43 results on '"Susanne C. Beck"'
Search Results
2. Contribution of the hexosamine biosynthetic pathway in the hyperglycemia-dependent and -independent breakdown of the retinal neurovascular unit
- Author
-
Yixin Wang, Rachana Eshwaran, Susanne C. Beck, Hans-Peter Hammes, Thomas Wieland, and Yuxi Feng
- Subjects
Cell Biology ,Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
3. Targeting CSF1R Alone or in Combination with PD1 in Experimental Glioma
- Author
-
Ghazaleh Tabatabai, Hannes Becker, Nicole Anderle, Susanne C. Beck, Marilin Koch, Anna-Lena Keller, Anja Pohl, Denis Canjuga, Nataliya Korinetska, Justyna Przystal, Foteini Tsiami, Martina Schmittnaegel, Jens Schittenhelm, Christian Schmees, Marcos Tatagiba, and Carola Ries
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Population ,Article ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Glioma ,medicine ,Cytotoxicity ,education ,RC254-282 ,education.field_of_study ,sequential therapy ,business.industry ,glioblastoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,CSF1R ,medicine.disease ,Primary tumor ,Blockade ,PD1 ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,immunotherapy ,business ,Ex vivo - Abstract
Simple Summary Glioblastomas are incurable tumors of the central nervous system. Currently, treatment strategies combine neurosurgical intervention, radiation therapy, and chemotherapy. Yet, clinical experience shows that tumors acquire escape mechanisms. Furthermore, the tumor-associated microenvironment, including macrophages expressing the receptor CSF1R, promote and nourish tumor cells. The so-called PD1/PDL1 axis is a major reason why tumors can grow with a “magic hat”; i.e., unrecognized from the immune system. The aim of our study was to assess treatment strategies that target macrophages in the microenvironment by blocking CSF1R alone or in combination with PD1 blockade. Using an immune competent mouse model and an ex vivo microtumor model using freshly resected glioblastoma material, we observed prolonged survival of treated mice and an improved “attack” of the immune system. We conclude that targeting CSF1R is a promising strategy that should be explored in clinical trials, potentially in combination with PD1 blockade. Abstract Glioblastoma is an aggressive primary tumor of the central nervous system. Targeting the immunosuppressive glioblastoma-associated microenvironment is an interesting therapeutic approach. Tumor-associated macrophages represent an abundant population of tumor-infiltrating host cells with tumor-promoting features. The colony stimulating factor-1/ colony stimulating factor-1 receptor (CSF-1/CSF1R) axis plays an important role for macrophage differentiation and survival. We thus aimed at investigating the antiglioma activity of CSF1R inhibition alone or in combination with blockade of programmed death (PD) 1. We investigated combination treatments of anti-CSF1R alone or in combination with anti-PD1 antibodies in an orthotopic syngeneic glioma mouse model, evaluated post-treatment effects and assessed treatment-induced cytotoxicity in a coculture model of patient-derived microtumors (PDM) and autologous tumor-infiltrating lymphocytes (TILs) ex vivo. Anti-CSF1R monotherapy increased the latency until the onset of neurological symptoms. Combinations of anti-CSF1R and anti-PD1 antibodies led to longterm survivors in vivo. Furthermore, we observed treatment-induced cytotoxicity of combined anti-CSF1R and anti-PD1 treatment in the PDM/TILs cocultures ex vivo. Our results identify CSF1R as a promising therapeutic target for glioblastoma, potentially in combination with PD1 inhibition.
- Published
- 2021
4. Accessory heterozygous mutations in cone photoreceptor CNGA3 exacerbate CNG channel–associated retinopathy
- Author
-
Richard G. Weleber, Robert Lukowski, M. W. Seeliger, Christina Brennenstuhl, Anne E. Bausch, Xiangang Zong, Sascha Venturelli, John R. Heckenlively, Vithiyanjali Sothilingam, Stylianos Michalakis, Susanne C. Beck, Günther Rudolph, Naoyuki Tanimoto, Ulrich Kellner, Peggy Reuter, Anja K. Mayer, Ditta Zobor, Susanne Kohl, Bernd Wissinger, Gesa Astrid Hahn, Britta Baumann, Paul A. Sieving, Xi-Qin Ding, Nicole Weisschuh, Christian P. Hamel, Martin Biel, Robert K. Koenekoop, Peter Ruth, Peter Charbel Issa, Timm Krätzig, Gesine Huber, Elvir Becirovic, Markus Burkard, Katrin Junger, Institute of Human Genetics [Erlangen, Allemagne], Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Institut des Neurosciences de Montpellier - Déficits sensoriels et moteurs (INM), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), and Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier)
- Subjects
0301 basic medicine ,Achromatopsia ,[SDV]Life Sciences [q-bio] ,Mutant ,Color Vision Defects ,Eye ,medicine.disease_cause ,Medical and Health Sciences ,Transgenic ,Mice ,Cone dystrophy ,2.1 Biological and endogenous factors ,Molecular genetics ,Aetiology ,Genetics ,Mutation ,General Medicine ,Phenotype ,Retinal Cone Photoreceptor Cells ,Ion Channel Gating ,Research Article ,Heterozygote ,medicine.medical_specialty ,Retinal Disorder ,Immunology ,Mutation, Missense ,Cyclic Nucleotide-Gated Cation Channels ,Mice, Transgenic ,Biology ,03 medical and health sciences ,Retinal Diseases ,medicine ,Animals ,Humans ,Allele ,Retinopathy ,Eye Disease and Disorders of Vision ,Animal ,Neurosciences ,medicine.disease ,Brain Disorders ,Disease Models, Animal ,Ophthalmology ,HEK293 Cells ,030104 developmental biology ,Amino Acid Substitution ,Disease Models ,Missense - Abstract
International audience; Mutations in CNGA3 and CNGB3, the genes encoding the subunits of the tetrameric cone photoreceptor cyclic nucleotide–gated ion channel, cause achromatopsia, a congenital retinal disorder characterized by loss of cone function. However, a small number of patients carrying the CNGB3/c.1208G>A;p.R403Q mutation present with a variable retinal phenotype ranging from complete and incomplete achromatopsia to moderate cone dysfunction or progressive cone dystrophy. By exploring a large patient cohort and published cases, we identified 16 unrelated individuals who were homozygous or (compound-)heterozygous for the CNGB3/c.1208G>A;p.R403Q mutation. In-depth genetic and clinical analysis revealed a co-occurrence of a mutant CNGA3 allele in a high proportion of these patients (10 of 16), likely contributing to the disease phenotype. To verify these findings, we generated a Cngb3R403Q/R403Q mouse model, which was crossbred with Cnga3-deficient (Cnga3–/–) mice to obtain triallelic Cnga3+/– Cngb3R403Q/R403Q mutants. As in human subjects, there was a striking genotype-phenotype correlation, since the presence of 1 Cnga3-null allele exacerbated the cone dystrophy phenotype in Cngb3R403Q/R403Q mice. These findings strongly suggest a digenic and triallelic inheritance pattern in a subset of patients with achromatopsia/severe cone dystrophy linked to the CNGB3/p.R403Q mutation, with important implications for diagnosis, prognosis, and genetic counseling.
- Published
- 2018
- Full Text
- View/download PDF
5. Gene Therapy Successfully Delays Degeneration in a Mouse Model ofPDE6A-Linked Retinitis Pigmentosa (RP43)
- Author
-
Christian Schön, Vithiyanjali Sothilingam, Regine Mühlfriedel, Marina Garcia Garrido, Susanne C. Beck, Naoyuki Tanimoto, Bernd Wissinger, François Paquet-Durand, Martin Biel, Stylianos Michalakis, Mathias W. Seeliger, and null the RD-Cure Consortium
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,genetic structures ,Transgene ,Genetic enhancement ,030105 genetics & heredity ,03 medical and health sciences ,In vivo ,Retinitis pigmentosa ,Genetics ,medicine ,Molecular Biology ,Retina ,biology ,medicine.diagnostic_test ,business.industry ,Therapeutic effect ,medicine.disease ,medicine.anatomical_structure ,Rhodopsin ,biology.protein ,Molecular Medicine ,sense organs ,business ,Electroretinography - 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...
- Published
- 2017
- Full Text
- View/download PDF
6. Retinitis pigmentosa: impact of differentPde6apoint mutations on the disease phenotype
- Author
-
Dragana Trifunović, Stylianos Michalakis, Christian Schön, Vithiyanjali Sothilingam, Elena Buena-Atienza, Angelique Heckmann, Kangwei Jiao, Marina Garcia Garrido, Sukirthini Balendran, Mathias W. Seeliger, Martin Biel, Bernd Wissinger, Regine Mühlfriedel, Tanja Koepfli, François Paquet-Durand, Ayse Sahaboglu, and Susanne C. Beck
- Subjects
Retinal degeneration ,Cell Survival ,Biology ,Compound heterozygosity ,medicine.disease_cause ,Retina ,Photoreceptor cell ,Mice ,Retinal Rod Photoreceptor Cells ,Locus heterogeneity ,Retinitis pigmentosa ,Genetics ,medicine ,Animals ,Humans ,Point Mutation ,Eye Proteins ,Molecular Biology ,Genetics (clinical) ,Cyclic Nucleotide Phosphodiesterases, Type 6 ,Mutation ,Calpain ,Genetic heterogeneity ,Point mutation ,General Medicine ,medicine.disease ,Disease Models, Animal ,medicine.anatomical_structure ,Caspases ,Retinitis Pigmentosa - 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.
- Published
- 2015
- Full Text
- View/download PDF
7. Gene Therapy Successfully Delays Degeneration in a Mouse Model of
- Author
-
Christian, Schön, Vithiyanjali, Sothilingam, Regine, Mühlfriedel, Marina, Garcia Garrido, Susanne C, Beck, Naoyuki, Tanimoto, Bernd, Wissinger, François, Paquet-Durand, Martin, Biel, Stylianos, Michalakis, and Mathias W, Seeliger
- Abstract
Retinitis pigmentosa type 43 (RP43) is a blinding disease caused by mutations in the gene for rod phosphodiesterase 6 alpha (
- Published
- 2017
8. A retinal model of cerebral malaria
- Author
-
François Paquet-Durand, Sylvia Bolz, Benjamin Mordmüller, Marina Garcia-Garrido, Naoyuki Tanimoto, Susanne C. Beck, Wolfgang Hoffmann, Regine Mühlfriedel, Ulrich Schraermeyer, Mathias W. Seeliger, Soumyaparna Das, Le Chang, Timm Schubert, and Gesine Huber
- Subjects
0301 basic medicine ,Plasmodium berghei ,medicine.medical_treatment ,Malaria, Cerebral ,lcsh:Medicine ,Dihydroartemisinin ,Gene Expression ,Mice, Transgenic ,Disease ,Retina ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Genes, Reporter ,parasitic diseases ,medicine ,Electroretinography ,Animals ,lcsh:Science ,Cellular localization ,Multidisciplinary ,biology ,business.industry ,lcsh:R ,Retinal ,medicine.disease ,biology.organism_classification ,Ophthalmoscopy ,Disease Models, Animal ,030104 developmental biology ,Phenotype ,chemistry ,Cerebral Malaria ,Immunology ,lcsh:Q ,business ,030217 neurology & neurosurgery ,Malaria ,Biomarkers ,Tomography, Optical Coherence ,Retinopathy - 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.
