Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Cells ; 10(3)2021 03 21.
Article in English | MEDLINE | ID: mdl-33800998

ABSTRACT

Vision loss is among the characteristic symptoms of neuronal ceroid lipofuscinosis (NCL), a fatal neurodegenerative lysosomal storage disorder. Here, we performed an in-depth analysis of retinal degeneration at the molecular and cellular levels in mice lacking the lysosomal aspartyl protease cathepsin D, an animal model of congenital CLN10 disease. We observed an early-onset accumulation of storage material as indicated by elevated levels of saposin D and subunit C of the mitochondrial ATP synthase. The accumulation of storage material was accompanied by reactive astrogliosis and microgliosis, elevated expression of the autophagy marker sequestosome 1/p62 and a dysregulated expression of several lysosomal proteins. The number of cone photoreceptor cells was reduced as early as at postnatal day 5. At the end stage of the disease, the outer nuclear layer was almost atrophied, and all cones were lost. A significant loss of rod and cone bipolar cells, amacrine cells and ganglion cells was found at advanced stages of the disease. Results demonstrate that cathepsin D deficiency results in an early-onset and rapidly progressing retinal dystrophy that involves all retinal cell types. Data of the present study will serve as a reference for studies aimed at developing treatments for retinal degeneration in CLN10 disease.


Subject(s)
Cathepsin D/deficiency , Neuronal Ceroid-Lipofuscinoses/pathology , Retina/pathology , Animals , Autophagy , Cathepsin D/metabolism , Disease Models, Animal , Gliosis/pathology , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proton-Translocating ATPases/metabolism , Neuronal Ceroid-Lipofuscinoses/complications , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Protein Subunits/metabolism , Retinal Degeneration/complications , Retinal Degeneration/pathology , Sequestosome-1 Protein/metabolism
3.
Ophthalmologe ; 118(2): 106-112, 2021 Feb.
Article in German | MEDLINE | ID: mdl-33030566

ABSTRACT

BACKGROUND: Neuronal ceroid lipofuscinosis (NCL) is a group of rare and fatal neurodegenerative lysosomal storage diseases. Progressive retinal degeneration and loss of vision are among the characteristic symptoms of affected patients. A brain-directed enzyme replacement therapy has been shown to significantly attenuate the neurological symptoms in CLN2 patients and is currently the only approved therapy for NCL; however, there is presently no treatment option for retinal dystrophy in NCL. OBJECTIVE: This short review aims to give an overview of preclinical studies that have developed and evaluated therapeutic strategies for the treatment of retinal dystrophy in animal models of different NCL forms. MATERIAL AND METHODS: The key findings of preclinical studies that have achieved positive therapeutic effects on retinal structure and/or function using different treatment strategies are summarized and discussed. RESULTS AND CONCLUSION: The published data on preclinical studies demonstrate the efficacy of different therapeutic strategies to attenuate retinal degeneration and vision loss in animal models for different NCL forms. It remains to be seen whether these promising results can be confirmed in future clinical studies.


Subject(s)
Neuronal Ceroid-Lipofuscinoses , Retinal Dystrophies , Animals , Disease Models, Animal , Enzyme Replacement Therapy , Humans , Neuronal Ceroid-Lipofuscinoses/complications , Neuronal Ceroid-Lipofuscinoses/diagnosis , Neuronal Ceroid-Lipofuscinoses/therapy , Retina , Retinal Dystrophies/diagnosis , Retinal Dystrophies/therapy , Tripeptidyl-Peptidase 1
4.
Exp Eye Res ; 176: 258-265, 2018 11.
Article in English | MEDLINE | ID: mdl-30237104

ABSTRACT

Neuroprotection is among the potential treatment options for glaucoma and other retinal pathologies characterized by the loss of retinal ganglion cells (RGCs). Here, we examined the impact of a neural stem (NS) cell-based intravitreal co-administration of two neuroprotective factors on the survival of axotomized RGCs. To this aim we used lentiviral vectors to establish clonal NS cell lines ectopically expressing either glial cell line-derived neurotrophic factor (GDNF) or ciliary neurotrophic factor (CNTF). The modified NS cell lines were intravitreally injected either separately or as a 1:1 mixture into adult mice one day after an optic nerve lesion, and the number of surviving RGCs was determined in retinal flat-mounts two, four and eight weeks after the lesion. For the transplantation experiments, we selected a GDNF- and a CNTF-expressing NS cell line that promoted the survival of axotomized RGCs with a similar efficacy. Eight weeks after the lesion, GDNF-treated retinas contained 3.8- and CNTF-treated retinas 3.7-fold more RGCs than control retinas. Of note, the number of surviving RGCs was markedly increased when both factors were administered simultaneously, with 14.3-fold more RGCs than in control retinas eight weeks after the lesion. GDNF and CNTF thus potently and synergistically rescued RGCs from axotomy-induced cell death, indicating that combinatorial neuroprotective approaches represent a promising strategy to effectively promote the survival of RGCs under pathological conditions.


