Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Publication year range
1.
Hum Mol Genet ; 25(6): 1165-75, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26740549

ABSTRACT

Most inherited blinding diseases are characterized by compromised retinal function and progressive degeneration of photoreceptors. However, the factors that affect the life span of photoreceptors in such degenerative retinal diseases are rather poorly understood. Here, we explore the role of hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1) in this context. HCN1 is known to adjust retinal function under mesopic conditions, and although it is expressed at high levels in rod and cone photoreceptor inner segments, no association with any retinal disorder has yet been found. We investigated the effects of an additional genetic deletion of HCN1 on the function and survival of photoreceptors in a mouse model of CNGB1-linked retinitis pigmentosa (RP). We found that the absence of HCN1 in Cngb1 knockout (KO) mice exacerbated photoreceptor degeneration. The deleterious effect was reduced by expression of HCN1 using a viral vector. Moreover, pharmacological inhibition of HCN1 also enhanced rod degeneration in Cngb1 KO mice. Patch-clamp recordings revealed that the membrane potentials of Cngb1 KO and Cngb1/Hcn1 double-KO rods were both significantly depolarized. We also found evidence for altered calcium homeostasis and increased activation of the protease calpain in Cngb1/Hcn1 double-KO mice. Finally, the deletion of HCN1 also exacerbated degeneration of cone photoreceptors in a mouse model of CNGA3-linked achromatopsia. Our results identify HCN1 as a major modifier of photoreceptor degeneration and suggest that pharmacological inhibition of HCN channels may enhance disease progression in RP and achromatopsia patients.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Retinitis Pigmentosa/pathology , Animals , Color Vision Defects/genetics , Color Vision Defects/metabolism , Color Vision Defects/pathology , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Disease Models, Animal , Disease Progression , Membrane Potentials/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retina/metabolism , Retina/pathology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Vision, Ocular
2.
Hum Mol Genet ; 24(19): 5486-99, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26188004

ABSTRACT

Mutations in the PDE6A gene can cause rod photoreceptors degeneration and the blinding disease retinitis pigmentosa (RP). While a number of pathogenic PDE6A mutations have been described, little is known about their impact on compound heterozygous situations and potential interactions of different disease-causing alleles. Here, we used a novel mouse model for the Pde6a R562W mutation in combination with an existing line carrying the V685M mutation to generate compound heterozygous Pde6a V685M/R562W animals, exactly homologous to a case of human RP. We compared the progression of photoreceptor degeneration in these compound heterozygous mice with the homozygous V685M and R562W mutants, and additionally with the D670G line that is known for a relatively mild phenotype. We investigated PDE6A expression, cyclic guanosine mono-phosphate accumulation, calpain and caspase activity, in vivo retinal function and morphology, as well as photoreceptor cell death and survival. This analysis confirms the severity of different Pde6a mutations and indicates that compound heterozygous mutants behave like intermediates of the respective homozygous situations. Specifically, the severity of the four different Pde6a situations may be categorized by the pace of photoreceptor degeneration: V685M (fastest) > V685M/R562W > R562W > D670G (slowest). While calpain activity was strongly increased in all four mutants, caspase activity was not. This points to the execution of non-apoptotic cell death and may lead to the identification of new targets for therapeutic interventions. For individual RP patients, our study may help to predict time-courses for Pde6a-related retinal degeneration and thereby facilitate the definition of a window-of-opportunity for clinical interventions.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Point Mutation , Retina/physiopathology , Retinitis Pigmentosa/pathology , Animals , Calpain/metabolism , Caspases/metabolism , Cell Survival , Disease Models, Animal , Humans , Mice , Retina/metabolism , Retina/pathology , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/physiopathology
3.
Hum Mol Genet ; 23(14): 3759-71, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24565864

ABSTRACT

Mutations in the CRB1 gene lead to retinal dystrophies ranging from Leber congenital amaurosis (LCA) to early-onset retinitis pigmentosa (RP), due to developmental defects or loss of adhesion between photoreceptors and Müller glia cells, respectively. Whereas over 150 mutations have been found, no clear genotype-phenotype correlation has been established. Mouse Crb1 knockout retinas show a mild phenotype limited to the inferior quadrant, whereas Crb2 knockout retinas display a severe degeneration throughout the retina mimicking the phenotype observed in RP patients associated with CRB1 mutations. Crb1Crb2 double mutant retinas have severe developmental defects similar to the phenotype observed in LCA patients associated with CRB1 mutations. Therefore, CRB2 is a candidate modifying gene of human CRB1-related retinal dystrophy. In this study, we studied the cellular localization of CRB1 and CRB2 in human retina and tested the influence of the Crb2 gene allele on Crb1-retinal dystrophies in mice. We found that in contrast to mice, in the human retina CRB1 protein was expressed at the subapical region in photoreceptors and Müller glia cells, and CRB2 only in Müller glia cells. Genetic ablation of one allele of Crb2 in heterozygote Crb1(+/-) retinas induced a mild retinal phenotype, but in homozygote Crb1 knockout mice lead to an early and severe phenotype limited to the entire inferior retina. Our data provide mechanistic insight for CRB1-related LCA and RP.