- Published
- 2017
9. Alterations of the tunica vasculosa lentis in the rat model of retinopathy of prematurity
- Author
-
Robert J. Munro, Tara L. Favazza, Vithiyanjali Sothilingam, Naoyuki Tanimoto, Susanne C. Beck, Marina Garcia Garrido, Mathias W. Seeliger, James D Akula, Ronald M. Hansen, Anne B. Fulton, and Christina Seide
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,genetic structures ,Retinal Artery ,Persistent Hyperplastic Primary Vitreous ,Retinal Neovascularization ,Retina ,Article ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Physiology (medical) ,Ophthalmology ,Lens, Crystalline ,Electroretinography ,medicine ,Animals ,Humans ,Rats, Long-Evans ,Retinopathy of Prematurity ,Tunica vasculosa lentis ,Fluorescein Angiography ,medicine.diagnostic_test ,business.industry ,Infant, Newborn ,Retinal ,Retinopathy of prematurity ,Anatomy ,medicine.disease ,Fluorescein angiography ,eye diseases ,Sensory Systems ,Rats ,Oxygen ,Arterioles ,Disease Models, Animal ,medicine.anatomical_structure ,Animals, Newborn ,chemistry ,Persistent hyperplastic primary vitreous ,cardiovascular system ,sense organs ,business - Abstract
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.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).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.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.
- Published
- 2013
- Full Text
- View/download PDF
10. Endothelial SRF/MRTF ablation causes vascular disease phenotypes in murine retinae
- Author
-
Dongjeong Park, Heidemarie Riehle, Ralf H. Adams, Hartwig Wolburg, Christine Stritt, Mathias W. Seeliger, Susanne C. Beck, Alfred Nordheim, Christine Weinl, Eric N. Olson, and Gesine Huber
- Subjects
Vascular Endothelial Growth Factor A ,Cytoplasm ,Serum Response Factor ,Pathology ,medicine.medical_specialty ,genetic structures ,Angiogenesis ,Neovascularization, Physiologic ,Biology ,Retina ,Neovascularization ,Mice ,chemistry.chemical_compound ,Retinal Diseases ,Serum response factor ,medicine ,Animals ,RNA, Messenger ,Vascular Diseases ,Cell Nucleus ,Mice, Knockout ,Neovascularization, Pathologic ,Vascular disease ,Retinal ,General Medicine ,medicine.disease ,eye diseases ,Tamoxifen ,Vascular endothelial growth factor A ,Phenotype ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Trans-Activators ,cardiovascular system ,sense organs ,Norrie disease ,medicine.symptom ,Gene Deletion ,Transcription Factors ,Research Article - Abstract
Retinal vessel homeostasis ensures normal ocular functions. Consequently, retinal hypovascularization and neovascularization, causing a lack and an excess of vessels, respectively, are hallmarks of human retinal pathology. We provide evidence that EC-specific genetic ablation of either the transcription factor SRF or its cofactors MRTF-A and MRTF-B, but not the SRF cofactors ELK1 or ELK4, cause retinal hypovascularization in the postnatal mouse eye. Inducible, EC-specific deficiency of SRF or MRTF-A/MRTF-B during postnatal angiogenesis impaired endothelial tip cell filopodia protrusion, resulting in incomplete formation of the retinal primary vascular plexus, absence of the deep plexi, and persistence of hyaloid vessels. All of these features are typical of human hypovascularization-related vitreoretinopathies, such as familial exudative vitreoretinopathies including Norrie disease. In contrast, conditional EC deletion of Srf in adult murine vessels elicited intraretinal neovascularization that was reminiscent of the age-related human pathologies retinal angiomatous proliferation and macular telangiectasia. These results indicate that angiogenic homeostasis is ensured by differential stage-specific functions of SRF target gene products in the developing versus the mature retinal vasculature and suggest that the actin-directed MRTF-SRF signaling axis could serve as a therapeutic target in the treatment of human vascular retinal diseases.
- Published
- 2013
- Full Text
- View/download PDF
11. Murine Autoimmune Optic Neuritis Is Not Phenotypically Altered by the Retinal Degeneration 8 Mutation
- Author
-
Mathias W. Seeliger, Sarah K. Williams, Richard Fairless, Valerie Gimmy, Petra Weissgerber, Susanne C. Beck, Veit Flockerzi, Ricarda Diem, Ulrich Wissenbach, Aleksandar Stojic, and Vithiyanjali Sothilingam
- Subjects
0301 basic medicine ,Retinal degeneration ,Retinal Ganglion Cells ,Pathology ,genetic structures ,DNA Mutational Analysis ,Polymerase Chain Reaction ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Mice, Knockout ,biology ,medicine.diagnostic_test ,Immunohistochemistry ,medicine.anatomical_structure ,Phenotype ,Retinal ganglion cell ,Optic nerve ,Female ,Tomography, Optical Coherence ,medicine.medical_specialty ,Optic Neuritis ,Genotype ,Nerve Tissue Proteins ,Myelin oligodendrocyte glycoprotein ,Autoimmune Diseases ,03 medical and health sciences ,medicine ,Electroretinography ,In Situ Nick-End Labeling ,Animals ,Optic neuritis ,Retina ,business.industry ,Retinal ,Optic Nerve ,DNA ,medicine.disease ,eye diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Mutation ,biology.protein ,Evoked Potentials, Visual ,sense organs ,business ,030217 neurology & neurosurgery - Abstract
Purpose To investigate whether the presence of the retinal degeneration 8 (rd8) mutation in C57BL/6 mice alters the phenotype of autoimmune optic neuritis (AON). Methods C57BL/6J and C57BL/6N mice were genotyped for the rd8 mutation and fundus analyses and examination of retinal layer morphology were performed in vivo by scanning laser ophthalmoscopy and optical coherence tomography. Visual function was assessed by recording electroretinographs, and visual evoked potentials and retinae and optic nerves were assessed histopathologically. Retinal ganglion cell numbers were determined by retrograde labeling with fluorogold. Mice were then immunized with myelin oligodendrocyte glycoprotein 35-55 to induce AON before assessment of retinal ganglion cell degeneration, inflammatory infiltration of retinae and optic nerves, and demyelination. Furthermore, visual function was assessed by visual evoked potentials. Results All C57BL/6N mice were homozygous for the mutation (Crb1rd8/rd8) and had pathology typical of the rd8 mutation; however, this was not seen in the C57BL/6J (Crb1wt/wt) mice. Following induction of AON, no differences were seen between the Crb1rd8/rd8 and Crb1wt/wt mice regarding disease parameters nor regarding inner retinal degeneration either in the retina as a whole or in the inferior nasal quadrant. Conclusions The presence of the rd8 mutation in C57BL/6 mice does not affect the course of AON and should not provide a confounding factor in the interpretation of experimental results obtained in this model. However, it could be dangerous in other models of ocular pathology.
- Published
- 2017
12. Retina-specific activation of a sustained hypoxia-like response leads to severe retinal degeneration and loss of vision
- Author
-
Mathias W. Seeliger, Naoyuki Tanimoto, Susanne C. Beck, Christian Caprara, Gesine Huber, Marijana Samardzija, Christina Lange, Christian Grimm, University of Zurich, and Lange, C
- Subjects
Retinal degeneration ,genetic structures ,Blindness ,Leukemia Inhibitory Factor ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Basic Helix-Loop-Helix Transcription Factors ,Hypoxia, Brain ,Caspase ,Mice, Knockout ,0303 health sciences ,Cell Death ,Neovascularization, Pathologic ,biology ,Cell biology ,medicine.anatomical_structure ,Neurology ,Von Hippel-Lindau Tumor Suppressor Protein ,10076 Center for Integrative Human Physiology ,Vasculature ,Hypoxia-Inducible Factor 1 ,medicine.symptom ,10018 Ophthalmology Clinic ,medicine.medical_specialty ,Programmed cell death ,Cell Survival ,Retinal Artery ,Mice, Transgenic ,610 Medicine & health ,Retina ,lcsh:RC321-571 ,Von Hippel-Lindau ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,HIF ,Transcription factor ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,030304 developmental biology ,Retinal ,Hypoxia (medical) ,medicine.disease ,eye diseases ,Disease Models, Animal ,Endocrinology ,chemistry ,2808 Neurology ,biology.protein ,570 Life sciences ,sense organs ,Leukemia inhibitory factor ,030217 neurology & neurosurgery - Abstract
Loss of vision and blindness in human patients is often caused by the degeneration of neuronal cells in the retina. In mouse models, photoreceptors can be protected from death by hypoxic preconditioning. Preconditioning in low oxygen stabilizes and activates hypoxia inducible transcription factors (HIFs), which play a major role in the hypoxic response of tissues including the retina. We show that a tissue-specific knockdown of von Hippel-Lindau protein (VHL) activated HIF transcription factors in normoxic conditions in the retina. Sustained activation of HIF1 and HIF2 was accompanied by persisting embryonic vasculatures in the posterior eye and the iris. Embryonic vessels persisted into adulthood and led to a severely abnormal mature vessel system with vessels penetrating the photoreceptor layer in adult mice. The sustained hypoxia-like response also activated the leukemia inhibitory factor (LIF)-controlled endogenous molecular cell survival pathway. However, this was not sufficient to protect the retina against massive cell death in all retinal layers of adult mice. Caspases 1, 3 and 8 were upregulated during the degeneration as were several VHL target genes connected to the extracellular matrix. Misregulation of these genes may influence retinal structure and may therefore facilitate growth of vessels into the photoreceptor layer. Thus, an early and sustained activation of a hypoxia-like response in retinal cells leads to abnormal vasculature and severe retinal degeneration in the adult mouse retina.
- Published
- 2011
- Full Text
- View/download PDF
13. Restoration of Cone Vision in the CNGA3−/− Mouse Model of Congenital Complete Lack of Cone Photoreceptor Function
- Author
-
Vidhyasankar Krishnamoorthy, Susanne Koch, Gesine Huber, E. Fahl, François Paquet-Durand, Xiangang Zong, Hildegard Büning, Susanne C. Beck, Lin Bai, Elvir Becirovic, Tim Gollisch, Martin Biel, Stylianos Michalakis, Regine Mühlfriedel, Naoyuki Tanimoto, Mathias W. Seeliger, and M. Dominik Fischer
- Subjects
Opsin ,Achromatopsia ,genetic structures ,Genetic Vectors ,Cyclic Nucleotide-Gated Cation Channels ,Sensory system ,Biology ,Retinal Cone Photoreceptor Cells ,Congenital Abnormalities ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Cloning, Molecular ,Molecular Biology ,Vision, Ocular ,030304 developmental biology ,Mice, Knockout ,Pharmacology ,0303 health sciences ,Retina ,Retinal ,Genetic Therapy ,Anatomy ,Dependovirus ,medicine.disease ,eye diseases ,Cone cell ,Cell biology ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,030221 ophthalmology & optometry ,Molecular Medicine ,Original Article ,sense organs ,Muller glia - Abstract
Congenital absence of cone photoreceptor function is associated with strongly impaired daylight vision and loss of color discrimination in human achromatopsia. Here, we introduce viral gene replacement therapy as a potential treatment for this disease in the CNGA3(-/-) mouse model. We show that such therapy can restore cone-specific visual processing in the central nervous system even if cone photoreceptors had been nonfunctional from birth. The restoration of cone vision was assessed at different stages along the visual pathway. Treated CNGA3(-/-) mice were able to generate cone photoreceptor responses and to transfer these signals to bipolar cells. In support, we found morphologically that treated cones expressed regular cyclic nucleotide-gated (CNG) channel complexes and opsins in outer segments, which previously they did not. Moreover, expression of CNGA3 normalized cyclic guanosine monophosphate (cGMP) levels in cones, delayed cone cell death and reduced the inflammatory response of Müller glia cells that is typical of retinal degenerations. Furthermore, ganglion cells from treated, but not from untreated, CNGA3(-/-) mice displayed cone-driven, light-evoked, spiking activity, indicating that signals generated in the outer retina are transmitted to the brain. Finally, we demonstrate that this newly acquired sensory information was translated into cone-mediated, vision-guided behavior.
