Dystrophies/therapy , Retinal Ganglion Cells/chemistry , Retinal Ganglion Cells/physiology , c-Mer Tyrosine Kinase
9.
Dev Growth Differ ; 53(3): 357-65, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21477161

ABSTRACT

We attempted to test whether the differentiated NIH/3T3 fibroblasts could be differentiated into neuronal cells without any epigenetic modification. First, a neurosphere assay was carried out, and we successfully generated neurosphere-like cells by floating cultures of NIH/3T3 fibroblasts in neural stem cell medium. These spheres have the ability to form sub-spheres after three passages, and express the neural progenitor markers Nestin, Sox2, Pax6, and Musashi-1. Second, after shifting to a differentiating medium and culturing for an additional 8 days, cells in these spheres expressed the neuronal markers ß-tubulin and neurofilament 200 and the astrocytic marker glial fibrillary acidic protein (GFAP). Finally, after treating the spheres with all-trans retinoic acid and taurine, the expression of ß-tubulin was increased and the staining of photoreceptor markers rhodopsin and recoverin was observed. The present study shows that NIH/3T3 fibroblasts can generate neurosphere-like, neuron-like, and even photoreceptor-like cells under defined conditions, suggesting that the differentiated non-neuronal cells NIH/3T3 fibroblasts, but not pluripotent cells such as embryonic stem cells or induced pluripotent stem cells, may have the potential to be transdifferentiated into neuronal cells without adding any epigenetic modifier. This transdifferentiation may be due to the possible neural progenitor potential of NIH/3T3 fibroblasts that remains dormant under normal conditions.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Differentiation , Fibroblasts/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Spheroids, Cellular/metabolism , Animals , Cell Culture Techniques , Fibroblasts/cytology , Mice , NIH 3T3 Cells , Neurons/cytology , Spheroids, Cellular/cytology
10.
Jpn J Ophthalmol ; 54(2): 156-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20401566

ABSTRACT

BACKGROUND: To examine whether different types of retinal ganglion cells (RGCs) in the cat retina have different survival rates when exposed to N-methyl-D-aspartate (NMDA). METHODS: NMDA injury was induced by intravitreal administration of NMDA at final concentrations of 0.2, 0.4, 0.6, and 0.8 mM. The total number of surviving RGCs and their distribution were counted by retrograde labeling with a fluorescent dye. Measurements of the proportions of the main RGC types (alpha, beta, and neither alpha nor beta cells) were obtained by using intracellular injections of Lucifer yellow. RESULTS: The mean percentage of surviving RGCs in the NMDA-injected retina was reduced to 59.4% (0.2 mM NMDA), 35.8% (0.4 mM), 10.8% (0.6 mM), and 14.1% (0.8 mM). At 0.2 mM, the survival rate of alpha cells was reduced to 56%, but that of beta cells remained at 81%. At 0.8 mM, the survival rate of alpha cells was 19%, while beta cells rapidly decreased to 9.9%. No difference was detected in NMDA vulnerability between ON- and OFF-center RGCs. CONCLUSIONS: Different RGC types display different susceptibilities to NMDA injury. A specific examination of the functions of different types of RGCs would be helpful in detecting retinal excitotoxicity such as in chronic retinal ischemia.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , N-Methylaspartate/toxicity , Retinal Ganglion Cells/drug effects , Animals , Cats , Cell Count , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Injections , Isoquinolines/administration & dosage , Phenotype , Retinal Ganglion Cells/cytology , Vitreous Body
11.
Mol Vis ; 16: 378-91, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20300562