Subject(s)
Carrier Proteins/metabolism , Ependymoglial Cells/metabolism , Eye Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Retinal Dystrophies/metabolism , Adult , Aged , Aged, 80 and over , Animals , Carrier Proteins/genetics , Disease Models, Animal , Eye Proteins/genetics , Gene Knockout Techniques , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Tissue Proteins/genetics , Photoreceptor Cells/metabolism
4.
Hum Mol Genet ; 23(13): 3384-401, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24493795

ABSTRACT

In humans, the Crumbs homolog-1 (CRB1) gene is mutated in autosomal recessive Leber congenital amaurosis and early-onset retinitis pigmentosa. In mammals, the Crumbs family is composed of: CRB1, CRB2, CRB3A and CRB3B. Recently, we showed that removal of mouse Crb2 from retinal progenitor cells, and consequent removal from Müller glial and photoreceptor cells, results in severe and progressive retinal degeneration with concomitant loss of retinal function that mimics retinitis pigmentosa due to mutations in the CRB1 gene. Here, we studied the effects of cell-type-specific loss of CRB2 from the developing mouse retina using targeted conditional deletion of Crb2 in photoreceptors or Müller cells. We analyzed the consequences of targeted loss of CRB2 in the adult mouse retina using adeno-associated viral vectors encoding Cre recombinase and short hairpin RNA against Crb2. In vivo retinal imaging by means of optical coherence tomography on retinas lacking CRB2 in photoreceptors showed progressive thinning of the photoreceptor layer and cellular mislocalization. Electroretinogram recordings under scotopic conditions showed severe attenuation of the a-wave, confirming the degeneration of photoreceptors. Retinas lacking CRB2 in developing photoreceptors showed early onset of abnormal lamination, whereas retinas lacking CRB2 in developing Müller cells showed late onset retinal disorganization. Our data suggest that in the developing retina, CRB2 has redundant functions in Müller glial cells, while CRB2 has essential functions in photoreceptors. Our data suggest that short-term loss of CRB2 in adult mouse photoreceptors, but not in Müller glial cells, causes sporadic loss of adhesion between photoreceptors and Müller cells.


Subject(s)
Membrane Proteins/metabolism , Photoreceptor Cells/metabolism , Retinitis Pigmentosa/etiology , Retinitis Pigmentosa/metabolism , Animals , Ependymoglial Cells/metabolism , Female , Immunohistochemistry , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Retinitis Pigmentosa/genetics
5.
Hum Mol Genet ; 23(6): 1538-50, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24163243

ABSTRACT

Mutations in CACNA1F encoding the α1-subunit of the retinal Cav1.4 L-type calcium channel have been linked to Cav1.4 channelopathies including incomplete congenital stationary night blindness type 2A (CSNB2), Åland Island eye disease (AIED) and cone-rod dystrophy type 3 (CORDX3). Since CACNA1F is located on the X chromosome, Cav1.4 channelopathies are typically affecting male patients via X-chromosomal recessive inheritance. Occasionally, clinical symptoms have been observed in female carriers, too. It is currently unknown how these mutations lead to symptoms in carriers and how the retinal network in these females is affected. To investigate these clinically important issues, we compared retinal phenotypes in Cav1.4-deficient and Cav1.4 heterozygous mice and in human female carrier patients. Heterozygous Cacna1f carrier mice have a retinal mosaic consistent with differential X-chromosomal inactivation, characterized by adjacent vertical columns of affected and non-affected wild-type-like retinal network. Vertical columns in heterozygous mice are well comparable to either the wild-type retinal network of normal mice or to the retina of homozygous mice. Affected retinal columns display pronounced rod and cone photoreceptor synaptopathy and cone degeneration. These changes lead to vastly impaired vision-guided navigation under dark and normal light conditions and reduced retinal electroretinography (ERG) responses in Cacna1f carrier mice. Similar abnormal ERG responses were found in five human CACNA1F carriers, four of which had novel mutations. In conclusion, our data on Cav1.4 deficient mice and human female carriers of mutations in CACNA1F are consistent with a phenotype of mosaic CSNB2.