- Published
- 2010
- Full Text
- View/download PDF
14. Cone loss is delayed relative to rod loss during induced retinal degeneration in the diurnal cone-rich rodent Arvicanthis ansorgei
- Author
-
Gesine Huber, M. W. Seeliger, Naoyuki Tanimoto, Domitille L. Boudard, David Hicks, and Susanne C. Beck
- Subjects
Male ,Retinal degeneration ,Pathology ,medicine.medical_specialty ,genetic structures ,Photoreceptor cell ,chemistry.chemical_compound ,Retinal Rod Photoreceptor Cells ,medicine ,Animals ,Scotopic vision ,Retina ,biology ,General Neuroscience ,Retinal Degeneration ,Arvicanthis ,Retinal ,Anatomy ,biology.organism_classification ,medicine.disease ,Circadian Rhythm ,Muridae ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,Rhodopsin ,Nerve Degeneration ,Retinal Cone Photoreceptor Cells ,biology.protein ,sense organs ,Transducin - Abstract
Cone photoreceptor breakdown underlies functional vision loss in many blinding diseases. Cone loss is often secondary to that of rods, but little experimental data are available on the relationship between the two populations. Because of its high cone numbers, we used the diurnal rodent Arvicanthis ansorgei to explore changes in rod and cone survival and function during chemically-induced retinal degeneration. Adult animals received intraperitoneal injections of N-methyl-N-nitrosourea (MNU), and changes in retinal fundus appearance, histology, phenotype, apoptosis (TUNEL staining) and functionality (scotopic and photopic electroretinography) were monitored as a function of post-treatment time and retinal topography. Relative to control animals injected with vehicle only, MNU-injected animals showed time-, region- and population-specific changes as measured by morphological and immunochemical criteria. Histological (gradual thinning of photoreceptor layer) and phenotypical (reduced immunostaining of rhodopsin and rod transducin, and mid wavelength cone opsin and cone arrestin) modifications were first observed in superior central retina at 11 days post-injection. These degenerative changes spread into the superior peripheral and inferior hemisphere during the following 10 days. Rod loss preceded that of cones as visualized by differential immunolabelling and presence of apoptotic cells in rod but not cone cells. By 3 months post-injection, degeneration of the photoreceptor layer was complete in the superior hemisphere, but only partial in the inferior hemisphere. Despite the persistence of cone photoreceptors, scotopic and photopic electroretinography performed at 90 days post-treatment showed that both rod and cone function were severely compromised. In conclusion, MNU-induced retinal degeneration in Arvicanthis follows a predictable spatial and temporal pattern allowing clear separation of rod- and cone-specific pathogenic mechanisms. Compared to other rodents in which MNU has been used, Arvicanthis ansorgei demonstrates pronounced resistance to photoreceptor cell loss.
- Published
- 2010
- Full Text
- View/download PDF
15. Cooperative Phagocytes
- Author
-
Martin Zinkernagel, Christian Grimm, Sandrine Joly, Andreas Reichenbach, Elke Ulbricht, M. W. Seeliger, Marijana Samardzija, Bernhard Odermatt, Karl S. Lang, Johannes Hirrlinger, Petra G. Hirrlinger, Mike Francke, Charlotte E. Remé, and Susanne C. Beck
- Subjects
Pathology ,medicine.medical_specialty ,Retina ,genetic structures ,Microglia ,Phagocyte ,Retinal ,Biology ,eye diseases ,Pathology and Forensic Medicine ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,medicine ,Optic nerve ,Neuroglia ,Macrophage ,sense organs ,Bone marrow - Abstract
Phagocytosis is essential for the removal of photoreceptor debris following retinal injury. We used two mouse models, mice injected with green fluorescent protein-labeled bone marrow cells or green fluorescent protein-labeled microglia, to study the origin and activation patterns of phagocytic cells after acute blue light-induced retinal lesions. We show that following injury, blood-borne macrophages enter the eye via the optic nerve and ciliary body and soon migrate into the injured retinal area. Resident microglia are also activated rapidly throughout the entire retina and adopt macrophage characteristics only in the injured region. Both blood-borne- and microglia-derived macrophages were involved in the phagocytosis of dead photoreceptors. No obvious breakdown of the blood-retinal barrier was observed. Ccl4, Ccl12, Tgfb1, Csf1, and Tnf were differentially expressed in both the isolated retina and the eyecup of wild-type mice. Debris-laden macrophages appeared to leave the retina into the general circulation, suggesting their potential to become antigen-presenting cells. These experiments provide evidence that both local and immigrant macrophages remove apoptotic photoreceptors and cell debris in the injured retina.
- Published
- 2009
- Full Text
- View/download PDF
16. Retinal degenerative and hypoxic ischemic disease
- Author
-
E. Fahl, Mathias W. Seeliger, Anne Moskowitz, Susanne C. Beck, Anne B. Fulton, Julie A. Mocko, James D Akula, Ilan Y. Benador, Ronald M. Hansen, and Maureen E. Harris
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,genetic structures ,Ischemia ,Biology ,Retina ,Article ,chemistry.chemical_compound ,Degenerative disease ,Retinal Rod Photoreceptor Cells ,Physiology (medical) ,Electroretinography ,medicine ,Animals ,Humans ,Retinopathy of Prematurity ,Hypoxia ,medicine.diagnostic_test ,Retinal Degeneration ,Infant, Newborn ,Retinal Vessels ,Retinopathy of prematurity ,Retinal ,Hypoxia (medical) ,medicine.disease ,eye diseases ,Sensory Systems ,Rats ,Surgery ,Disease Models, Animal ,Ophthalmology ,medicine.anatomical_structure ,chemistry ,sense organs ,medicine.symptom ,Neuroscience ,Retinopathy - Abstract
A broad spectrum of retinal diseases affects both the retinal vasculature and the neural retina, including photoreceptor and postreceptor layers. The accepted clinical hallmarks of acute retinopathy of prematurity (ROP) are dilation and tortuosity of the retinal vasculature. Additionally, significant early and persistent effects on photoreceptor and postreceptor neural structures and function are demonstrated in ROP. In this paper, we focus on the results of longitudinal studies of electroretinographic (ERG) and vascular features in rats with induced retinopathies that model the gamut of human ROP, mild to severe. Two potential targets for pharmaceutical interventions emerge from the observations. The first target is immature photoreceptors because the status of the photoreceptors at an early age predicts later vascular outcome; this approach is appealing as it holds promise to prevent ROP. The second target is the interplay of the neural and vascular retinal networks, which develop cooperatively. Beneficial pharmaceutical interventions may be measured in improved visual outcome as well as lessening of the vascular abnormalities.
- Published
- 2008
- Full Text
- View/download PDF
17. Influence of the β2-Subunit of L-Type Voltage-Gated Cav Channels on the Structural and Functional Development of Photoreceptor Ribbon Synapses
- Author
-
Vithiyanjali Sothilingam, Marina Garcia Garrido, Janka Dörr, Elisabeth Roth, Veit Flockerzi, Petra Weissgerber, Andreas Beck, Rashmi Katiyar, Frank Schmitz, Mathias W. Seeliger, and Susanne C. Beck
- Subjects
Male ,genetic structures ,Calcium Channels, L-Type ,Blotting, Western ,Presynaptic Terminals ,Mice, Transgenic ,Neurotransmission ,Ribbon synapse ,Synaptic Transmission ,Synapse ,chemistry.chemical_compound ,Mice ,Microscopy, Electron, Transmission ,Retinal Rod Photoreceptor Cells ,medicine ,Electroretinography ,Animals ,Active zone ,Retina ,Voltage-gated ion channel ,medicine.diagnostic_test ,Retinal ,Immunohistochemistry ,eye diseases ,Cell biology ,medicine.anatomical_structure ,chemistry ,Synapses ,Retinal Cone Photoreceptor Cells ,Female ,sense organs - Abstract
PURPOSE The cacnb2 gene encodes the β2 subunit (Cavβ2) of voltage-gated Ca2+ channels in photoreceptors, and its targeted deletion in mice has previously been shown to cause altered retinal morphology and synaptic transmission. The purpose of this study was to provide a detailed morphologic study combined with experiments on the altered functions of photoreceptor ribbon synapses lacking Cavβ2. METHODS A cacnb2-deficient mouse strain was generated and deletion of the Cavβ2 in the retina documented by biochemical and immunhistochemical approaches. Ultrastructural changes of photoreceptor ribbon synapses were examined by electronmicroscopy and functional implications of the lack of Cavβ2 studied by depolarization-induced Ca2+ influx into isolated photoreceptor cells and electroretinography. RESULTS Voltage-gated Ca2+ influx into rod photoreceptors lacking Cavβ2 was abolished and the typical rod ribbon-type active zones were absent in Cavβ2-deficient retinas. The active zone and the architecture of the presynaptic terminals were severely altered in rod synapses. Cone photoreceptor and the bipolar cell ribbon synapses were largely spared from ultrastructural changes although peanut agglutinin (PNA) labelling and photopic ERG analyses demonstrated that also cone pathways were disturbed in Cavβ2-deficient retinas. CONCLUSIONS The presence of the Cavβ2 is essential for the structural integrity and function of the rod photoreceptor synapse. The Cavβ2 is less essential for the morphology of cone and bipolar cell ribbon synapses, although the impaired photopic electroretinogram suggests a functional alteration also of the cone-mediated signaling in Cavβ2-deficient retinas.
- Published
- 2015
18. Scale Adjustments to Facilitate Two-Dimensional Measurements in OCT Images
- Author
-
Marina, Garcia Garrido, Regine L, Mühlfriedel, Susanne C, Beck, Christine, Wallrapp, and Mathias W, Seeliger
- Subjects
Mice, 129 Strain ,genetic structures ,Green Fluorescent Proteins ,Retinal Vessels ,Eye ,eye diseases ,Retina ,Ophthalmoscopy ,Mice ,Image Processing, Computer-Assisted ,Animals ,sense organs ,Tomography, Optical Coherence ,Research Article - 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.