ABSTRACT

PURPOSE: To characterize an N-ethyl-N-nitrosourea-induced dominant mouse mutant, M-1156, that exhibits progressive retinal degeneration and to investigate the pathogenesis of the retinal phenotype in the mutant. METHODS: A positional candidate gene approach was used to identify the causative gene in the M-1156 mutant. Funduscopic examination, light microscopy, transmission electron microscopy, and electroretinography were performed to analyze the M-1156 phenotype. Real-time quantitative PCR, immunohistochemistry, and western blotting were also performed. RESULTS: Linkage analysis enabled the mutant gene to be mapped to a region of chromosome 19 containing Rom1, which encodes rod outer segment membrane protein 1. Sequence analysis demonstrated that the mutation consisted of a single base T-->C substitution at position 1,195 in Rom1 (M96760, National Center for Biotechnology Information [NCBI]) and that the mutant allele was expressed. A putative missense mutation designated Rom1(Rgsc1156) that was identified in the M-1156 mutant mouse causes a Trp to Arg substitution at position 182 in the translated protein. Rom1(Rgsc1156) heterozygotes were found to have a mottled retina and narrowed arteries in the fundus oculi. Photomicrographs of the retina revealed significant differences among the genotypes in the thickness of the outer nuclear layer and in the length of the outer segments of the photoreceptors. The alterations were more marked in the homozygotes than in the heterozygotes. Electron micrographs showed that the diameters of the discs varied in the heterozygotes and that the discs were more compactly stacked than in the wild type. There were significant differences among the genotypes in the amplitude of the a-wave in single-flash electroretinograms, but there were no significant differences among the photopic electroretinograms. Real-time quantitative PCR showed that there were no significant differences among the genotypes in Rom1 or peripherin/rds (Prph2) mRNA levels relative to the rhodopsin (Rho) mRNA level. Rom1 and Prph2 immunoreactivity were decreased in the retinas of the Rom1(Rgsc1156) mutants. Semiquantitative western blot analysis of retinas from 3-week-old Rom1(Rgsc1156) mutants demonstrated significant decreases in Rom1, Prph2, and Rho protein levels in all of the genotypes. CONCLUSIONS: The Trp182Arg substitution in Rom1(Rgsc1156) mutants causes retinal degeneration. The results suggested that Trp182Arg mutant Rom1 causes a decrease in the levels of wild-type Prph2 and Rom1, which in turn cause a reduction in the level of Prph2 containing tetramers in the disc rim region and ultimately result in unstable, disorganized outer segments and photoreceptor degeneration.


Subject(s)
Eye Proteins/genetics , Genes, Dominant/genetics , Membrane Proteins/genetics , Mutagenesis , Mutation/genetics , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Animals , Base Sequence , DNA Mutational Analysis , Electroretinography , Ethylnitrosourea , Eye Proteins/metabolism , Fundus Oculi , Gene Expression Regulation , Haplotypes/genetics , Immunohistochemistry , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Peripherins , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/ultrastructure , Retinal Degeneration/metabolism , Tetraspanins
12.
Exp Eye Res ; 90(3): 429-36, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20036655

ABSTRACT

To test the hypothesis that transduction of the channelrhodopsin-2 (ChR2) gene, a microbial-type rhodopsin gene, into retinal ganglion cells of genetically blind rats will restore functional vision, we recorded visually evoked potentials and tested the experimental rats for the presence of optomotor responses. The N-terminal fragment of the ChR2 gene was fused to the fluorescent protein Venus and inserted into an adeno-associated virus to make AAV2-ChR2V. AAV2-ChR2V was injected intravitreally into the eyes of 6-month-old dystrophic RCS (rdy/rdy) rats. Visual function was evaluated six weeks after the injection by recording visually evoked potentials (VEPs) and testing optomotor responses. The expression of ChR2V in the retina was investigated histologically. We found that VEPs could not be recorded from 6-month-old dystrophic RCS rats that had not been injected with AAV2-ChR2V. In contrast, VEPs were elicited from RCS rats six weeks after injection with AAV2-ChR2V. The VEPs were recorded at stimulation rates <20Hz, which was the same as that of normal rats. Optomotor responses were also significantly better after the AAV2-ChR2V injection. Expression of ChR2V was observed mainly in the retinal ganglion cells. These findings demonstrate that visual function can be restored in blind rats by transducing the ChR2V gene into retinal ganglion cells.


Subject(s)
Blindness/therapy , Carrier Proteins/genetics , Dependovirus/genetics , Genetic Therapy/methods , Retinal Degeneration/therapy , Retinal Ganglion Cells/metabolism , Animals , Blindness/genetics , Blindness/physiopathology , Evoked Potentials, Visual/physiology , Gene Expression , Male , Nystagmus, Optokinetic/physiology , Photic Stimulation , Rats , Rats, Mutant Strains , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Stilbamidines/metabolism , Transduction, Genetic
13.
PLoS One ; 4(11): e7679, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19893752