Subject(s)
Ciliary Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neural Stem Cells/transplantation , Neuroprotective Agents/administration & dosage , Retinal Ganglion Cells/drug effects , Animals , Axotomy , Cell Survival/drug effects , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , Drug Synergism , Genetic Vectors , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Nerve Crush , Neural Stem Cells/metabolism , Neuroprotective Agents/metabolism , Optic Nerve Injuries
5.
Invest Ophthalmol Vis Sci ; 57(11): 4989-4998, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27654426

ABSTRACT

PURPOSE: Neuronal ceroid lipofuscinoses comprise a genetically heterogeneous group of mainly childhood-onset neurodegenerative lysosomal storage disorders. Progressive loss of vision is among the typical clinical symptoms of these fatal disorders. Here, we performed a detailed analysis of retinal degeneration in mice deficient in the lysosomal membrane protein CLN7, a novel animal model of CLN7 disease. METHODS: Immunohistochemical analyses of retinas at different ages were performed to qualitatively and quantitatively characterize retinal degeneration in CLN7-deficient mice. Storage material in mutant retinas was analyzed by electron microscopy, and expression levels of various lysosomal proteins were studied using immunohistochemistry, immunoblot analyses, and quantitative real-time PCR. RESULTS: We observed an early onset and rapidly progressing degeneration of photoreceptor cells in CLN7-deficient mice, resulting in the loss of more than 70% rod photoreceptors in 4-month-old animals. The number of cone photoreceptors was not detectably altered at this age. Loss of rod photoreceptors was accompanied by reactive astrogliosis and microgliosis. Immunohistochemical and immunoblot analyses revealed accumulation of subunit c of mitochondrial ATP synthase and saposin D in mutant retinas, and electron microscopic analyses demonstrated the presence of curvilinear bodies or fingerprint-like profiles in various cell types of CLN7-deficient retinas. We also found a marked dysregulation of various lysosomal proteins in mutant retinas. CONCLUSIONS: We conclude that the retina of CLN7-deficient mice represents a useful model to elucidate the pathomechanisms ultimately leading to neurodegeneration in CLN7 disease, and to evaluate the efficacy of strategies aimed at developing treatments for this fatal neurodegenerative lysosomal storage disorder.

6.
PLoS One ; 10(5): e0127204, 2015.
Article in English | MEDLINE | ID: mdl-25992714

ABSTRACT

A sustained intraocular administration of neurotrophic factors is among the strategies aimed at establishing treatments for currently untreatable degenerative retinal disorders. In the present study we have analyzed the neuroprotective effects of a continuous neural stem (NS) cell-based intraocular delivery of ciliary neurotrophic factor (CNTF) on photoreceptor cells in the nclf mouse, an animal model of the neurodegenerative lysosomal storage disorder variant late infantile neuronal ceroid lipofuscinosis (vLINCL). To this aim, we genetically modified adherently cultivated NS cells with a polycistronic lentiviral vector encoding a secretable variant of CNTF together with a Venus reporter gene (CNTF-NS cells). NS cells for control experiments (control-NS cells) were modified with a vector encoding the reporter gene tdTomato. Clonal CNTF-NS and control-NS cell lines were established using fluorescent activated cell sorting and intravitreally grafted into 14 days old nclf mice at the onset of retinal degeneration. The grafted cells preferentially differentiated into astrocytes that were attached to the posterior side of the lenses and the vitreal side of the retinas and stably expressed the transgenes for at least six weeks, the latest post-transplantation time point analyzed. Integration of donor cells into host retinas, ongoing proliferation of grafted cells or adverse effects of the donor cells on the morphology of the host eyes were not observed. Quantitative analyses of host retinas two, four and six weeks after cell transplantation revealed the presence of significantly more photoreceptor cells in eyes with grafted CNTF-NS cells than in eyes with grafted control-NS cells. This is the first demonstration that a continuous intraocular administration of a neurotrophic factor attenuates retinal degeneration in an animal model of neuronal ceroid lipofuscinosis.