Subject(s)
Calcium Channels/genetics , Eye Diseases, Hereditary/pathology , Genetic Diseases, X-Linked/pathology , Myopia/pathology , Night Blindness/pathology , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Animals , Calcium Channels/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Disease Models, Animal , Electroretinography , Eye Diseases, Hereditary/genetics , Female , Genetic Diseases, X-Linked/genetics , Heterozygote , Humans , Male , Mice , Mice, Knockout , Mutation, Missense , Myopia/genetics , Night Blindness/genetics , Phenotype , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , X Chromosome , X Chromosome Inactivation
6.
PLoS Genet ; 9(12): e1003976, 2013.
Article in English | MEDLINE | ID: mdl-24339791

ABSTRACT

Development in the central nervous system is highly dependent on the regulation of the switch from progenitor cell proliferation to differentiation, but the molecular and cellular events controlling this process remain poorly understood. Here, we report that ablation of Crb1 and Crb2 genes results in severe impairment of retinal function, abnormal lamination and thickening of the retina mimicking human Leber congenital amaurosis due to loss of CRB1 function. We show that the levels of CRB1 and CRB2 proteins are crucial for mouse retinal development, as they restrain the proliferation of retinal progenitor cells. The lack of these apical proteins results in altered cell cycle progression and increased number of mitotic cells leading to an increased number of late-born cell types such as rod photoreceptors, bipolar and Müller glia cells in postmitotic retinas. Loss of CRB1 and CRB2 in the retina results in dysregulation of target genes for the Notch1 and YAP/Hippo signaling pathways and increased levels of P120-catenin. Loss of CRB1 and CRB2 result in altered progenitor cell cycle distribution with a decrease in number of late progenitors in G1 and an increase in S and G2/M phase. These findings suggest that CRB1 and CRB2 suppress late progenitor pool expansion by regulating multiple proliferative signaling pathways.


Subject(s)
Central Nervous System/metabolism , Leber Congenital Amaurosis/genetics , Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Retina/growth & development , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Proliferation , Central Nervous System/growth & development , Central Nervous System/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Humans , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mitosis/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Retina/cytology , Retina/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Stem Cells/metabolism
7.
Hum Mol Genet ; 22(1): 35-50, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23001562

ABSTRACT

In humans, the Crumbs homolog-1 (CRB1) gene is mutated in progressive types of autosomal recessive retinitis pigmentosa and Leber congenital amaurosis. However, there is no clear genotype-phenotype correlation for CRB1 mutations, which suggests that other components of the CRB complex may influence the severity of retinal disease. Therefore, to understand the physiological role of the Crumbs complex proteins, we generated and analysed conditional knockout mice lacking CRB2 in the developing retina. Progressive disorganization was detected during late retinal development. Progressive thinning of the photoreceptor layer and sites of cellular mislocalization was detected throughout the CRB2-deficient retina by confocal scanning laser ophthalmoscopy and spectral domain optical coherence tomography. Under scotopic conditions using electroretinography, the attenuation of the a-wave was relatively stronger than that of the b-wave, suggesting progressive degeneration of photoreceptors in adult animals. Histological analysis of newborn mice showed abnormal lamination of immature rod photoreceptors and disruption of adherens junctions between photoreceptors, Müller glia and progenitor cells. The number of late-born progenitor cells, rod photoreceptors and Müller glia cells was increased, concomitant with programmed cell death of rod photoreceptors. The data suggest an essential role for CRB2 in proper lamination of the photoreceptor layer and suppression of proliferation of late-born retinal progenitor cells.


Subject(s)
Eye Proteins/genetics , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Retina/metabolism , Retinitis Pigmentosa/genetics , Animals , Apoptosis , Base Sequence , DNA Primers , Electroretinography , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells, Vertebrate/pathology , Polymerase Chain Reaction , Retina/growth & development , Retinitis Pigmentosa/pathology , Tomography, Optical Coherence
8.
Hum Mol Genet ; 21(20): 4486-96, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22802073

ABSTRACT

Retinitis pigmentosa (RP) is a group of genetically heterogeneous, severe retinal diseases commonly leading to legal blindness. Mutations in the CNGB1a subunit of the rod cyclic nucleotide-gated (CNG) channel have been found to cause RP in patients. Here, we demonstrate the efficacy of gene therapy as a potential treatment for RP by means of recombinant adeno-associated viral (AAV) vectors in the CNGB1 knockout (CNGB1(-/-)) mouse model. To enable efficient packaging and rod-specific expression of the relatively large CNGB1a cDNA (~4 kb), we used an AAV expression cassette with a short rod-specific promoter and short regulatory elements. After injection of therapeutic AAVs into the subretinal space of 2-week-old CNGB1(-/-) mice, we assessed the restoration of the visual system by analyzing (i) CNG channel expression and localization, (ii) retinal function and morphology and (iii) vision-guided behavior. We found that the treatment not only led to expression of full-length CNGB1a, but also restored normal levels of the previously degraded CNGA1 subunit of the rod CNG channel. Both proteins co-localized in rod outer segments and formed regular CNG channel complexes within the treated area of the CNGB1(-/-) retina, leading to significant morphological preservation and a delay of retinal degeneration. In the electroretinographic analysis, we also observed restoration of rod-driven light responses. Finally, treated CNGB1(-/-) mice performed significantly better than untreated mice in a rod-dependent vision-guided behavior test. In summary, this work provides a proof-of-concept for the treatment of rod channelopathy-associated RP by AAV-mediated gene replacement.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Nerve Tissue Proteins/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Animals , Cyclic Nucleotide-Gated Cation Channels/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Electroretinography , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Retinitis Pigmentosa/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Rod Cell Outer Segment/metabolism
9.
Adv Exp Med Biol ; 801: 733-9, 2014.
Article in English | MEDLINE | ID: mdl-24664765