- Published
- 2015
19. A splice site mutation in the murine Opa1 gene features pathology of autosomal dominant optic atrophy
- Author
-
Lukas Rüttiger, Hans F. Wehrl, Thomas Peters, Marlies Knipper, Bernd Wissinger, Susanne C. Beck, Frank Schuettauf, Felix Tonagel, Simone Schimpf, Juergen Laufs, Marcel V. Alavi, Ulrich Schraermeyer, Bernd J. Pichler, and Stefanie Bette
- Subjects
Retinal Ganglion Cells ,Pathology ,medicine.medical_specialty ,Transcription, Genetic ,Mutant ,Biology ,DNA, Mitochondrial ,Retinal ganglion ,Retina ,GTP Phosphohydrolases ,Mice ,Hearing ,Microscopy, Electron, Transmission ,Mutant protein ,Optic Atrophy, Autosomal Dominant ,Electroretinography ,medicine ,Animals ,Amino Acids ,Cells, Cultured ,Mice, Inbred C3H ,Splice site mutation ,Optic Nerve ,Exons ,medicine.disease ,Magnetic Resonance Imaging ,eye diseases ,Mitochondria ,Ganglion ,Disease Models, Animal ,medicine.anatomical_structure ,Sensory Thresholds ,Mutation ,Optic nerve ,RNA Splice Sites ,Neurology (clinical) ,DNA, Circular ,Mitochondrial optic neuropathies ,Haploinsufficiency - Abstract
Autosomal dominant optic atrophy (adOA) is a juvenile onset, progressive ocular disorder characterized by bilateral loss of vision, central visual field defects, colour vision disturbances, and optic disc pallor. adOA is most frequently associated with mutations in OPA1 encoding a dynamin-related large GTPase that localizes to mitochondria. Histopathological studies in adOA patients have shown a degeneration of retinal ganglion cells (RGCs) and a loss of axons in the optic nerve. However little is known about the molecular mechanism and pathophysiology of adOA due to the lack of appropriate in vivo models. Here we report a first mouse model carrying a splice site mutation (c.1065 + 5G --> A) in the Opa1 gene. The mutation induces a skipping of exon 10 during transcript processing and leads to an in-frame deletion of 27 amino acid residues in the GTPase domain. Western blot analysis showed no evidence of a shortened mutant protein but a approximately 50% reduced OPA1 protein level supporting haploinsufficiency as a major disease mechanism in adOA. Homozygous mutant mice die in utero during embryogenesis with first notable developmental delay at E8.5 as detected by magnetic resonance imaging (MRI). Heterozygous mutants are viable and of normal habitus but exhibit an age-dependent loss of RGCs that eventually progresses to a severe degeneration of the ganglion cell and nerve fibre layer. In addition optic nerves of mutant mice showed a reduced number of axons, and a swelling and abnormal shape of the remaining axons. Mitochondria in these axons showed disorganized cristae structures. All these defects recapitulate crucial features of adOA in humans and therefore document the validity and importance of this model for future research.
- Published
- 2006
- Full Text
- View/download PDF
20. Mpp4 recruits Psd95 and Veli3 towards the photoreceptor synapse
- Author
-
Inge Versteeg, Naoyuki Tanimoto, Albena Kantardzhieva, Felix Tonagel, Mathias W. Seeliger, Jan Klooster, Serge A. van de Pavert, Bob Nunes Cardozo, Susanne C. Beck, Agnes G.S.H. van Rossum, Wendy M. Aartsen, and Jan Wijnholds
- Subjects
Retinal degeneration ,Scaffold protein ,Down-Regulation ,Outer plexiform layer ,Nerve Tissue Proteins ,Biology ,Retina ,Synapse ,Mice ,Retinal Diseases ,Cell polarity ,Electroretinography ,Genetics ,medicine ,Animals ,Photoreceptor Cells ,Molecular Biology ,Genetics (clinical) ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Models, Genetic ,Cell Membrane ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,General Medicine ,medicine.disease ,Immunohistochemistry ,Cell biology ,Microscopy, Electron ,medicine.anatomical_structure ,Membrane protein ,Disks Large Homolog 4 Protein ,Synapses ,sense organs ,Guanylate Kinases ,Signal Transduction - Abstract
Membrane-associated guanylate kinase (MAGUK) proteins function as scaffold proteins contributing to cell polarity and organizing signal transducers at the neuronal synapse membrane. The MAGUK protein Mpp4 is located in the retinal outer plexiform layer (OPL) at the presynaptic plasma membrane and presynaptic vesicles of photoreceptors. Additionally, it is located at the outer limiting membrane (OLM) where it might be involved in OLM integrity. In Mpp4 knockout mice, loss of Mpp4 function only sporadically causes photoreceptor displacement, without changing the Crumbs (Crb) protein complex at the OLM, adherens junctions or synapse structure. Scanning laser ophthalmology revealed no retinal degeneration. The minor morphological effects suggest that Mpp4 is a candidate gene for mild retinopathies only. At the OPL, Mpp4 is essential for correct localization of Psd95 and Veli3 at the presynaptic photoreceptor membrane. Psd95 labeling is absent of presynaptic membranes in both rods and cones but still present in cone basal contacts and dendritic contacts. Total retinal Psd95 protein levels are significantly reduced which suggests Mpp4 to be involved in Psd95 turnover, whereas Veli3 proteins levels are not changed. These protein changes in the photoreceptor synapse did not result in an altered electroretinograph. These findings suggest that Mpp4 coordinates Psd95/Veli3 assembly and maintenance at synaptic membranes. Mpp4 is a critical recruitment factor to organize scaffolds at the photoreceptor synapse and is likely to be associated with synaptic plasticity and protein complex transport.
- Published
- 2006
- Full Text
- View/download PDF
21. Mutations in the unfolded protein response regulator ATF6 cause the cone dysfunction disorder achromatopsia
- Author
-
Jennifer Staller, Francesco Benedicenti, Wei-Chieh Chiang, Mathias W. Seeliger, Susanne C. Beck, Irene Gonzalez Menendez, Michel Michaelides, Frans P.M. Cremers, Franco Stanzial, Bernd Wissinger, Susanne Kohl, Eberhart Zrenner, Jonathan H. Lin, Marina Garcia Garrido, Irma Lopez, Nicole Weisschuh, Vithiyanjali Sothilingam, Ditta Zobor, Stanley Chang, Susanne Roosing, Günther Rudolph, Jill Beis, Francesca Inzana, Tim M. Strom, Ajoy Vincent, Anneke I. den Hollander, Anthony T. Moore, Elise Héon, Randal J. Kaufman, Huanan Ren, Robert K. Koenekoop, Stephen H. Tsang, and Andrew R. Webster
- Subjects
Adult ,Male ,Candidate gene ,Achromatopsia ,Adolescent ,Transcription, Genetic ,genetic structures ,Mutation, Missense ,Color Vision Defects ,Biology ,Retinal Cone Photoreceptor Cells ,Article ,Young Adult ,Genetics ,medicine ,Animals ,Humans ,Sensory disorders Radboud Institute for Molecular Life Sciences [Radboudumc 12] ,Child ,Genetic Association Studies ,Aged, 80 and over ,Mice, Knockout ,GNAT2 ,ATF6 ,Endoplasmic reticulum ,Middle Aged ,medicine.disease ,Disease gene identification ,eye diseases ,Activating Transcription Factor 6 ,Pedigree ,Mice, Inbred C57BL ,HEK293 Cells ,Unfolded Protein Response ,Unfolded protein response ,Female ,sense organs - Abstract
Item does not contain fulltext Achromatopsia (ACHM) is an autosomal recessive disorder characterized by color blindness, photophobia, nystagmus and severely reduced visual acuity. Using homozygosity mapping and whole-exome and candidate gene sequencing, we identified ten families carrying six homozygous and two compound-heterozygous mutations in the ATF6 gene (encoding activating transcription factor 6A), a key regulator of the unfolded protein response (UPR) and cellular endoplasmic reticulum (ER) homeostasis. Patients had evidence of foveal hypoplasia and disruption of the cone photoreceptor layer. The ACHM-associated ATF6 mutations attenuate ATF6 transcriptional activity in response to ER stress. Atf6(-/-) mice have normal retinal morphology and function at a young age but develop rod and cone dysfunction with increasing age. This new ACHM-related gene suggests a crucial and unexpected role for ATF6A in human foveal development and cone function and adds to the list of genes that, despite ubiquitous expression, when mutated can result in an isolated retinal photoreceptor phenotype.
- Published
- 2015
- Full Text
- View/download PDF
22. VEGF Mediates ApoE4-Induced Neovascularization and Synaptic Pathology in the Choroid and Retina
- Author
-
Marina Garcia Garrido, Ran Antes, Daniel M. Michaelson, Dov Weinberger, Idit Maharshak, Mathias W. Seeliger, Tami Livnat, Tamar Kadar, Susanne C. Beck, and Shiran Salomon-Zimri
- Subjects
Apolipoprotein E ,Vascular Endothelial Growth Factor A ,Pathology ,medicine.medical_specialty ,Apolipoprotein E4 ,Ependymoglial Cells ,Apolipoprotein E3 ,Inflammation ,Mice, Transgenic ,Retinal Neovascularization ,Retina ,Neovascularization ,Basal (phylogenetics) ,chemistry.chemical_compound ,Alzheimer Disease ,mental disorders ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,biology ,Choroid ,Retinal Vessels ,Retinal ,eye diseases ,Choroidal Neovascularization ,Mice, Inbred C57BL ,Disease Models, Animal ,Choroidal neovascularization ,medicine.anatomical_structure ,Neurology ,chemistry ,Astrocytes ,Synapses ,Synaptophysin ,biology.protein ,lipids (amino acids, peptides, and proteins) ,sense organs ,Neurology (clinical) ,medicine.symptom ,human activities - Abstract
Apolipoprotein E4 (apoE4), the most prevalent genetic risk factor for Alzheimer's disease (AD), is associated with neuronal and vascular impairments. Recent findings suggest that retina of apoE4 mice have synaptic and functional impairments. We presently investigated the effects of apoE4 on retinal and choroidal vasculature and the possible role of VEGF in these effects. There were no histological differences between the retinal and choroidal vasculatures of naive apoE3 and apoE4 mice. In contrast, laserdriven choroidal injury induced higher levels of choroidal neovascularization (CNV) in apoE4 than in apoE3 mice. These effects were associated with an inflammatory response and with activation of the Muller cells and asrocytic markers gluthatione synthetase and GFAP, all of which were more pronounced in the apoE4 mice. CNV also induced a transient increase in the levels of the synaptic markers synaptophysin and PSD95 which were however similar in the apoE4 and apoE3 naive mice. Retinal and choroidal VEGF and apoE levels were lower in naive apoE4 than in corresponding apoE3 mice. In contrast, VEGF and apoE levels rose more pronouncedly following laser injury in the apoE4 than in apoE3 mice. Taken together, these findings suggest that the apoE4-induced retinal impairments, under basal conditions, may be related to reduced VEGF levels in the eyes of these mice. The hyper-neovascularization in the apoE4 mice might be driven by increased inflammation and the associated surge in VEGF following injury. Retinal and choroidal VEGF and apoE levels were lower in naive apoE4 than in corresponding apoE3 mice. In contrast, VEGF and apoE levels rose more pronouncedly following laser injury in the apoE4 than in apoE3 mice. Taken together, these findings suggest that the apoE4-induced retinal impairments, under basal conditions, may be related to reduced VEGF levels in the eyes of these mice. The hyper-neovascularization in the apoE4 mice might be driven by increased inflammation and the associated surge in VEGF following injury.
- Published
- 2014
23. Targeted ablation of Crb2 in photoreceptor cells induces retinitis pigmentosa
- Author
-
Lucie P. Pellissier, Susanne C. Beck, John G. Flannery, Joost Verhaagen, Marina Garcia Garrido, Mathias W. Seeliger, Jan Klooster, Rogier M. Vos, Jan Wijnholds, Vithiyanjali Sothilingam, Celso Henrique Alves, Christina Seide, Takahisa Furukawa, Royal Netherlands Academy of Arts and Sciences (KNAW), Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, Department of Retinal Signal Processing, Netherlands Institute for Neuroscience-KNAW, Osaka University, Osaka Bioscience Institute, University of California [Berkeley], University of California, Rotterdamse Vereniging Blindenbelangen, Landelijke St. voor Blinden en Slechtzienden, St. Blindenhulp, St. Oogfonds Nederland, St. Retina Nederland, Netherlands Institute for Neuroscience, Foundation Fighting Blindness TA-GT-0811-0540-NIN TA-GT-0313-0607-NIN, Netherlands Organisation for Health Research and Development (ZonMw) 43200004, European Project: 200234,HEALTH,FP7-HEALTH-2007-A,CRUMBS IN SIGHT(2008), University of California [Berkeley] (UC Berkeley), University of California (UC), and Netherlands Institute for Neuroscience (NIN)
- Subjects
Male ,rétinite pigmentaire ,[SDV.OT]Life Sciences [q-bio]/Other [q-bio.OT] ,genetic structures ,Knockout ,Ependymoglial Cells ,Biology ,Inbred C57BL ,Medical and Health Sciences ,Small hairpin RNA ,chemistry.chemical_compound ,Mice ,Retinitis pigmentosa ,Genetics ,medicine ,Animals ,Photoreceptor Cells ,Molecular Biology ,Genetics (clinical) ,Retinal regeneration ,Mice, Knockout ,Genetics & Heredity ,Retina ,CRB1 ,Gene therapy of the human retina ,Membrane Proteins ,Retinal ,General Medicine ,Biological Sciences ,medicine.disease ,Immunohistochemistry ,eye diseases ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,chemistry ,Neuroglia ,Female ,sense organs ,Retinitis Pigmentosa - 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,weshowed that removal ofmouse 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 tomutations in theCRB1gene. Here,westudied 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. © The Author 2014.Published by Oxford University Press. All rights reserved.
- Published
- 2014
- Full Text
- View/download PDF
24. Gene therapy restores vision and delays degeneration in the CNGB1(-/-) mouse model of retinitis pigmentosa
- Author
-
Stylianos, Michalakis, Susanne, Koch, Vithiyanjali, Sothilingam, Marina, Garcia Garrido, Naoyuki, Tanimoto, Elisabeth, Schulze, Elvir, Becirovic, Fred, Koch, Christina, Seide, Susanne C, Beck, Mathias W, Seeliger, Regine, Mühlfriedel, and Martin, Biel
- Subjects
Mice, Knockout ,Retinal Degeneration ,Cyclic Nucleotide-Gated Cation Channels ,Nerve Tissue Proteins ,Recovery of Function ,Dependovirus ,Disease Models, Animal ,Mice ,Retinal Rod Photoreceptor Cells ,Electroretinography ,Animals ,Maze Learning ,Retinitis Pigmentosa ,Vision, Ocular - 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.
- Published
- 2014
25. Mitogenic and adhesive effects of tenascin-C on human hematopoietic cells are mediated by various functional domains
- Author
-
Claudia A. Müller, Martina Seiffert, Harold P. Erickson, Gerd Klein, Susanne C. Beck, and Franka Schermutzki
- Subjects
Binding Sites ,biology ,Heparin ,Cell growth ,Tenascin C ,Tenascin ,Hematopoietic Stem Cells ,Peptide Mapping ,Molecular biology ,Recombinant Proteins ,Extracellular matrix ,Fibronectin ,Haematopoiesis ,medicine.anatomical_structure ,embryonic structures ,Cell Adhesion ,biology.protein ,medicine ,Humans ,Bone marrow ,Mitogens ,Binding site ,Cell adhesion ,Molecular Biology ,Cells, Cultured - Abstract
In the adult organism, the extracellular matrix molecule tenascin-C is prominently expressed in the bone marrow. Bone marrow mononuclear cells can adhere to plastic-immobilized tenascin-C, and in the present study we have used bacterial expression proteins to map the domains of tenascin-C responsible for binding of hematopoietic cells. A strong binding site was found to be located within the fibrinogen-like domain, and this binding could be inhibited by heparin, suggesting interactions with membrane-bound heparan sulfate proteoglycans. A second strong binding site was identified within the fibronectin type III-like repeats 6-8, and was also inhibitable by heparin. Adhesion to both attachment sites could not be blocked by various anti-integrin antibodies. A third hematopoietic cell binding site is located in the fibronectin type III-like repeats 1-5, which harbor an RGD sequence in the third fibronectin type III-like repeat. Binding to this domain, however, seems to be RGD-independent, since RGD-containing peptides could not inhibit cell binding; the addition of heparin also did not block adhesion to this domain. Since contradictory results had been reported on a proliferative effect of soluble tenascin-C, we also analyzed its activity on hematopoietic cells. The heterogeneous bone marrow mononuclear cells show a striking proliferative response in the presence of tenascin-C which is concentration-dependent. This result indicates a strong mitogenic activity of tenascin-C on primary hematopoietic cells. Using recombinant fragments of human tenascin-C, we identified several mitogenic domains within the tenascin-C molecule. These adhesive and mitogenic effects of tenascin-C suggest a direct functional association with proliferation and differentiation of hematopoietic cells within the bone marrow microenvironment.
- Published
- 1998
- Full Text
- View/download PDF
26. Gene Therapy Restores Vision and Delays Degeneration in the CNGB1−/− Mouse Model of Retinitis Pigmentosa
- Author
-
Susanne C. Beck, Fred Koch, E. Schulze, Susanne Koch, Martin Biel, Sothilingam, Mathias W. Seeliger, Garcia Garrido M, Elvir Becirovic, Christina Seide, Stylianos Michalakis, Naoyuki Tanimoto, and Regine Mühlfriedel
- Subjects
Retinal degeneration ,Retina ,medicine.medical_specialty ,Gene therapy of the human retina ,genetic structures ,medicine.diagnostic_test ,business.industry ,Genetic enhancement ,Retinal ,medicine.disease ,eye diseases ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Ophthalmology ,Retinitis pigmentosa ,medicine ,sense organs ,Retinal Rod Photoreceptor Cells ,business ,Electroretinography - 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.
- Published
- 2014
- Full Text
- View/download PDF
27. Loss of CRB2 in the mouse retina mimics human retinitis pigmentosa due to mutations in the CRB1 gene
- Author
-
Jan Wijnholds, Naoyuki Tanimoto, Mariyam Murtaza, André Le Bivic, Lucie P. Pellissier, Susanne C. Beck, Iswariyaraja Sridevi Gurubaran, Christiaan N. Levelt, Fabrice Richard, Bokyung Park, Gesine Huber, Alicia Sanz Sanz, Mathias W. Seeliger, P. Rashbass, Marina Garcia Garrido, Celso Henrique Alves, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), University of Sheffield [Sheffield], Institut de Biologie du Développement de Marseille (IBDM), Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS), Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, Rotterdamse Vereniging Blinden-belangen, Landelijke St. voor Blinden en Slechtzienden, St. Blindenhulp, St. Oogfonds Nederland, St. Retina Nederland, Netherlands Institute for Neuroscience, Netherlands Organisation for Health Research and Development ZonMw 43200004, Deutsche Forschungsgemeinschaft DFG Se837/5-2 Se837/6-1 Se837/6-2 Se837/7-1, German Ministry of Education and Research BMBF 0314106, French National Research Agency (ANR) BLAN 07-2-186738, European Project: 200234,HEALTH,FP7-HEALTH-2007-A,CRUMBS IN SIGHT(2008), and Molecular Visual Plasticity
- Subjects
genetic structures ,Apoptosis ,MESH: Base Sequence ,MESH: Electroretinography ,MESH: Mice, Knockout ,Polymerase Chain Reaction ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,MESH: Eye Proteins ,MESH: Animals ,MESH: Nerve Tissue Proteins ,Genetics (clinical) ,Mice, Knockout ,0303 health sciences ,CRB1 ,Gene therapy of the human retina ,medicine.diagnostic_test ,MESH: Retina ,General Medicine ,Anatomy ,Scanning laser ophthalmoscopy ,Cell biology ,MESH: Tomography, Optical Coherence ,medicine.anatomical_structure ,MESH: Membrane Proteins ,Muller glia ,Retinitis Pigmentosa ,Tomography, Optical Coherence ,Photoreceptor Cells, Vertebrate ,MESH: DNA Primers ,MESH: Mutation ,Nerve Tissue Proteins ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Retina ,03 medical and health sciences ,MESH: Mice, Inbred C57BL ,Retinitis pigmentosa ,Genetics ,medicine ,Electroretinography ,Animals ,Humans ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,Eye Proteins ,Molecular Biology ,MESH: Mice ,030304 developmental biology ,DNA Primers ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,MESH: Humans ,Base Sequence ,MESH: Apoptosis ,Membrane Proteins ,Retinal ,MESH: Polymerase Chain Reaction ,medicine.disease ,eye diseases ,Mice, Inbred C57BL ,chemistry ,Mutation ,MESH: Photoreceptor Cells, Vertebrate ,MESH: Retinitis Pigmentosa ,sense organs ,030217 neurology & neurosurgery - Abstract
International audience; 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.
- Published
- 2012
- Full Text
- View/download PDF
28. Gene therapy restores vision and delays degeneration in the CNGB1(-/-) mouse model of retinitis pigmentosa
- Author
-
Mathias W. Seeliger, Vithiyanjali Sothilingam, Marina Garcia Garrido, Martin Biel, Stylianos Michalakis, Naoyuki Tanimoto, Regine Mühlfriedel, Elvir Becirovic, Christina Seide, Susanne Koch, Fred Koch, and Susanne C. Beck
- Subjects
Retinal degeneration ,medicine.medical_specialty ,Rhodopsin ,DNA, Complementary ,genetic structures ,Genetic enhancement ,Genetic Vectors ,Cyclic Nucleotide-Gated Cation Channels ,Nerve Tissue Proteins ,Biology ,chemistry.chemical_compound ,Mice ,Ophthalmology ,Retinitis pigmentosa ,Genetics ,medicine ,Electroretinography ,Animals ,Cyclic nucleotide-gated ion channel ,Molecular Biology ,Genetics (clinical) ,Mice, Knockout ,Retina ,Gene therapy of the human retina ,Retinal Degeneration ,Retinal ,General Medicine ,Genetic Therapy ,Dependovirus ,medicine.disease ,Rod Cell Outer Segment ,eye diseases ,Tissue Degeneration ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,sense organs ,Retinitis Pigmentosa - 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.
- Published
- 2012
29. Mice lacking Period 1 and Period 2 circadian clock genes exhibit blue cone photoreceptor defects
- Author
-
Naoyuki Tanimoto, David Hicks, Marie-Paule Felder-Schmittbuhl, Mathias W. Seeliger, Susanne C. Beck, Marina Garcia-Garrido, Christina Seide, Mohammed Bennis, Ouafa Ait-Hmyed, and Vithiyanjali Sothilingam
- Subjects
endocrine system ,genetic structures ,Transcription, Genetic ,Arrestins ,Period (gene) ,Circadian clock ,Biology ,Retina ,chemistry.chemical_compound ,Mice ,OPN1MW ,medicine ,Animals ,Genetics ,General Neuroscience ,Rod Opsins ,Nuclear Receptor Subfamily 1, Group F, Member 2 ,Retinal ,Cell Differentiation ,Period Circadian Proteins ,eye diseases ,Mice, Mutant Strains ,Cell biology ,PER2 ,Mice, Inbred C57BL ,medicine.anatomical_structure ,chemistry ,ARR3 ,Retinal Cone Photoreceptor Cells ,sense organs ,PER1 - 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.
- Published
- 2012
30. Gene therapy restores missing cone-mediated vision in the CNGA3-/- mouse model of achromatopsia
- Author
-
Stylianos, Michalakis, Regine, Mühlfriedel, Naoyuki, Tanimoto, Vidhyasankar, Krishnamoorthy, Susanne, Koch, M Dominik, Fischer, Elvir, Becirovic, Lin, Bai, Gesine, Huber, Susanne C, Beck, Edda, Fahl, Hildegard, Büning, Jennifer, Schmidt, Xiangang, Zong, Tim, Gollisch, Martin, Biel, and Mathias W, Seeliger
- Subjects
Color Vision ,Cyclic Nucleotide-Gated Cation Channels ,Color Vision Defects ,Genetic Therapy ,Recovery of Function ,Dependovirus ,Recombinant Proteins ,Mice ,HEK293 Cells ,Retinal Rod Photoreceptor Cells ,Electroretinography ,Retinal Cone Photoreceptor Cells ,Animals ,Humans - Published
- 2011
31. Implantation of ultrathin, biofunctionalized polyimide membranes into the subretinal space of rats
- Author
-
Blanca Arango-Gonzalez, Milada Šírová, Hagen Thielecke, Sylvie Julien, Etienne Schacht, Ulrich Schraermeyer, Eberhart Zrenner, Blanka Rihova, Heiko Büth, Tobias Peters, Pavel Rossmann, Sandra Van Vlierberghe, Peter Dubruel, Susanne C. Beck, Focke Ziemssen, and Publica
- Subjects
Materials science ,genetic structures ,Cell Survival ,Biophysics ,Implantation Site ,Bioengineering ,Microscopy, Atomic Force ,Retina ,Biomaterials ,Prosthesis Implantation ,chemistry.chemical_compound ,Organ Culture Techniques ,Microscopy, Electron, Transmission ,Fibrocyte ,medicine ,Cell Adhesion ,Animals ,Rats, Wistar ,medicine.diagnostic_test ,Retinal ,Membranes, Artificial ,Fluorescein angiography ,eye diseases ,Scanning laser ophthalmoscopy ,Rats ,medicine.anatomical_structure ,chemistry ,Mechanics of Materials ,Ceramics and Composites ,Microscopy, Electron, Scanning ,sense organs ,Choroid ,Implant ,Tomography, Optical Coherence ,Biomedical engineering - Abstract
Subretinal implants aim to replace the photoreceptor function in patients suffering from degenerative retinal disease by topically applying electrical stimuli in the subretinal space. Critical obstacles in the design of high-resolution subretinal implants include the proximity of stimulating electrodes to the target cells and enabling nutrient flow between the retina and the choroid. The present work evaluates the adhesion, migration and survival of retinal cells on an ultrathin (5 μm), highly porous (O 1 μm spaced 3 μm), gelatin-coated polyimide (PI) membrane. The biocompatibility was examined in mice indicating a good tolerance upon subcutaneous implantation with only a mild inflammatory response. In addition, organotypic cultures of rat retina evidenced that the porous membrane allowed the necessary nutrient flow for the retinal cell survival and maintenance. A transscleral implantation technique was applied to position the membrane into the subretinal space of rats. The effect on the obtained retinal integration was investigated in vivo using scanning laser ophthalmoscopy (SLO) and optical coherence tomography (OCT). In 12 out of 18 rat eyes, the implant was successfully placed subretinally. SLO and OCT demonstrated complete retinal attachment and fluorescein angiography showed no retinal vascular abnormalities over and around the implant, immediately after and up to four weeks after the implantation. Histological examination of the eyes showed a close attachment of a thin fibrocyte layer to the implant, the occlusion of the pores by living cells and the survival of some photoreceptors at the implantation site.
- Published
- 2010
32. Vax2 regulates retinoic acid distribution and cone opsin expression in the vertebrate eye
- Author
-
Gesine Huber, Pascal Dollé, Ivan Conte, Maria Teresa Pizzo, Mathias W. Seeliger, Tiziana Caramico, Giovanna Alfano, Raffaella Avellino, Benedetta Arnò, Susanne C. Beck, Sandro Banfi, Naoyuki Tanimoto, Alfano, G., Conte, I., Caramico, T., Avellino, R., Arno', Barbara, Pizzo, M. T., Tanimoto, N., Beck, S. C., Huber, G., Dolle, P., Seeliger, M. W., Banfi, S., Alfano, G, Conte, I, Caramico, T, Avellino, R, Arno, B, Pizzo, Mt, Tanimoto, N, Beck, Sc, Huber, G, Dollé, P, Seeliger, M, and Banfi, Sandro
- Subjects
Male ,Mouse ,genetic structures ,Retinoic acid ,Oryzias ,Eye ,Animals, Genetically Modified ,chemistry.chemical_compound ,Mice ,CYP26C1 ,Cytochrome P-450 Enzyme System ,Pregnancy ,OPN1MW ,Cytochrome P450 Family 26 ,In Situ Hybridization ,Genetics ,Mice, Knockout ,Cone opsins ,Gene Expression Regulation, Developmental ,Homeodomain Protein ,Retinoic Acid 4-Hydroxylase ,Cell biology ,Opsin ,medicine.anatomical_structure ,Vax2 ,Retinal Cone Photoreceptor Cells ,Female ,Retinal Cone Photoreceptor Cell ,Oryzias latipes (medaka) ,Mice, Transgenic ,Tretinoin ,Biology ,Rod Opsin ,Retinal ganglion ,CYP26A1 ,medicine ,Animals ,Molecular Biology ,Oryzia ,Homeodomain Proteins ,Cone opsin ,Retina ,Opsins ,Animal ,Gene Expression Profiling ,Rod Opsins ,eye diseases ,chemistry ,Eye development ,Homeobox ,sense organs ,Developmental Biology - Abstract
Vax2 is an eye-specific homeobox gene, the inactivation of which in mouse leads to alterations in the establishment of a proper dorsoventral eye axis during embryonic development. To dissect the molecular pathways in which Vax2 is involved, we performed a transcriptome analysis of Vax2–/– mice throughout the main stages of eye development. We found that some of the enzymes involved in retinoic acid (RA) metabolism in the eye show significant variations of their expression levels in mutant mice. In particular, we detected an expansion of the expression domains of the RA-catabolizing enzymes Cyp26a1 and Cyp26c1, and a downregulation of the RA-synthesizing enzyme Raldh3. These changes determine a significant expansion of the RA-free zone towards the ventral part of the eye. At postnatal stages of eye development, Vax2 inactivation led to alterations of the regional expression of the cone photoreceptor genes Opn1sw (S-Opsin) and Opn1mw (M-Opsin), which were significantly rescued after RA administration. We confirmed the above described alterations of gene expression in the Oryzias latipes (medaka fish) model system using both Vax2 gain- and loss-of-function assays. Finally, a detailed morphological and functional analysis of the adult retina in mutant mice revealed that Vax2 is necessary for intraretinal pathfinding of retinal ganglion cells in mammals. These data demonstrate for the first time that Vax2 is both necessary and sufficient for the control of intraretinal RA metabolism, which in turn contributes to the appropriate expression of cone opsins in the vertebrate eye.
- Published
- 2010
- Full Text
- View/download PDF
33. Degeneration of the mouse retina upon dysregulated activity of serum response factor
- Author
-
Jenny, Sandström, Peter, Heiduschka, Susanne C, Beck, Ulrike, Philippar, Matthias W, Seeliger, Ulrich, Schraermeyer, and Alfred, Nordheim
- Subjects
Serum Response Factor ,genetic structures ,Recombinant Fusion Proteins ,Retinal Degeneration ,Gene Dosage ,Gene Expression ,Gene Expression Regulation, Developmental ,Herpes Simplex Virus Protein Vmw65 ,Transfection ,eye diseases ,Mice, Mutant Strains ,Retina ,Disease Models, Animal ,Mice ,embryonic structures ,cardiovascular system ,Electroretinography ,Animals ,Eosine Yellowish-(YS) ,sense organs ,Transgenes ,Hematoxylin ,Embryonic Stem Cells ,Research Article - Abstract
Purpose Our aim was to generate and phenotypically characterize a transgenic mouse line expressing a constitutively active variant of the transcription regulatory protein serum response factor (SRF), namely the SRF-VP16 protein. This new mouse strain has been registered under the designation Gt(ROSA)26Sortm1(SRF-VP16)Antu. We found phenotypic changes upon ectopic expression of SRF-VP16, especially in the mouse retina. Methods Using homologous recombination, we integrated an SRF-VP16 conditional (i.e., “flox-STOP” repressed) expression transgene into the Rosa26 locus of murine embryonic stem (ES) cells. These engineered ES cells were used to derive the Gt(ROSA)26Sortm1(SRF-VP16)Antu mouse strain. Semiquantitative real-time PCR was used to determine expression of the SRF-VP16 transgene at the mRNA level, both in young (P20 and P30) and adult (six months old) Gt(ROSA)26Sortm1(SRF-VP16)Antu mice. We also investigated the transcript levels of endogenous Srf and several SRF target genes. Retinal function was tested by electroretinography in both young and adult mice. Morphological abnormalities could be visualized by hematoxylin and eosin staining of sectioned, paraffin-embedded eye tissue samples. Scanning-laser ophthalmoscopy was used to investigate retinal vascularization and degeneration in adult mice. Results We show that the SRF-VP16 mRNA is expressed to a low but significant degree in the retinas of young and adult animals of the Gt(ROSA)26Sortm1(SRF-VP16)Antu mouse strain, even in the absence of Cre-mediated deletion of the “flox-STOP” cassette. In the retinas of these transgenic mice, endogenous Srf displays elevated transcript levels. Ectopic retinal expression of constitutively active SRF-VP16 is correlated with the malfunction of retinal neurons in both heterozygous and homozygous animals of both age groups (P20 and adult). Additionally, mislamination of retinal cell layers and cellular rosette formations are found in retinas of both heterozygous and homozygous animals of young age. In homozygous individuals, however, the cellular rosettes are more widespread over the fundus. At adult age, retinas both from animals that are heterozygous and homozygous for the floxSTOP/SRF-VP16 transgene display severe degeneration, mainly of the photoreceptor cell layer. Wild-type age-matched littermates, however, do not show any degeneration. The severity of the observed effects correlates with dosage of the transgene. Conclusions This is the first report suggesting an influence of the transcription factor SRF on the development and function of the murine retina. Ectopic SRF-VP16 mRNA expression in the retinas of young animals is correlated with photoreceptor layer mislamination and impaired retinal function. At an advanced age of six months, degenerative processes are detected in SRF-VP16 transgenic retinas accompanied by impaired retinal function. The Gt(ROSA)26Sortm1(SRF-VP16)Antu mouse strain represents a genetic SRF gain-of-function mouse model that will complement the current SRF loss-of-function models. It promises to provide new insight into the hitherto poorly defined role of SRF in retinal development and function, including potential contributions to ophthalmologic disorders. Furthermore, using conditional Cre-mediated activation of SRF-VP16, the described mouse strain will enable assessment of the impact of dysregulated SRF activity on the physiologic functions of various other organs.
- Published
- 2010
34. In vivo assessment of retinal vascular wall dimensions
- Author
-
M. Dominik Fischer, Gesine Huber, Markus N. Preising, Susanne C. Beck, Mathias W. Seeliger, Hans-Peter Hammes, Naoyuki Tanimoto, Christoph Kernstock, Regine Mühlfriedel, Eric Tröger, Birgit Lorenz, and Yuxi Feng
- Subjects
Indocyanine Green ,Pathology ,medicine.medical_specialty ,Retinal Artery ,Green Fluorescent Proteins ,Mice, Transgenic ,Biology ,chemistry.chemical_compound ,Mice ,In vivo ,Arteriole ,medicine.artery ,medicine ,Electroretinography ,Animals ,Humans ,Body Weights and Measures ,Fluorescein Angiography ,Coloring Agents ,Retina ,Venule ,medicine.diagnostic_test ,Lasers ,Retinal ,Anatomy ,Retinal Vein ,Actins ,Scanning laser ophthalmoscopy ,Mice, Inbred C57BL ,Ophthalmoscopy ,medicine.anatomical_structure ,chemistry ,Angiography ,Indocyanine green ,Tomography, Optical Coherence - Abstract
PURPOSE: Retinal blood vessel diameter and arteriovenous ratio (AVR) are commonly used diagnostic parameters. Because vascular walls are typically not visible in funduscopy, clinical AVR estimation is based on the lumen rather than the entire vessel diameter. Here the authors used a transgenic mouse model to quantify AVR in vivo based on total vessel dimensions (wall and lumen). METHODS: Confocal scanning laser ophthalmoscopy (cSLO) and indocyanine green angiography of the retinal vasculature were performed in wild-type and transgenic mice expressing green fluorescent protein (GFP) under the transcriptional control of the smooth muscle type α-actin (αSMA) promoter. Spectral-domain-OCT and ERG were performed to control for integrity of retinal structure and function in vivo and histology to demonstrate the location of GFP expression. RESULTS: Native cSLO imaging and angiography yielded only inner vessel diameters similar to those observed through clinical funduscopy. In αSMA-GFP mice, autofluorescence imaging of the GFP-marked vascular walls also allowed the determination of outer vessel diameters. The mean AVR based on either inner diameter (AVR(id) = 0.72 ± 0.08) or outer diameter (AVR(od) = 0.97 ± 0.09) measurements were significantly different (P < 0.01). CONCLUSIONS: Transgenic αSMA-GFP expression in murine vessel wall components allowed quantification of retinal vessel outer diameters in vivo. Although arterioles and venules differ in lumen and vessel wall width, they share a common outer diameter, leading to an AVR(od) close to unity. Because vessel walls are primary targets in common hypertensive and metabolic diseases, αSMA-GFP transgenic mice may prove valuable in the detailed assessment of such disorders in vivo.
- Published
- 2010
35. Structural and functional phenotyping in the cone-specific photoreceptor function loss 1 (cpfl1) mouse mutant - a model of cone dystrophies
- Author
-
M Dominik, Fischer, Naoyuki, Tanimoto, Susanne C, Beck, Gesine, Huber, Karin, Schaeferhoff, Stylianos, Michalakis, Olaf, Riess, Bernd, Wissinger, Martin, Biel, Michael, Bonin, and Mathias W, Seeliger
- Subjects
Disease Models, Animal ,Mice ,Structure-Activity Relationship ,Imaging, Three-Dimensional ,Phenotype ,Time Factors ,Green Fluorescent Proteins ,Electroretinography ,Animals ,Eye Proteins ,Mice, Mutant Strains ,Retina ,Retinitis Pigmentosa - Abstract
We performed a comprehensive in vivo assessment of retinal morphology and function in cpfl1 (cone photoreceptor function loss 1) mice to better define the disease process in this model of cone dystrophies.Mice were examined using electroretinography (ERG), confocal scanning laser ophthalmoscopy (cSLO), and spectral domain optical coherence tomography (SD-OCT). Cross-breeding cpfl1 mutants with mice expressing green fluorescent protein (GFP) under control of red-green cone opsin promoter allowed for an in vivo timeline analysis of number and distribution of cone photoreceptors using the autofluorescence (AF) mode of the cSLO.Light-evoked responses of cone origin were practically absent in cpfl1 mice, whereas rod system function appeared normal. In vivo imaging revealed a progressive loss of cone photoreceptors with a major decline between PW4 and PW8, while retinal architecture and layering remained essentially intact.While the absence of substantial light-evoked cone responses in the cpfl1 mice is evident from early on, the course of physical cone degeneration is protracted and has a major drop between PW4 and PW8. However, these changes do not lead to significant alterations in retinal architecture, probably due to the relatively low number and wide dissemination of cone photoreceptor cells within the afoveate mouse retina.
- Published
- 2010
36. Induction of STAT3-related genes in fast degenerating cone photoreceptors of cpfl1 mice
- Author
-
M. W. Seeliger, Bernd Wissinger, Karin Schaeferhoff, Stylianos Michalakis, Gesine Huber, Naoyuki Tanimoto, M. D. Fischer, Olaf Riess, Susanne C. Beck, Elvir Becirovic, N. Rieger, Michael Bonin, and Martin Biel
- Subjects
Retinal degeneration ,STAT3 Transcription Factor ,Pathology ,medicine.medical_specialty ,genetic structures ,Mutant ,Degeneration (medical) ,Biology ,medicine.disease_cause ,Cellular and Molecular Neuroscience ,Mice ,medicine ,Animals ,STAT3 ,Molecular Biology ,Loss function ,Oligonucleotide Array Sequence Analysis ,Pharmacology ,Mutation ,medicine.diagnostic_test ,Retinal Degeneration ,Cell Biology ,medicine.disease ,Mice, Mutant Strains ,Cell biology ,Gliosis ,Gene Expression Regulation ,biology.protein ,Retinal Cone Photoreceptor Cells ,Molecular Medicine ,sense organs ,medicine.symptom ,Electroretinography - Abstract
Cone dystrophies are genetic diseases characterized by loss of cone photoreceptor function and severe impairment of daylight vision. Loss of function is accompanied by a progressive degeneration of cones limiting potential therapeutic interventions. In this study we combined microarray-based gene-expression analysis with electroretinography and immunohistochemistry to characterize the pathological processes in the cone photoreceptor function loss 1 (cpfl1) mouse model. The cpfl1-mouse is a naturally arising mouse mutant with a loss-of-function mutation in the cone-specific Pde6c gene. Cpfl1-mice displayed normal rod-specific light responses while cone-specific responses were strongly diminished. Despite the lack of a general retinal degeneration, the cone-specific functional defect resulted in a marked activation of GFAP, a hallmark of Muller-cell gliosis. Microarray-based network-analysis confirmed activation of Muller-glia-specific transcripts. Unexpectedly, we found up-regulation of the cytokine LIF and the anti-apoptotic transcription factor STAT3 in cpfl1 cone photoreceptors. We postulate that STAT3-related pathways are induced in cpfl1 cone photoreceptors to counteract degeneration.
- Published
- 2010
37. Study of gene-targeted mouse models of splicing factor gene Prpf31 implicated in human autosomal dominant retinitis pigmentosa (RP)
- Author
-
Theo van Veen, Per Ekström, Naoyuki Tanimoto, Marian M. Humphries, Cécilia Maubaret, Susanne C. Beck, Olga V. Makarova, Evgeny M. Makarov, Shomi S. Bhattacharya, Christina Chakarova, Thierry Léveillard, E. Fahl, François Paquet-Durand, Peter Humphries, Kinga M. Bujakowska, Paul F. Kenna, and Mathias W. Seeliger
- Subjects
Retinal degeneration ,PRPF31 ,Genotype ,Usher syndrome ,Blotting, Western ,Biology ,Retina ,Gene Knockout Techniques ,Mice ,Retinitis pigmentosa ,medicine ,Electroretinography ,In Situ Nick-End Labeling ,Animals ,Humans ,Point Mutation ,Gene Knock-In Techniques ,Allele ,Eye Proteins ,Genes, Dominant ,Genetics ,Mice, Knockout ,Mice, Inbred BALB C ,Gene targeting ,medicine.disease ,Molecular biology ,Penetrance ,eye diseases ,Mice, Inbred C57BL ,Ophthalmoscopy ,Disease Models, Animal ,Knockout mouse ,Gene Targeting ,Electrophoresis, Polyacrylamide Gel ,Female ,Retinitis Pigmentosa - Abstract
PURPOSE. Pre-mRNA processing factor 31 (PRPF31) is a ubiquitous protein needed for the assembly of the pre-mRNA splicing machinery. It has been shown that mutations in this gene cause autosomal dominant retinitis pigmentosa 11 (RP11), which is characterized by rod-cell degeneration. Interestingly, mutations in this ubiquitously expressed gene do not lead to phenotypes other than retinal malfunction. Furthermore, the dominant inheritance pattern has shown incomplete penetrance, which poses interesting questions about the disease mechanism of RP11. METHODS. To characterize PRPF31 function in the rod cells, two animal models have been generated. One was a heterozygous knock-in mouse (Prpf31(A216P/+)) carrying a point mutation p.A216P, which has previously been identified in RP11 patients. The second was a heterozygous knockout mouse (Prpf31(+/-)). Retinal degeneration in RP11 mouse models was monitored by electroretinography and histology. RESULTS. Generation of the mouse models is presented, as are results of ERGs and retinal morphology. No degenerative phenotype on fundus examination was found in Prpf31(A216P/+) and Prpf31(+/-) mice. Prpf31(A216P/A216P) and Prpf31(-/-) genotypes were embryonic lethal. CONCLUSIONS. The results imply that Prpf31 is necessary for survival, and there is no compensation mechanism in mouse for the lack of this splicing factor. The authors suggest that p.A216P mutation in Prpf31 does not exert a dominant negative effect and that one Prpf31 wild-type allele is sufficient for maintenance of the healthy retina in mice. (Less)
- Published
- 2009
38. In conditions of limited chromophore supply rods entrap 11-cis-retinal leading to loss of cone function and cell death
- Author
-
Marijana Samardzija, Yvan Arsenijevic, Andreas Wenzel, Vitus Oberhauser, Corinne Kostic, E. Fahl, Christian Grimm, Sandrine Joly, Markus Thiersch, Naoyuki Tanimoto, Mathias W. Seeliger, Johannes von Lintig, Susanne C. Beck, University of Zurich, and Samardzija, M
- Subjects
10018 Ophthalmology Clinic ,cis-trans-Isomerases ,2716 Genetics (clinical) ,Opsin ,11-cis retinal ,genetic structures ,610 Medicine & health ,Biology ,Retinal Cone Photoreceptor Cells ,Mice ,1311 Genetics ,Retinal Rod Photoreceptor Cells ,1312 Molecular Biology ,Genetics ,Animals ,Humans ,Eye Proteins ,Molecular Biology ,Genetics (clinical) ,Amino Acid Substitution ,Carrier Proteins/genetics ,Cell Death ,Cone Opsins/metabolism ,Eye Proteins/genetics ,Mutation/genetics ,Protein Transport ,Retinal Cone Photoreceptor Cells/cytology ,Retinal Cone Photoreceptor Cells/metabolism ,Retinal Pigments/metabolism ,Retinal Rod Photoreceptor Cells/metabolism ,Retinaldehyde/metabolism ,General Medicine ,Anatomy ,Cone Opsins ,eye diseases ,RPE65 ,Rhodopsin ,Cis-trans-Isomerases ,Retinaldehyde ,Mutation ,Biophysics ,biology.protein ,sense organs ,Carrier Proteins ,Retinal Pigments - Abstract
RPE65 is a retinoid isomerase required for the production of 11-cis-retinal, the chromophore of both cone and rod visual pigments. We recently established an R91W knock-in mouse strain as homologous animal model for patients afflicted by this mutation in RPE65. These mice have impaired vision and can only synthesize minute amounts of 11-cis-retinal. Here, we investigated the consequences of this chromophore insufficiency on cone function and pathophysiology. We found that the R91W mutation caused cone opsin mislocalization and progressive geographic cone atrophy. Remnant visual function was mostly mediated by rods. Ablation of rod opsin corrected the localization of cone opsin and improved cone retinal function. Thus, our analyses indicate that under conditions of limited chromophore supply rods and cones compete for 11-cis-retinal that derives from regeneration pathway(s) which are reliant on RPE65. Due to their higher number and the instability of cone opsin, rods are privileged under this condition while cones suffer chromophore deficiency and degenerate. These findings reinforce the notion that in patients any effective gene therapy with RPE65 needs to target the cone-rich macula directly to locally restore the cones' chromophore supply outside the reach of rods.
- Published
- 2009
39. A portable albumin binder from a DNA-encoded chemical library
- Author
-
Dario Neri, Frank Bootz, Samu Melkko, Christof Baltes, Christoph E. Dumelin, Jörg Scheuermann, Yixin Zhang, Mathias W. Seeliger, Markus Rudin, Eveline Trachsel, Susanne C. Beck, Luca Mannocci, Felicitas Kranz, Sabrina Trüssel, Thomas Müggler, Mihaela Drumea-Mirancea, Fabian Buller, and University of Zurich
- Subjects
1503 Catalysis ,Stereochemistry ,Oligonucleotides ,Protein Array Analysis ,Contrast Media ,610 Medicine & health ,1600 General Chemistry ,Catalysis ,Chemical library ,Small Molecule Libraries ,170 Ethics ,Mice ,Structure-Activity Relationship ,chemistry.chemical_compound ,Animals ,Humans ,10237 Institute of Biomedical Engineering ,A-DNA ,Organic Chemicals ,Serum Albumin ,Chemistry ,Albumin ,General Chemistry ,General Medicine ,Combinatorial chemistry ,Protein Binding - Published
- 2008
- Full Text
- View/download PDF
40. Crb1 is a determinant of retinal apical Müller glia cell features
- Author
-
Alicia Sanz Sanz, Jan Klooster, Inge Versteeg, Wendy M. Aartsen, Mathias W. Seeliger, Jan Wijnholds, Rogier M. Vos, Serge A. van de Pavert, and Susanne C. Beck
- Subjects
Retinal degeneration ,Aging ,Light ,Nerve Tissue Proteins ,Optic Atrophy, Hereditary, Leber ,Biology ,Retina ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,Microscopy, Electron, Transmission ,Retinitis pigmentosa ,medicine ,Animals ,Genetic Predisposition to Disease ,Photoreceptor Cells ,Retinal regeneration ,Mice, Knockout ,Retinal pigment epithelium ,Gene therapy of the human retina ,Microvilli ,Neovascularization, Pathologic ,Gene Expression Profiling ,Intrinsically photosensitive retinal ganglion cells ,Retinal Degeneration ,Retinal ,Anatomy ,medicine.disease ,eye diseases ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Neurology ,chemistry ,Gene Expression Regulation ,sense organs ,Neuroglia ,Photic Stimulation ,Retinitis Pigmentosa - Abstract
Mutations in the human Crumbs homologue-1 (CRB1) gene cause retinal blinding diseases, such as Leber congenital amaurosis and retinitis pigmentosa. In the previous studies we have shown that Crb1 resides in retinal Muller glia cells and that loss of Crb1 results in retinal degeneration (particularly in the inferior temporal quadrant of the mouse eye). Degeneration is increased by exposure to white light. Here, we studied the role of light and aging to gain a better understanding of the factors involved in the progress of retinal disease. Our data reveal that light is neither sufficient nor required to induce retinal disorganization and degeneration in young Crb1(-/-) mutant mice, suggesting that it rather modulates the retinal phenotype. Gene expression profiling showed that expression of five genes is altered in light-exposed Crb1(-/-) mutant retinas. Three of the five genes are involved in chromosome stabilization (Pituitary tumor transforming gene 1 or Pttg1, Establishment of cohesion 1 homolog 1 or Esco1, and a gene similar to histone H2B). In aged retinas, degeneration of photoreceptors, inner retinal neurons, and retinal pigment epithelium was practically limited to the inferior temporal quadrant. Loss of Crb1 in Muller glia cells resulted in an irregular number and size of their apical villi. We propose that Crb1 is required to regulate number and size of these Muller glia cell villi. The subsequent loss of retinal integrity resulted in neovascularization, in which blood vessels of the choroid protruded into the neural retina.
- Published
- 2007
41. No evidence for prooxidative effects of homocysteine in vascular endothelial cells
- Author
-
Juergen Frank, Hans Konrad Biesalski, Susanne C. Beck, and Andrea Flaccus
- Subjects
medicine.medical_specialty ,Time Factors ,Medicine (miscellaneous) ,Vascular Cell Adhesion Molecule-1 ,Inflammation ,Biology ,medicine.disease_cause ,Thiobarbituric Acid Reactive Substances ,Monocytes ,Proinflammatory cytokine ,Cell Line ,Internal medicine ,medicine ,Cell Adhesion ,Humans ,Endothelial dysfunction ,Homocysteine ,chemistry.chemical_classification ,Reactive oxygen species ,Nutrition and Dietetics ,Dose-Response Relationship, Drug ,Cell adhesion molecule ,Monocyte ,Endothelial Cells ,medicine.disease ,Intercellular Adhesion Molecule-1 ,Cell biology ,Endothelial stem cell ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Endothelium, Vascular ,medicine.symptom ,E-Selectin ,Reactive Oxygen Species ,Oxidative stress - Abstract
Many epidemiological studies predict a role for homocysteine (HCys) in cardiovascular disease occurrence, progression, and risk factors. In vitro studies demonstrated that HCys is an atherogenic determinant that promotes oxidant stress, inflammation, endothelial dysfunction and cell proliferation. This study originally attempted to examine the mechanism by which exposure of endothelial cells to HCys (0-250 microM) initiates inflammatory reaction and oxidative stress, by (i) investigating whether physiological and pathophysiological concentrations of HCys exhibit a prooxidative activity in vitro, (ii) examining the interaction of monocyte adhesion (Mono Mac 6) to monolayers of human microvascular endothelial cells (HMEC-1) exposed to different HCys concentrations, and (iii) examining if adherent monocytes increase reactive oxygen species either in endothelial cells or in monocytes themselves. However, our results demonstrate that HCys had neither prooxidative nor cytotoxic effects on endothelial cells. Only a moderate time- and concentration-dependent increase in monocyte adhesion up to 28.3 +/- 5.5% was achieved relative to control after 4 h of HCys stimulation. This effect was accompanied by an increased VCAM and ICAM-1 mRNA expression. This "proinflammatory" effect appeared also when HMEC-1 cells were incubated with cysteine or glutathione at the concentration range 0-250 microM, demonstrating a non-specific rather than a specific HCys effect. In addition, adherent monocytes did not increase ROS formation neither in endothelial cells nor in monocytes themselves, indicating no direct or indirect cytotoxic or prooxidative effects of HCys.
- Published
- 2006
42. Biochemical but not clinical vitamin A deficiency results from mutations in the gene for retinol binding protein
- Author
-
Jürgen Frank, Susanne C. Beck, Harald Gollnick, Ram Reifen, Hans Konrad Biesalski, Mathias W. Seeliger, Beate Illek, Felix Oliver Heinrich, Bernd Wissinger, and Eberhart Zrenner
- Subjects
medicine.medical_specialty ,Retinyl Esters ,Adolescent ,Molecular Sequence Data ,Retinoic acid ,Medicine (miscellaneous) ,medicine.disease_cause ,Nuclear Family ,chemistry.chemical_compound ,Exon ,Internal medicine ,medicine ,Humans ,Point Mutation ,Prealbumin ,Vitamin A ,Mutation ,Nutrition and Dietetics ,biology ,Base Sequence ,Vitamin A Deficiency ,Point mutation ,Retinol ,Exons ,medicine.disease ,Vitamin A deficiency ,Retinol-Binding Proteins ,Transthyretin ,Retinol binding protein ,Zinc ,Endocrinology ,chemistry ,biology.protein ,Female ,Diterpenes ,Retinol-Binding Proteins, Plasma - Abstract
Background: Two German sisters aged 14 and 17 y were admitted to the Tubingen eye hospital with a history of night blindness. In both siblings, plasma retinol binding protein (RBP) concentrations were below the limit of detection (
- Published
- 1999
43. Novel Rodent Models for Macular Research
- Author
-
Naoyuki Tanimoto, Gesine Huber, Susanne C. Beck, Regine Muehlfriedel, Leo Peichl, Coni Imsand, Yuxi Feng, Mathias W. Seeliger, Hans-Peter Hammes, Christian Grimm, Franziska vom Hagen, Severin R. Heynen, and University of Zurich
- Subjects
10018 Ophthalmology Clinic ,medicine.medical_specialty ,Retinal Disorder ,genetic structures ,Rodent ,lcsh:Medicine ,Rodentia ,610 Medicine & health ,1100 General Agricultural and Biological Sciences ,Central region ,Ophthalmology/Diabetic Retinopathy ,03 medical and health sciences ,0302 clinical medicine ,Macula Lutea ,1300 General Biochemistry, Genetics and Molecular Biology ,ddc:570 ,biology.animal ,Ophthalmology ,Animals ,Medicine ,lcsh:Science ,030304 developmental biology ,1000 Multidisciplinary ,0303 health sciences ,Retina ,Multidisciplinary ,biology ,business.industry ,lcsh:R ,Retinal Vessels ,Ophthalmology/Macular Disorders ,eye diseases ,Disease Models, Animal ,Ophthalmology clinic ,medicine.anatomical_structure ,030221 ophthalmology & optometry ,Ophthalmology/Retinal Disorders ,lcsh:Q ,sense organs ,business ,Neuroscience ,Tomography, Optical Coherence ,Research Article - Abstract
Background: Many disabling human retinal disorders involve the central retina, particularly the macula. However, the commonly used rodent models in research, mouse and rat, do not possess a macula. The purpose of this study was to identify small laboratory rodents with a significant central region as potential new models for macular research. Methodology/Principal Findings: Gerbillus perpallidus, Meriones unguiculatus and Phodopus campbelli, laboratory rodents less commonly used in retinal research, were subjected to confocal scanning laser ophthalmoscopy (cSLO), fluorescein and indocyanine green angiography, and spectral-domain optical coherence tomography (SD-OCT) using standard equipment (Heidelberg Engineering HRA1 and Spectralis™) adapted to small rodent eyes. The existence of a visual streak-like pattern was assessed on the basis of vascular topography, retinal thickness, and the topography of retinal ganglion cells and cone photoreceptors. All three species examined showed evidence of a significant horizontal streak-like specialization. cSLO angiography and retinal wholemounts revealed that superficial retinal blood vessels typically ramify and narrow into a sparse capillary net at the border of the respective area located dorsal to the optic nerve. Similar to the macular region, there was an absence of larger blood vessels in the streak region. Furthermore, the thickness of the photoreceptor layer and the population density of neurons in the ganglion cell layer were markedly increased in the visual streak region. Conclusions/Significance: The retinal specializations of Gerbillus perpallidus, Meriones unguiculatus and Phodopus campbelli resemble features of the primate macula. Hence, the rodents reported here may serve to study aspects of macular development and diseases like age-related macular degeneration and diabetic macular edema, and the preclinical assessment of therapeutic strategies.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.