Subject(s)
Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/therapeutic use , Genetic Therapy , Neural Stem Cells/transplantation , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/therapy , Photoreceptor Cells/pathology , Animals , Cell Count , Cell Line , Clone Cells , Disease Models, Animal , Gene Expression , Genes, Reporter , Genetic Vectors/metabolism , Immunoblotting , Injections, Intraocular , Intravitreal Injections , Lentivirus/genetics , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Photoreceptor Cells/metabolism , Retinal Degeneration/therapy , Stem Cell Transplantation
7.
Invest Ophthalmol Vis Sci ; 55(11): 7029-39, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25270193

ABSTRACT

PURPOSE: To analyze the neuroprotective effect of intravitreally grafted neural stem (NS) cells genetically modified to secrete ciliary neurotrophic factor (CNTF) on intraorbitally lesioned retinal ganglion cells (RGCs) in adult mice. METHODS: Adherently cultivated NS cells were genetically modified to express a secretable variant of mouse CNTF together with the fluorescent reporter protein Venus. Clonal CNTF-secreting NS cell lines were established using fluorescence activated cell sorting, and intravitreally grafted into adult mice 1 day after an intraorbital crush of the optic nerve. Brn-3a-positive RGCs were counted in flat-mounted retinas at different postlesion intervals to evaluate the neuroprotective effect of the CNTF-secreting NS cells on the axotomized RGCs. Anterograde axonal tracing experiments were performed to analyze the regrowth of the injured RGC axons in CNTF-treated retinas. RESULTS: Intravitreally grafted NS cells preferentially differentiated into astrocytes that survived in the host eyes, stably expressed CNTF, and significantly attenuated the loss of the axotomized RGCs over a period of at least 4 months, the latest postlesion time point analyzed. Depending on the postlesion interval analyzed, the number of RGCs in eyes with grafted CNTF-secreting NS cells was 2.8-fold to 6.4-fold higher than in eyes with grafted control NS cells. The CNTF-secreting NS cells additionally induced long-distance regrowth of the lesioned RGC axons. CONCLUSIONS: Genetically modified clonal NS cell lines may serve as a useful tool for preclinical studies aimed at evaluating the therapeutic potential of a sustained cell-based intravitreal administration of neuroprotective factors in mouse models of glaucoma.


Subject(s)
Ciliary Neurotrophic Factor/administration & dosage , Genetic Therapy/methods , Neural Stem Cells , Optic Nerve Diseases/therapy , Retinal Ganglion Cells/pathology , Stem Cell Transplantation , Animals , Cell Line , Disease Models, Animal , Immunohistochemistry , Injections , Mice , Mice, Inbred C57BL , Optic Nerve Diseases/pathology , Orbit , Retinal Ganglion Cells/drug effects
8.
Neurobiol Dis ; 65: 12-24, 2014 May.
Article in English | MEDLINE | ID: mdl-24423645

ABSTRACT

Mutations in the major facilitator superfamily domain containing 8 (MFSD8) gene coding for the lysosomal CLN7 membrane protein result in CLN7 disease, a lysosomal storage disease of childhood. CLN7 disease belongs to a group of inherited disorders, called neuronal ceroid lipofuscinoses (NCL), which are characterized by the accumulation of autofluorescent ceroid lipopigments, neuroinflammation, photoreceptor- and neurodegeneration. We have disrupted the Mfsd8 gene by insertion of a lacZ gene-trap cassette between exons 1 and 2 in mice and have analyzed the impact of Cln7 depletion on neuronal and visceral tissues. Analysis of lacZ reporter gene activity in heterozygous Mfsd8((wt/tm1a)) mice showed strong Mfsd8 mRNA expression in the cerebral cortex, in the hippocampus and in the kidney. Homozygous Mfsd8((tm1a/tm1a)) mice were viable and fertile and resembled biochemically the NCL-phenotype of human CLN7 patients including the accumulation of autofluorescent material in the brain and peripheral tissues and of subunit c of mitochondrial ATP synthase in the cerebellum and nuclei of distinct brain regions, and the degeneration of photoreceptor cells in the retina. Lysosomal storage was found in large neurons of the medulla, the hippocampus and in Purkinje cells of the cerebellum in mutant mice. The ultrastructure of the storage material revealed dense lamellar bodies with irregular forms within cerebellar and hippocampal neurons. In the brain loss of Cln7 was accompanied by mild reactive microgliosis and subtle astrogliosis by 10months of age, respectively. In summary we have generated a mouse model which is partly valuable as some but not all neuropathological features of human CLN7 disease are recapitulated thus representing an animal model to study CLN7-specific disease mechanisms.