ABSTRACT

Retinitis pigmentosa (RP) is a severe retinal disease characterized by a progressive degeneration of rod photoreceptors and a secondary loss of cone function. Here, we used CNGB1-deficient (CNGB1(-/-)) mice, a mouse model for autosomal recessive RP, to evaluate the efficacy of adeno-associated virus (AAV) vector-mediated gene therapy for the treatment of RP. The treatment restored normal expression of rod CNG channels and rod-driven light responses in the CNGB1(-/-) retina. This led to a substantial delay of retinal degeneration and long-term preservation of retinal morphology. Finally, treated CNGB1(-/-) mice performed significantly better than untreated mice in a rod-dependent vision-guided behavior test. In summary, this study holds promise for the treatment of rod channelopathy-associated retinitis pigmentosa by AAV-mediated gene replacement.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Dependovirus/genetics , Nerve Tissue Proteins/genetics , Recovery of Function/genetics , Retinal Degeneration/therapy , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/therapy , Animals , Disease Models, Animal , Electroretinography , Maze Learning , Mice , Mice, Knockout , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Vision, Ocular/physiology
10.
Glia ; 61(10): 1629-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893895

ABSTRACT

MPP3 and CRB1 both interact directly with PALS1/MPP5 and through this scaffold protein may form a large protein complex. To investigate the role of MPP3 in the retina we have analyzed conditional mutant Mpp3 knockout mice. Ultrastructural localization studies revealed that MPP3 is predominantly localized in apical villi of Müller glia cells. Retinas lacking MPP3 developed late onset retinal degeneration, with sporadic foci of rosette formation in the central part of the retina. Retinal degeneration in Mpp3 cKO mice was accelerated by exposure to moderate levels of white light. Electroretinography recordings in aging mice under both scotopic and photopic conditions ranged from normal to mildly subnormal, while the magnitude correlated with the strength and extent of morphological alterations. Loss of MPP3 resulted in significant loss of PALS1 at the subapical region adjacent to adherens junctions, and loss of MPP3 in Pals1 conditional knockdown retinas significantly accelerated the onset of retinal degeneration. These data suggest that MPP3 is required for maintaining proper levels of PALS1 at the subapical region, and indicate that the MPP3 gene is a candidate modulator of the Crumbs complex.


Subject(s)
Cell Adhesion/physiology , Ependymoglial Cells/metabolism , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Nucleoside-Phosphate Kinase/metabolism , Photoreceptor Cells/metabolism , Animals , Catenins/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules/metabolism , Electroretinography , Ependymoglial Cells/ultrastructure , Fluorescein Angiography , Gene Expression Regulation/genetics , Guanylate Kinases/deficiency , Light/adverse effects , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Nectins , Photoreceptor Cells/ultrastructure , Retina/cytology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Tomography, Optical Coherence , Visual Pathways/metabolism , Delta Catenin
11.
Eur J Neurosci ; 37(7): 1048-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23351077

ABSTRACT

Many aspects of retinal physiology are modulated by circadian clocks, but it is unclear whether clock malfunction impinges directly on photoreceptor survival, differentiation or function. Eyes from wild-type (WT) and Period1 (Per1) and Period2 (Per2) mutant mice (Per1(Brdm1) Per2(Brdm1) ) were examined for structural (histology, in vivo imaging), phenotypical (RNA expression, immunohistochemistry) and functional characteristics. Transcriptional levels of selected cone genes [red/green opsin (Opn1mw), blue cone opsin (Opn1sw) and cone arrestin (Arr3)] and one circadian clock gene (RORb) were quantified by real-time polymerase chain reaction. Although there were no changes in general retinal histology or visual responses (electroretinograms) between WT and Per1(Brdm1) Per2(Brdm1) mice, compared with age-matched controls, Per1(Brdm1) Per2(Brdm1) mice showed scattered retinal deformations by fundus inspection. Also, mRNA expression levels and immunostaining of blue cone opsin were significantly reduced in mutant mice. Especially, there was an alteration in the dorsal-ventral patterning of blue cones. Decreased blue cone opsin immunoreactivity was present by early postnatal stages, and remained throughout maturation. General photoreceptor differentiation was retarded in young mutant mice. In conclusion, deletion of both Per1 and Per2 clock genes leads to multiple discrete changes in retina, notably patchy tissue disorganization, reductions in cone opsin mRNA and protein levels, and altered distribution. These data represent the first direct link between Per1 and Per2 clock genes, and cone photoreceptor differentiation and function.