ABSTRACT

Channelrhodopsin-2 (ChR2), one of the archea-type rhodopsins from green algae, is a potentially useful optogenetic tool for restoring vision in patients with photoreceptor degeneration, such as retinitis pigmentosa. If the ChR2 gene is transferred to retinal ganglion cells (RGCs), which send visual information to the brain, the RGCs may be repurposed to act as photoreceptors. In this study, by using a transgenic rat expressing ChR2 specifically in the RGCs under the regulation of a Thy-1.2 promoter, we tested the possibility that direct photoactivation of RGCs could restore effective vision. Although the contrast sensitivities of the optomotor responses of transgenic rats were similar to those observed in the wild-type rats, they were enhanced for visual stimuli of low-spatial frequency after the degeneration of native photoreceptors. This result suggests that the visual signals derived from the ChR2-expressing RGCs were reinterpreted by the brain to form behavior-related vision.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , Rhodopsin/biosynthesis , Rhodopsin/genetics , Thy-1 Antigens/genetics , Vision, Ocular , Animals , Animals, Genetically Modified , Behavior, Animal , Brain/metabolism , Mice , Models, Biological , Neurons/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Rats , Retinal Ganglion Cells/metabolism , Thy-1 Antigens/metabolism
14.
J Genet ; 88(4): 409-15, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20090204

ABSTRACT

Photoreceptor cells are the only retinal neurons that can absorb photons. Their degeneration due to some diseases or injuries leads to blindness. Retinal prostheses electrically stimulating surviving retinal cells and evoking a pseudo light sensation have been investigated over the past decade for restoring vision. Currently, a gene therapy approach is under development. Channelrhodopsin-2 derived from the green alga Chlamydomonas reinhardtii, is a microbial-type rhodopsin. Its specific characteristic is that it functions as a light-driven cation-selective channel. It has been reported that the channelrhodopsin-2 transforms inner light-insensitive retinal neurons to light-sensitive neurons. Herein, we introduce new strategies for restoring vision by using channelrhodopsins and discuss the properties of adeno-associated virus vectors widely used in gene therapy.


Subject(s)
Algal Proteins/physiology , Blindness/therapy , Chlamydomonas reinhardtii/metabolism , Genetic Therapy/methods , Ion Pumps/physiology , Rhodopsin/physiology , Algal Proteins/genetics , Animals , Blindness/genetics , Chlamydomonas reinhardtii/genetics , Humans , Ion Pumps/genetics , Light Signal Transduction/genetics , Light Signal Transduction/physiology , Models, Biological , Rhodopsin/genetics , Signal Transduction/genetics , Signal Transduction/physiology
15.
Mol Vis ; 14: 413-7, 2008 Mar 04.
Article in English | MEDLINE | ID: mdl-18334962

ABSTRACT

PURPOSE: To examine the myocilin (MYOC) gene for mutations in Japanese patients with primary open-angle glaucoma (POAG) and to determine the phenotypes of the patients with the mutations. METHODS: One-hundred thirty-eight unrelated Japanese patients with POAG were studied. Genomic DNA was extracted from leukocytes of peripheral blood, and the three coding exons including the intron-exon boundaries were amplified by polymerase chain reaction (PCR) and directly sequenced bi-directionally. RESULTS: Two sequence variants were identified, one novel non-synonymous amino acid change (p.Gln297His) and one reported synonymous amino acid change (p.Ala363Thr). These mutations were not detected in the 118 ethnically-matched controls. p.Gln297His was found in a 70-year-old man, who developed POAG at a late age, and his intraocular pressure was high. p.Ala363Thr was found in two cases, and both patients developed POAG at an early age and had high intraocular pressures that responded poorly to medical treatment. CONCLUSIONS: Two non-synonymous variants, p.Gln297His and p.Ala363Thr, indicate that they are involved in the pathogenesis of POAG. p.Ala363Thr has been found in another Japanese population and would be useful in genetic testing.