Subject(s)
Disease Models, Animal , Gene Expression Regulation, Enzymologic/genetics , Membrane Transport Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Animals , Brain/metabolism , Brain/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Humans , Kidney/enzymology , Kidney/pathology , Kidney/ultrastructure , Liver/enzymology , Mice , Mice, Inbred C57BL , Microscopy, Electron , Neuronal Ceroid-Lipofuscinoses/pathology , Neuronal Ceroid-Lipofuscinoses/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology , Retina/ultrastructure , Subcellular Fractions/metabolism , Subcellular Fractions/pathology , alpha-Mannosidase/metabolism , beta-Galactosidase/metabolism , beta-N-Acetylhexosaminidases/metabolism
9.
Invest Ophthalmol Vis Sci ; 54(10): 6952-9, 2013 Oct 23.
Article in English | MEDLINE | ID: mdl-24084090

ABSTRACT

PURPOSE: Mutations in the CLN6 gene cause variant late-infantile neuronal ceroid lipofuscinosis, a lysosomal storage disorder clinically characterized by progressive loss of vision, dementia, seizures, and early death. Here, we analyzed the time course of photoreceptor loss and the role of lysosomes in nclf mice, an animal model of the human CLN6 disease. METHODS: Labeling of apoptotic cells, activated astrocytes, and Müller cells, and expression analyses of glial fibrillary acidic protein, rhodopsin, and lysosomal proteins were performed on nclf mice during the course of retinal degeneration. In addition, the distribution and variability of storage material was examined at the ultrastructural level. RESULTS: Progressive apoptotic loss of photoreceptor cells was observed in nclf mice, resulting in reduction of the outer nuclear layer to approximately 3 rows of photoreceptor cells at 9 months of age. Onset of reactive gliosis was observed in 1-month-old nclf mice. Ultrastructural analysis revealed lysosomal storage material containing curvilinear and fingerprint-like inclusions in various retinal cell types. Expression levels of soluble mannose 6-phosphate-containing lysosomal enzymes, such as cathepsin D and the lysosomal membrane protein Lamp1, were increased in retinal cells of nclf mice. CONCLUSIONS: Accumulation of heterogeneous nondegraded macromolecules in dysfunctional lysosomes and autolysosomes impairs photoreceptor cells, ultimately leading to early-onset apoptotic death with subsequent activation of astrocytes and Müller cells in the retina of nclf mice. The defined steps of photoreceptor degeneration suggest that nclf mice might serve as an ideal animal model for experimental therapeutic approaches aimed at attenuating vision loss in neuronal ceroid lipofuscinosis.


Subject(s)
Neuronal Ceroid-Lipofuscinoses , Photoreceptor Cells , Proteins/metabolism , Animals , Disease Models, Animal , Immunohistochemistry , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Microscopy, Electron , Neuronal Ceroid-Lipofuscinoses/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Time Factors
10.
Stem Cells Transl Med ; 2(12): 1001-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24167317

ABSTRACT

A continuous intraocular delivery of neurotrophic factors (NFs) is being explored as a strategy to rescue photoreceptor cells and visual functions in degenerative retinal disorders that are currently untreatable. To establish a cell-based intraocular delivery system for a sustained administration of NFs to the dystrophic mouse retina, we used a polycistronic lentiviral vector to genetically modify adherently cultivated murine neural stem (NS) cells. The vector concurrently encoded a gene of interest, a reporter gene, and a resistance gene and thus facilitated the selection, cloning, and in vivo tracking of the modified cells. To evaluate whether modified NS cells permit delivery of functionally relevant quantities of NFs to the dystrophic mouse retina, we expressed a secretable variant of ciliary neurotrophic factor (CNTF) in NS cells and grafted the cells into the vitreous space of Pde6b(rd1) and Pde6b(rd10) mice, two animal models of retinitis pigmentosa. In both mouse lines, grafted cells attached to the retina and lens, where they differentiated into astrocytes and some neurons. Adverse effects of the transplanted cells on the morphology of host retinas were not observed. Importantly, the CNTF-secreting NS cells significantly attenuated photoreceptor degeneration in both mutant mouse lines. The neuroprotective effect was significantly more pronounced when clonally derived NS cell lines selected for high expression levels of CNTF were grafted into Pde6b(rd1) mice. Intravitreal transplantations of modified NS cells may thus represent a useful method for preclinical studies aimed at evaluating the therapeutic potential of a cell-based intraocular delivery of NFs in mouse models of photoreceptor degeneration.


Subject(s)
Ciliary Neurotrophic Factor/metabolism , Genetic Therapy , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Retina/metabolism , Retinitis Pigmentosa/therapy , Animals , Cells, Cultured , Ciliary Neurotrophic Factor/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Disease Models, Animal , Genetic Vectors , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Neurogenesis , Photoreceptor Cells, Vertebrate/metabolism , Retina/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Spheroids, Cellular , Transduction, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...