Subject(s)
Period Circadian Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Rod Opsins/metabolism , Animals , Arrestins/genetics , Arrestins/metabolism , Cell Differentiation , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nuclear Receptor Subfamily 1, Group F, Member 2/genetics , Nuclear Receptor Subfamily 1, Group F, Member 2/metabolism , Period Circadian Proteins/metabolism , Retina/cytology , Retina/growth & development , Retina/physiology , Retinal Cone Photoreceptor Cells/cytology , Rod Opsins/genetics , Transcription, Genetic
12.
Doc Ophthalmol ; 127(1): 3-11, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23748796

ABSTRACT

PURPOSE: To study the relationship between retinal and tunica vasculosa lentis (TVL) disease in retinopathy of prematurity (ROP). Although the clinical hallmark of ROP is abnormal retinal blood vessels, the vessels of the anterior segment, including the TVL, are also altered. METHODS: ROP was induced in Long-Evans pigmented and Sprague Dawley albino rats; room-air-reared (RAR) rats served as controls. Then, fluorescein angiographic images of the TVL and retinal vessels were serially obtained with a scanning laser ophthalmoscope near the height of retinal vascular disease, ~20 days of age, and again at 30 and 64 days of age. Additionally, electroretinograms (ERGs) were obtained prior to the first imaging session. The TVL images were analyzed for percent coverage of the posterior lens. The tortuosity of the retinal arterioles was determined using Retinal Image multiScale Analysis (Gelman et al. in Invest Ophthalmol Vis Sci 46:4734-4738, 2005). RESULTS: In the youngest ROP rats, the TVL was dense, while in RAR rats, it was relatively sparse. By 30 days, the TVL in RAR rats had almost fully regressed, while in ROP rats, it was still pronounced. By the final test age, the TVL had completely regressed in both ROP and RAR rats. In parallel, the tortuous retinal arterioles in ROP rats resolved with increasing age. ERG components indicating postreceptoral dysfunction, the b-wave, and oscillatory potentials were attenuated in ROP rats. CONCLUSIONS: These findings underscore the retinal vascular abnormalities and, for the first time, show abnormal anterior segment vasculature in the rat model of ROP. There is delayed regression of the TVL in the rat model of ROP. This demonstrates that ROP is a disease of the whole eye.


Subject(s)
Disease Models, Animal , Lens, Crystalline/blood supply , Persistent Hyperplastic Primary Vitreous/physiopathology , Retina/physiopathology , Retinal Artery/pathology , Retinal Neovascularization/pathology , Retinopathy of Prematurity/physiopathology , Animals , Animals, Newborn , Arterioles/pathology , Electroretinography , Fluorescein Angiography , Humans , Infant, Newborn , Male , Oxygen/toxicity , Persistent Hyperplastic Primary Vitreous/diagnosis , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Retinopathy of Prematurity/diagnosis
13.
JAMA Ophthalmol ; 138(6): 643-651, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32352493

ABSTRACT

Importance: Achromatopsia linked to variations in the CNGA3 gene is associated with day blindness, poor visual acuity, photophobia, and involuntary eye movements owing to lack of cone photoreceptor function. No treatment is currently available. Objective: To assess safety and vision outcomes of supplemental gene therapy with adeno-associated virus (AAV) encoding CNGA3 (AAV8.CNGA3) in patients with CNGA3-linked achromatopsia. Design, Setting, and Participants: This open-label, exploratory nonrandomized controlled trial tested safety and vision outcomes of gene therapy vector AAV8.CNGA3 administered by subretinal injection at a single center. Nine patients (3 per dose group) with a clinical diagnosis of achromatopsia and confirmed biallelic disease-linked variants in CNGA3 were enrolled between November 5, 2015, and September 22, 2016. Data analysis was performed from June 6, 2017, to March 12, 2018. Intervention: Patients received a single unilateral injection of 1.0 × 1010, 5.0 × 1010, or 1.0 × 1011 total vector genomes of AAV8.CNGA3 and were followed up for a period of 12 months (November 11, 2015, to October 10, 2017). Main Outcomes and Measures: Safety as the primary end point was assessed by clinical examination of ocular inflammation. Systemic safety was assessed by vital signs, routine clinical chemistry testing, and full and differential blood cell counts. Secondary outcomes were change in visual function from baseline in terms of spatial and temporal resolution and chromatic, luminance, and contrast sensitivity throughout a period of 12 months after treatment. Results: Nine patients (mean [SD] age, 39.6 [11.9] years; age range, 24-59 years; 8 [89%] male) were included in the study. Baseline visual acuity letter score (approximate Snellen equivalent) ranged from 34 (20/200) to 49 (20/100), whereas baseline contrast sensitivity log scores ranged from 0.1 to 0.9. All 9 patients underwent surgery and subretinal injection of AAV8.CNGA3 without complications. No substantial safety problems were observed during the 12-month follow-up period. Despite the congenital deprivation of cone photoreceptor-mediated vision in achromatopsia, all 9 treated eyes demonstrated some level of improvement in secondary end points regarding cone function, including mean change in visual acuity of 2.9 letters (95% CI, 1.65-4.13; P = .006, 2-sided t test paired samples). Contrast sensitivity improved by a mean of 0.33 log (95% CI, 0.14-0.51 log; P = .003, 2-sided t test paired samples). Conclusions and Relevance: Subretinal gene therapy with AAV8.CNGA3 was not associated with substantial safety problems and was associated with cone photoreceptor activation in adult patients, as reflected by visual acuity and contrast sensitivity gains. Trial Registration: ClinicalTrials.gov Identifier: NCT02610582.


Subject(s)
Color Vision Defects/therapy , Cyclic Nucleotide-Gated Cation Channels/genetics , Genetic Therapy/methods , Retinal Cone Photoreceptor Cells/pathology , Visual Acuity , Adult , Color Vision Defects/diagnosis , Color Vision Defects/physiopathology , Electroretinography , Female , Follow-Up Studies , Humans , Male , Middle Aged , Retina , Retrospective Studies , Treatment Outcome , Young Adult
14.
Sci Rep ; 9(1): 3470, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837488

ABSTRACT

Malaria is a causative factor in about 500.000 deaths each year world-wide. Cerebral malaria is a particularly severe complication of this disease and thus associated with an exceedingly high mortality. Malaria retinopathy is an ocular manifestation often associated with cerebral malaria, and presumably shares a substantial part of its pathophysiology. Here, we describe that indeed murine malaria retinopathy reproduced the main hallmarks of the corresponding human disease. In the living animal, we were able to follow the circulation and cellular localization of malaria parasites transgenically labelled with GFP via non-invasive in vivo retinal imaging. We found that malaria parasites cross the blood-retinal-barrier and infiltrate the neuroretina, concomitant with an extensive, irreversible, and long-lasting retinal neurodegeneration. Furthermore, anti-malarial treatment with dihydroartemisinin strongly diminished the load of circulating parasites but resolved the symptoms of the retinopathy only in part. In summary, we introduce here a novel preclinical model for human cerebral malaria that is much more directly accessible for studies into disease pathophysiology and development of novel treatment approaches. In vivo retinal imaging may furthermore serve as a valuable tool for the early diagnosis of the human disease.


Subject(s)
Malaria, Cerebral/diagnosis , Malaria, Cerebral/parasitology , Retina/pathology , Animals , Biomarkers , Disease Models, Animal , Electroretinography/methods , Gene Expression , Genes, Reporter , Malaria, Cerebral/metabolism , Mice , Mice, Transgenic , Ophthalmoscopy , Phenotype , Plasmodium berghei , Retina/diagnostic imaging , Retina/metabolism , Tomography, Optical Coherence
15.
Sci Rep ; 8(1): 5970, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29654250

ABSTRACT

Mutations in the Norrin (NDP) gene cause severe developmental blood vessel defects in the retina leading to congenital blindness. In the retina of Ndph-knockout mice only the superficial capillary network develops. Here, a detailed characterization of this mouse model at late stages of the disease using in vivo retinal imaging revealed cystoid structures that closely resemble the ovoid cysts in the inner nuclear layer of the human retina with cystoid macular edema (CME). In human CME an involvement of Müller glia cells is hypothesized. In Ndph-knockout retinae we could demonstrate that activated Müller cells were located around and within these cystoid spaces. In addition, we observed extensive activation of retinal microglia and development of neovascularization. Furthermore, ex vivo analyses detected extravasation of monocytic cells suggesting a breakdown of the blood retina barrier. Thus, we could demonstrate that also in the developmental retinal vascular pathology present in the Ndph-knockout mouse inflammatory processes are active and may contribute to further retinal degeneration. This observation delivers a new perspective for curative treatments of retinal vasculopathies. Modulation of inflammatory responses might reduce the symptoms and improve visual acuity in these diseases.


Subject(s)
Eye Proteins/metabolism , Inflammation/pathology , Macular Edema/pathology , Neovascularization, Pathologic/pathology , Nerve Tissue Proteins/metabolism , Retina/pathology , Animals , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/pathology , Disease Models, Animal , Humans , Inflammation/metabolism , Macular Edema/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Visual Acuity/physiology
16.
Hum Gene Ther ; 28(12): 1180-1188, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29212391

ABSTRACT

Retinitis pigmentosa type 43 (RP43) is a blinding disease caused by mutations in the gene for rod phosphodiesterase 6 alpha (PDE6A). The disease process begins with a dysfunction of rod photoreceptors, subsequently followed by a currently untreatable progressive degeneration of the entire outer retina. Aiming at a curative approach via PDE6A gene supplementation, a novel adeno-associated viral (AAV) vector was developed for expression of the human PDE6A cDNA under control of the human rhodopsin promotor (rAAV8.PDE6A). This study assessed the therapeutic efficacy of rAAV8.PDE6A in the Pde6anmf363/nmf363-mutant mouse model of RP43. All mice included in this study were treated with sub-retinal injections of the vector at 2 weeks after birth. The therapeutic effect was monitored at 1 month and 6 months post injection. Biological function of the transgene was assessed in vivo by means of electroretinography. The degree of morphological rescue was investigated both in vivo using optical coherence tomography and ex vivo by immunohistological staining. It was found that the novel rAAV8.PDE6A vector resulted in a stable and efficient expression of PDE6A protein in rod photoreceptors of Pde6anmf363/nmf363 mice following treatment at both the short- and long-term time points. The treatment led to a substantial morphological preservation of outer nuclear layer thickness, rod outer segment structure, and prolonged survival of cone photoreceptors for at least 6 months. Additionally, the ERG analysis confirmed a restoration of retinal function in a group of treated mice. Taken together, this study provides successful proof-of-concept for the cross-species efficacy of the rAAV8.PDE6A vector developed for use in human patients. Importantly, the data show stable expression and rescue effects for a prolonged period of time, raising hope for future translational studies based on this approach.

17.
Front Neurosci ; 11: 292, 2017.
Article in English | MEDLINE | ID: mdl-28596720

ABSTRACT

Achromatopsia type 2 (ACHM2) is a severe, inherited eye disease caused by mutations in the CNGA3 gene encoding the α subunit of the cone photoreceptor cyclic nucleotide-gated (CNG) channel. Patients suffer from strongly impaired daylight vision, photophobia, nystagmus, and lack of color discrimination. We have previously shown in the Cnga3 knockout (KO) mouse model of ACHM2 that gene supplementation therapy is effective in rescuing cone function and morphology and delaying cone degeneration. In our preclinical approach, we use recombinant adeno-associated virus (AAV) vector-mediated gene transfer to express the murine Cnga3 gene under control of the mouse blue opsin promoter. Here, we provide novel data on the efficiency and permanence of such gene supplementation therapy in Cnga3 KO mice. Specifically, we compare the influence of two different AAV vector capsids, AAV2/5 (Y719F) and AAV2/8 (Y733F), on restoration of cone function, and assess the effect of age at time of treatment on the long-term outcome. The evaluation included in vivo analysis of retinal function using electroretinography (ERG) and immunohistochemical analysis of vector-driven Cnga3 transgene expression. We found that both vector capsid serotypes led to a comparable rescue of cone function over the observation period between 4 weeks and 3 months post treatment. In addition, a clear therapeutic effect was present in mice treated at 2 weeks of age as well as in mice treated at 3 months of age at the first assessment at 4 weeks after treatment. Importantly, the effect extended in both cases over the entire observation period of 12 months post treatment. However, the average ERG amplitude levels differed between the two groups, suggesting a role of the absolute age, or possibly, the associated state of the degeneration, on the achievable outcome. In summary, we found that the therapeutic time window of opportunity for AAV-mediated Cnga3 gene supplementation therapy in the Cnga3 KO mouse model extends at least to an age of 3 months, but is presumably limited by the condition, number and topographical distribution of remaining cones at the time of treatment. No impact of the choice of capsid on the therapeutic success was detected.

18.
PLoS One ; 12(6): e0178753, 2017.
Article in English | MEDLINE | ID: mdl-28575130

ABSTRACT

Loss of Norrin signalling due to mutations in the Norrie disease pseudoglioma gene causes severe vascular defects in the retina, leading to visual impairment and ultimately blindness. While the emphasis of experimental work so far was on the developmental period, we focus here on disease mechanisms that induce progression into severe adult disease. The goal of this study was the comprehensive analysis of the long-term effects of the absence of Norrin on vascular homeostasis and retinal function. In a mouse model of Norrie disease retinal vascular morphology and integrity were studied by means of in vivo angiography; the vascular constituents were assessed in detailed histological analyses using quantitative retinal morphometry. Finally, electroretinographic analyses were performed to assess the retinal function in adult Norrin deficient animals. We could show that the primary developmental defects not only persisted but developed into further vascular abnormalities and microangiopathies. In particular, the overall vessel homeostasis, the vascular integrity, and also the cellular constituents of the vascular wall were affected in the adult Norrin deficient retina. Moreover, functional analyses indicated to persistent hypoxia in the neural retina which was suggested as one of the major driving forces of disease progression. In summary, our data provide evidence that the key to adult Norrie disease are ongoing vascular modifications, driven by the persistent hypoxic conditions, which are ineffective to compensate for the primary Norrin-dependent defects.


Subject(s)
Blindness/congenital , Genetic Diseases, X-Linked/pathology , Nerve Tissue Proteins/deficiency , Nervous System Diseases/pathology , Retinal Vessels/pathology , Spasms, Infantile/pathology , Angiography , Animals , Blindness/diagnostic imaging , Blindness/genetics , Blindness/pathology , Capillaries/pathology , Cell Hypoxia , Disease Models, Animal , Disease Progression , Electroretinography , Eye Proteins/genetics , Eye Proteins/physiology , Genetic Diseases, X-Linked/diagnostic imaging , Genetic Diseases, X-Linked/genetics , Lasers , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Nervous System Diseases/diagnostic imaging , Nervous System Diseases/genetics , Ophthalmoscopy/methods , Retinal Degeneration , Retinal Vessels/diagnostic imaging , Spasms, Infantile/diagnostic imaging , Spasms, Infantile/genetics
19.
PLoS One ; 11(1): e0147728, 2016.
Article in English | MEDLINE | ID: mdl-26807953

ABSTRACT

PURPOSE: Vision originates in rods and cones at the outer retina. Already at these early stages, diverse processing schemes shape and enhance image information to permit perception over a wide range of lighting conditions. In this work, we address the role of hyperpolarization-activated and cyclic nucleotide-gated channels 1 (HCN1) in rod photoreceptors for the enhancement of rod system responsivity under conditions of light exposure. METHODS: To isolate HCN1 channel actions in rod system responses, we generated double mutant mice by crossbreeding Hcn1-/- mice with Cnga3-/- mice in which cones are non-functional. Retinal function in the resulting Hcn1-/- Cnga3-/- animals was followed by means of electroretinography (ERG) up to the age of four month. Retinal imaging via scanning laser ophthalmoscopy (SLO) and optical coherence tomography (OCT) was also performed to exclude potential morphological alterations. RESULTS: This study on Hcn1-/- Cnga3-/- mutant mice complements our previous work on HCN1 channel function in the retina. We show here in a functional rod-only setting that rod responses following bright light exposure terminate without the counteraction of HCN channels much later than normal. The resulting sustained signal elevation does saturate the retinal network due to an intensity-dependent reduction in the dynamic range. In addition, the lack of rapid adaptational feedback modulation of rod photoreceptor output via HCN1 in this double mutant limits the ability to follow repetitive (flicker) stimuli, particularly under mesopic conditions. CONCLUSIONS: This work corroborates the hypothesis that, in the absence of HCN1-mediated feedback, the amplitude of rod signals remains at high levels for a prolonged period of time, leading to saturation of the retinal pathways. Our results demonstrate the importance of HCN1 channels for regular vision.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Retina/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Vision, Ocular/physiology , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Electroretinography , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Mice , Mice, Knockout , Photic Stimulation , Retina/cytology , Retinal Cone Photoreceptor Cells/metabolism , Signal Transduction/physiology , Tomography, Optical Coherence
20.
PLoS One ; 10(6): e0131154, 2015.
Article in English | MEDLINE | ID: mdl-26110792

ABSTRACT

PURPOSE: To address the problem of unequal scales for the measurement of two-dimensional structures in OCT images, and demonstrate the use of intra¬ocular objects of known dimensions in the murine eye for the equal calibration of axes. METHODS: The first part of this work describes the mathematical foundation of major distortion effects introduced by X-Y scaling differences. Illustrations were generated with CorelGraph X3 software. The second part bases on image data obtained with a HRA2 Spectralis (Heidelberg Engineering) in SV129 wild-type mice. Subretinally and intravitreally implanted microbeads, alginate capsules with a diameter of 154±5 µm containing GFP-marked mesenchymal stem cells (CellBeads), were used as intraocular objects for calibration. RESULTS: The problems encountered with two-dimensional measurements in cases of unequal scales are demonstrated and an estimation of the resulting errors is provided. Commonly, the Y axis is reliably calibrated using outside standards like histology or manufacturer data. We show here that intraocular objects like dimensionally stable spherical alginate capsules allow for a two-dimensional calibration of the acquired OCT raw images by establishing a relation between X and Y axis data. For our setup, a correction factor of about 3.3 was determined using both epiretinally and subretinally positioned beads (3.350 ± 0.104 and 3.324 ± 0.083, respectively). CONCLUSIONS: In this work, we highlight the distortion-related problems in OCT image analysis induced by unequal X and Y scales. As an exemplary case, we provide data for a two-dimensional in vivo OCT image calibration in mice using intraocular alginate capsules. Our results demonstrate the need for a proper two-dimensional calibration of OCT data, and we believe that equal scaling will certainly improve the efficiency of OCT image analysis.


Subject(s)
Tomography, Optical Coherence/statistics & numerical data , Animals , Eye/anatomy & histology , Green Fluorescent Proteins , Image Processing, Computer-Assisted/statistics & numerical data , Mice , Mice, 129 Strain , Ophthalmoscopy/statistics & numerical data , Retina/anatomy & histology , Retinal Vessels/anatomy & histology
SELECTION OF CITATIONS
SEARCH DETAIL