Subject(s)
Asian People/genetics , Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Glaucoma, Open-Angle/genetics , Glycoproteins/genetics , Mutation/genetics , Aged , Base Sequence , DNA Mutational Analysis , Female , Humans , Japan , Male , Middle Aged , Pedigree
16.
Cell Struct Funct ; 33(1): 21-6, 2008.
Article in English | MEDLINE | ID: mdl-18285636

ABSTRACT

To investigate the effect of brain derived neurotrophic factor (BDNF) on the phagocytic activity in iris pigment epithelial (IPE) cells, purified porcine photoreceptor outer segments (POS) were applied to cultured IPE cells for three hours. To measure phagocytic activities, bound and total POS were differentially stained using a double immunofluorescence staining method. BDNF increased the binding of POS in IPE cells in a dose-dependent manner. Ingestion of POS, however, was not affected throughout the concentrations used in this study. To investigate the signal transduction pathways of BDNF, a phosphatidylinositol 3-kinase (PI3K) inhibitor, LY294002, and MAPK/ERK kinase (MEK) inhibitor, PD98059, were used for this study. LY294002 had no effect on the binding and ingestion of POS in BDNF-treated IPE cells. On the other hand, PD98059 completely inhibited the increase of POS binding in BDNF-treated cells and also decreased the ingestion of POS. These results indicate that increased POS binding activity by BDNF and the decreased ingestion of POS were mediated through the MAPK pathway.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Epithelial Cells/drug effects , Phagocytosis/drug effects , Animals , Blotting, Western , Cell Culture Techniques , Cells, Cultured , Chromones/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Flavonoids/pharmacology , Fluorescent Antibody Technique , Iris/cytology , Microscopy, Fluorescence , Morpholines/pharmacology , Phosphorylation/drug effects , Pigment Epithelium of Eye/cytology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Long-Evans , Rod Cell Outer Segment/metabolism , Swine
17.
J Biochem ; 142(3): 365-76, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17646173

ABSTRACT

Calpains are considered to be cytoplasmic enzymes, although several studies have shown that calpain-like protease activities also exist in mitochondria. We partially purified mitochondrial calpain from swine liver mitochondria and characterized. Only one type of mitochondrial calpain was detected by the column chromatographies. The mitochondrial calpain was stained with anti-mu-calpain and calpain small subunit antibodies. The susceptibility of mitochondrial calpain to calpain inhibitors and the optimum pH differ from those of cytosolic mu- and m-calpains. The Ca(2+)-dependency of mitochondrial calpain was similar to that of cytosolic mu-calpain. Therefore, we named the protease mitochondrial mu-like calpain. In zymogram analysis, two types of caseinolytic enzymes existed in mitochondria and showed different mobilities from cytosolic mu- and m-calpains. The upper major band was stained with anti-mu-calpain and calpain small subunit antibodies (mitochondrial calpain I, mitochondrial mu-like calpain). The lower band was stained only with anti-calpain small subunit antibody (mitochondrial calpain II, unknown mitochondrial calpain). Calpastatin was not detected in mitochondrial compartments. The mitochondrial calpain processed apoptosis-inducing factor (AIF) to truncated AIF (tAIF), releasing tAIF into the intermembrane space. These results indicate that mitochondrial calpain, which differs from mu- and m-calpains, seems to be a ubiquitous calpain and may play a role in mitochondrial apoptotic signalling.


Subject(s)
Calpain/metabolism , Mitochondria, Liver/enzymology , Animals , Apoptosis Inducing Factor/metabolism , Blotting, Western , Caseins/metabolism , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Hydrolysis , Subcellular Fractions/enzymology , Swine
18.
Invest Ophthalmol Vis Sci ; 48(8): 3821-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17652757

ABSTRACT

PURPOSE: To investigate whether the channelopsin-2 (Chop2) gene would restore visual responses in 10-month-old dystrophic Royal College of Surgeons (aged RCS; rdy/rdy) rats, the authors transferred the Chop2 gene into the retinal cells of aged RCS rats using the adenoassociated virus (AAV) vector. METHODS: The N-terminal fragment (residues 1-315) of Chop2 was fused to a fluorescent protein, Venus, in frame at the end of the Chop2 coding fragment. The viral vector construct (AAV-Chop2V) for the expression of the Chop2V in the retina was made by subcloning into an adenoassociated virus vector, including the CAG promoter. To evaluate the expression profile of Chop2V in the retina, the rats were killed and the eyes were removed and fixed with 4% paraformaldehyde in 0.1 M phosphate-buffered saline. Retinal wholemount specimens and cryosections were made. Under anesthetized conditions, electrodes for the recording of visually evoked potentials (VEPs) were implanted onto the visual cortex in aged-RCS (rdy/rdy) rats. AAV-Chop2V vectors were then injected into the vitreous cavity of the left eyes. As a control, AAV-Venus vectors were applied to the right eyes. VEPs were evoked by the flash of a blue, white, or red light-emitting diode (LED) and were recorded from the visual cortex of the rats at various time points after the AAV vector injection. RESULTS: Chop2V fluorescence was predominantly observed in retinal ganglion cells (RGCs). Some fluorescence was observed in the inner nuclear layer and the inner plexiform layer neurites. A tendency of recovery was observed in the VEPs of aged RCS (rdy/rdy) rats after the AAV-Chop2V injection but not after the AAV-Venus injection. The visual response of AAV-Chop2V-injected aged RCS (rdy/rdy) rats was less sensitive to the blue LED flash than that of nondystrophic RCS (+/+) rats. The AAV-Chop2V-injected aged RCS (rdy/rdy) rats were insensitive to the red LED flash, which evoked a robust VEP in the RCS (+/+) rats. CONCLUSIONS: The visual response of aged RCS (rdy/rdy) rats was partially restored by transduction of the Chop2 gene through AAV into the inner retinal neurons, mainly RGCs. These results suggest that the transduction of Chop2 would provide a new strategy to treat some retinitis pigmentosa (RP) symptoms independent of their etiology.


Subject(s)
Adenoviridae/genetics , Carrier Proteins/genetics , Chlamydomonas/genetics , Genetic Therapy/methods , Retinitis Pigmentosa/therapy , Aging , Animals , Disease Models, Animal , Electroretinography , Evoked Potentials, Visual , Gene Expression , Male , Molecular Sequence Data , Rats , Rats, Mutant Strains , Retinitis Pigmentosa/physiopathology
19.
Article in English | MEDLINE | ID: mdl-17474055

ABSTRACT

This paper summarizes the background and objectives of the establishment of the Tohoku University Biomedical Engineering Research Organization (TUBERO), established in July 2003 as a part of the "Development of Strategic Research Center" program, funded by the Ministry of Education, Culture, Sports, Science and Technology (MEXT) of Japan through its Special Coordination Funds for Promoting Science and Technology. This is the largest project among those funded by the budget for strategic research provided by MEXT. In relation to medical microsystems, one of the vital tasks of the biomedical engineering field selected by TUBERO, the efforts of Tohoku University and collaborative actions undertaken between Tohoku University on the one hand and local governments (Miyagi prefecture and Sendai city) and the national government on the other hand, are introduced. More than three years have passed since the launch of this five-year project; one-and-a-half years remain. It will take time for the results to take shape, but this paper will offer some insight into Tohoku University's stance toward biomedical engineering in general, including the TUBERO Project.


Subject(s)
Biomedical Engineering/organization & administration , Research/organization & administration , Universities/organization & administration , Humans , Japan
20.
J Ocul Pharmacol Ther ; 23(1): 35-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17341148

ABSTRACT

PURPOSE: The aim of this study was to determine whether indocyanine green (ICG) is toxic to cultured human retinal pigment epithelial (ARPE-19) cells, and whether hypothermia can protect the ARPE-19 cells against the ICG toxicity. METHODS: Cultured ARPE-19 cells were exposed to 0.25, 0.5, 1, 2.5, and 5 mg/mL of ICG dye at 37 and 4 degrees C for 30 min. The percentage of ARPE-19 cells that survived was determined by resazurin 1 day after the exposure. RESULTS: Exposure of the RPE cells to a hypotonic saline solution with an osmolarity equal to 5 mg/mL of ICG did not induce a statistically significant decrease in the percentage of RPE cells that survived. Exposure of the ARPE-19 cells to ICG induced a significant decrease in the percentage of cell survival at all concentrations of ICG (P<0.05), except in 0.25 mg/mL at 37 degrees C. At 4 degrees C, on the other hand, ICG induced a statistically significant decrease in the percentage of RPE cell survival only at 5 mg/mL of ICG (P<0.05). CONCLUSIONS: These results indicate that ICG is toxic to human RPE cells in culture, and that cell death cannot be attributed to the low osmolarity. Hypothermia of 4 degrees C has a protective effect against ICG toxicity.


Subject(s)
Coloring Agents/toxicity , Drug-Related Side Effects and Adverse Reactions/prevention & control , Hypothermia, Induced , Indocyanine Green/toxicity , Pigment Epithelium of Eye/cytology , Cell Survival/drug effects , Cells, Cultured/drug effects , Cytoprotection , Humans , Pigment Epithelium of Eye/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL