1
|
Doldur-Balli F, Smieszek SP, Keenan BT, Zimmerman AJ, Veatch OJ, Polymeropoulos CM, Birznieks G, Polymeropoulos MH. Screening effects of HCN channel blockers on sleep/wake behavior in zebrafish. Front Neurosci 2024; 18:1375484. [PMID: 38567282 PMCID: PMC10986788 DOI: 10.3389/fnins.2024.1375484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels generate electrical rhythmicity in various tissues although primarily heart, retina and brain. The HCN channel blocker compound, Ivabradine (Corlanor), is approved by the US Food and Drug Administration (FDA) as a medication to lower heart rate by blocking hyperpolarization activated inward current in the sinoatrial node. In addition, a growing body of evidence suggests a role for HCN channels in regulation of sleep/wake behavior. Zebrafish larvae are ideal model organisms for high throughput drug screening, drug repurposing and behavioral phenotyping studies. We leveraged this model system to investigate effects of three HCN channel blockers (Ivabradine, Zatebradine Hydrochloride and ZD7288) at multiple doses on sleep/wake behavior in wild type zebrafish. Results of interest included shorter latency to daytime sleep at 0.1 μM dose of Ivabradine (ANOVA, p: 0.02), moderate reduction in average activity at 30 μM dose of Zatebradine Hydrochloride (ANOVA, p: 0.024) in daytime, and increased nighttime sleep at 4.5 μM dose of ZD7288 (ANOVA, p: 0.036). Taken together, shorter latency to daytime sleep, decrease in daytime activity and increased nighttime sleep indicate that different HCN channel antagonists affected different parameters of sleep and activity.
Collapse
Affiliation(s)
- Fusun Doldur-Balli
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Brendan T. Keenan
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Amber J. Zimmerman
- Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Olivia J. Veatch
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS, United States
| | | | - Gunther Birznieks
- Vanda Pharmaceuticals Inc., Pennsylvania, Washington, DC, United States
| | | |
Collapse
|
2
|
Kim D, Roh H, Lee HM, Kim SJ, Im M. Localization of hyperpolarization-activated cyclic nucleotide-gated channels in the vertebrate retinas across species and their physiological roles. Front Neuroanat 2024; 18:1385932. [PMID: 38562955 PMCID: PMC10982330 DOI: 10.3389/fnana.2024.1385932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Transmembrane proteins known as hyperpolarization-activated cyclic nucleotide-gated (HCN) channels control the movement of Na+ and K+ ions across cellular membranes. HCN channels are known to be involved in crucial physiological functions in regulating neuronal excitability and rhythmicity, and pacemaker activity in the heart. Although HCN channels have been relatively well investigated in the brain, their distribution and function in the retina have received less attention, remaining their physiological roles to be comprehensively understood. Also, because recent studies reported HCN channels have been somewhat linked with the dysfunction of photoreceptors which are affected by retinal diseases, investigating HCN channels in the retina may offer valuable insights into disease mechanisms and potentially contribute to identifying novel therapeutic targets for retinal degenerative disorders. This paper endeavors to summarize the existing literature on the distribution and function of HCN channels reported in the vertebrate retinas of various species and discuss the potential implications for the treatment of retinal diseases. Then, we recapitulate current knowledge regarding the function and regulation of HCN channels, as well as their relevance to various neurological disorders.
Collapse
Affiliation(s)
- Daniel Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University (SNU), Seoul, Republic of Korea
| | - Hyeonhee Roh
- School of Electrical Engineering, College of Engineering, Korea University, Seoul, Republic of Korea
| | - Hyung-Min Lee
- School of Electrical Engineering, College of Engineering, Korea University, Seoul, Republic of Korea
| | - Sang Jeong Kim
- Department of Biomedical Sciences, College of Medicine, Seoul National University (SNU), Seoul, Republic of Korea
| | - Maesoon Im
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science & Technology (UST), Seoul, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Occelli LM, Zobel L, Stoddard J, Wagner J, Pasmanter N, Querubin J, Renner LM, Reynaga R, Winkler PA, Sun K, Marinho LFLP, O'Riordan CR, Frederick A, Lauer A, Tsang SH, Hauswirth WW, McGill TJ, Neuringer M, Michalakis S, Petersen-Jones SM. Development of a translatable gene augmentation therapy for CNGB1-retinitis pigmentosa. Mol Ther 2023; 31:2028-2041. [PMID: 37056049 PMCID: PMC10362398 DOI: 10.1016/j.ymthe.2023.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023] Open
Abstract
In this study, we investigate a gene augmentation therapy candidate for the treatment of retinitis pigmentosa (RP) due to cyclic nucleotide-gated channel beta 1 (CNGB1) mutations. We use an adeno-associated virus serotype 5 with transgene under control of a novel short human rhodopsin promoter. The promoter/capsid combination drives efficient expression of a reporter gene (AAV5-RHO-eGFP) exclusively in rod photoreceptors in primate, dog, and mouse following subretinal delivery. The therapeutic vector (AAV5-RHO-CNGB1) delivered to the subretinal space of CNGB1 mutant dogs restores rod-mediated retinal function (electroretinographic responses and vision) for at least 12 months post treatment. Immunohistochemistry shows human CNGB1 is expressed in rod photoreceptors in the treated regions as well as restoration of expression and trafficking of the endogenous alpha subunit of the rod CNG channel required for normal channel formation. The treatment reverses abnormal accumulation of the second messenger, cyclic guanosine monophosphate, which occurs in rod photoreceptors of CNGB1 mutant dogs, confirming formation of a functional CNG channel. In vivo imaging shows long-term preservation of retinal structure. In conclusion, this study establishes the long-term efficacy of subretinal delivery of AAV5-RHO-CNGB1 to rescue the disease phenotype in a canine model of CNGB1-RP, confirming its suitability for future clinical development.
Collapse
Affiliation(s)
- Laurence M Occelli
- College of Veterinary Medicine, Michigan State University, 736 Wilson Road, East Lansing, MI 48864, USA
| | - Lena Zobel
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Jonathan Stoddard
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185(th) Avenue, Beaverton, OR 97005, USA
| | - Johanna Wagner
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Nathaniel Pasmanter
- College of Veterinary Medicine, Michigan State University, 736 Wilson Road, East Lansing, MI 48864, USA
| | - Janice Querubin
- College of Veterinary Medicine, Michigan State University, 736 Wilson Road, East Lansing, MI 48864, USA
| | - Lauren M Renner
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185(th) Avenue, Beaverton, OR 97005, USA
| | - Rene Reynaga
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185(th) Avenue, Beaverton, OR 97005, USA
| | - Paige A Winkler
- College of Veterinary Medicine, Michigan State University, 736 Wilson Road, East Lansing, MI 48864, USA
| | - Kelian Sun
- College of Veterinary Medicine, Michigan State University, 736 Wilson Road, East Lansing, MI 48864, USA
| | - Luis Felipe L P Marinho
- College of Veterinary Medicine, Michigan State University, 736 Wilson Road, East Lansing, MI 48864, USA
| | | | - Amy Frederick
- Genomic Medicine Unit, Sanofi, 225 Second Avenue, Waltham, MA 02451, USA
| | - Andreas Lauer
- Casey Eye Institute, Oregon Health & Science University, 515 Campus Drive, Portland, OR 97239, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Columbia Stem Cell Initiative, Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - William W Hauswirth
- Department of Ophthalmology, College of Medicine, University of Florida, Box 100284 HSC, Gainesville, FL 32610, USA
| | - Trevor J McGill
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185(th) Avenue, Beaverton, OR 97005, USA; Casey Eye Institute, Oregon Health & Science University, 515 Campus Drive, Portland, OR 97239, USA
| | - Martha Neuringer
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185(th) Avenue, Beaverton, OR 97005, USA; Casey Eye Institute, Oregon Health & Science University, 515 Campus Drive, Portland, OR 97239, USA
| | - Stylianos Michalakis
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; Department of Ophthalmology, University Hospital, LMU Munich, 80336 Munich, Germany.
| | - Simon M Petersen-Jones
- College of Veterinary Medicine, Michigan State University, 736 Wilson Road, East Lansing, MI 48864, USA.
| |
Collapse
|
4
|
Hebbar S, Traikov S, Hälsig C, Knust E. Modulating the Kynurenine pathway or sequestering toxic 3-hydroxykynurenine protects the retina from light-induced damage in Drosophila. PLoS Genet 2023; 19:e1010644. [PMID: 36952572 PMCID: PMC10035932 DOI: 10.1371/journal.pgen.1010644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/30/2023] [Indexed: 03/25/2023] Open
Abstract
Tissue health is regulated by a myriad of exogenous or endogenous factors. Here we investigated the role of the conserved Kynurenine pathway (KP) in maintaining retinal homeostasis in the context of light stress in Drosophila melanogaster. cinnabar, cardinal and scarlet are fly genes that encode different steps in the KP. Along with white, these genes are known regulators of brown pigment (ommochrome) biosynthesis. Using white as a sensitized genetic background, we show that mutations in cinnabar, cardinal and scarlet differentially modulate light-induced retinal damage. Mass Spectrometric measurements of KP metabolites in flies with different genetic combinations support the notion that increased levels of 3-hydroxykynurenine (3OH-K) and Xanthurenic acid (XA) enhance retinal damage, whereas Kynurenic Acid (KYNA) and Kynurenine (K) are neuro-protective. This conclusion was corroborated by showing that feeding 3OH-K results in enhanced retinal damage, whereas feeding KYNA protects the retina in sensitized genetic backgrounds. Interestingly, the harmful effects of free 3OH-K are diminished by its sub-cellular compartmentalization. Sequestering of 3OH-K enables the quenching of its toxicity through conversion to brown pigment or conjugation to proteins. This work enabled us to decouple the role of these KP genes in ommochrome formation from their role in retinal homeostasis. Additionally, it puts forward new hypotheses on the importance of the balance of KP metabolites and their compartmentalization in disease alleviation.
Collapse
Affiliation(s)
- Sarita Hebbar
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sofia Traikov
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Catrin Hälsig
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
5
|
Inamdar SM, Lankford CK, Baker SA. Photoreceptor Ion Channels in Signaling and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:269-276. [PMID: 37440044 DOI: 10.1007/978-3-031-27681-1_39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Photoreceptors (PRs) in the neural retina convert photon capture into an electrical signal that is communicated across a chemical synapse to second-order neurons in the retina and on through the rest of the visual pathway. This information is decoded in the visual cortex to create images. The activity of PRs depends on the concerted action of several voltage-gated ion channels that will be discussed in this chapter.
Collapse
Affiliation(s)
- Shivangi M Inamdar
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA, USA.
| | - Colten K Lankford
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA, USA
| | - Sheila A Baker
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
6
|
Chrystal PW, Lambacher NJ, Doucette LP, Bellingham J, Schiff ER, Noel NCL, Li C, Tsiropoulou S, Casey GA, Zhai Y, Nadolski NJ, Majumder MH, Tagoe J, D'Esposito F, Cordeiro MF, Downes S, Clayton-Smith J, Ellingford J, Mahroo OA, Hocking JC, Cheetham ME, Webster AR, Jansen G, Blacque OE, Allison WT, Au PYB, MacDonald IM, Arno G, Leroux MR. The inner junction protein CFAP20 functions in motile and non-motile cilia and is critical for vision. Nat Commun 2022; 13:6595. [PMID: 36329026 PMCID: PMC9633640 DOI: 10.1038/s41467-022-33820-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
Motile and non-motile cilia are associated with mutually-exclusive genetic disorders. Motile cilia propel sperm or extracellular fluids, and their dysfunction causes primary ciliary dyskinesia. Non-motile cilia serve as sensory/signalling antennae on most cell types, and their disruption causes single-organ ciliopathies such as retinopathies or multi-system syndromes. CFAP20 is a ciliopathy candidate known to modulate motile cilia in unicellular eukaryotes. We demonstrate that in zebrafish, cfap20 is required for motile cilia function, and in C. elegans, CFAP-20 maintains the structural integrity of non-motile cilia inner junctions, influencing sensory-dependent signalling and development. Human patients and zebrafish with CFAP20 mutations both exhibit retinal dystrophy. Hence, CFAP20 functions within a structural/functional hub centered on the inner junction that is shared between motile and non-motile cilia, and is distinct from other ciliopathy-associated domains or macromolecular complexes. Our findings suggest an uncharacterised pathomechanism for retinal dystrophy, and potentially for motile and non-motile ciliopathies in general.
Collapse
Affiliation(s)
- Paul W Chrystal
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada.
| | - Nils J Lambacher
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Lance P Doucette
- Department of Ophthalmology & Visual Science, University of Alberta, Edmonton, AB, Canada
| | | | - Elena R Schiff
- Moorfields Eye Hospital, London, UK
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Nicole C L Noel
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Chunmei Li
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Sofia Tsiropoulou
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Geoffrey A Casey
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Yi Zhai
- Department of Ophthalmology & Visual Science, University of Alberta, Edmonton, AB, Canada
| | - Nathan J Nadolski
- Division of Anatomy, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Mohammed H Majumder
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Julia Tagoe
- Lethbridge Outreach Genetics Service, Alberta Health Services, Lethbridge, AB, Canada
| | - Fabiana D'Esposito
- Western Eye Hospital, Imperial College Healthcare NHS Trust, London, UK
- ICORG, Imperial College London, London, UK
| | | | - Susan Downes
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jill Clayton-Smith
- Manchester Centre for Genomic Medicine, Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
| | - Jamie Ellingford
- Manchester Centre for Genomic Medicine, Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
- Genomics England, London, UK
| | - Omar A Mahroo
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, London, UK
| | - Jennifer C Hocking
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Division of Anatomy, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Andrew R Webster
- UCL Institute of Ophthalmology, London, UK
- Moorfields Eye Hospital, London, UK
| | - Gert Jansen
- Department of Cell Biology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - W Ted Allison
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada.
| | - Ping Yee Billie Au
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Ian M MacDonald
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada.
- Department of Ophthalmology & Visual Science, University of Alberta, Edmonton, AB, Canada.
| | - Gavin Arno
- UCL Institute of Ophthalmology, London, UK.
- Moorfields Eye Hospital, London, UK.
- North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| | - Michel R Leroux
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada.
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
7
|
Mutations within the cGMP-binding domain of CNGA1 causing autosomal recessive retinitis pigmentosa in human and animal model. Cell Death Dis 2022; 8:387. [PMID: 36115851 PMCID: PMC9482621 DOI: 10.1038/s41420-022-01185-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/08/2022]
Abstract
Retinitis pigmentosa is a group of progressive inherited retinal dystrophies that may present clinically as part of a syndromic entity or as an isolated (nonsyndromic) manifestation. In an Indian family suffering from retinitis pigmentosa, we identified a missense variation in CNGA1 affecting the cyclic nucleotide binding domain (CNBD) and characterized a mouse model developed with mutated CNBD. A gene panel analysis comprising 105 known RP genes was used to analyze a family with autosomal-recessive retinitis pigmentosa (arRP) and revealed that CNGA1 was affected. From sperm samples of ENU mutagenesis derived F1 mice, we re-derived a mutant with a Cnga1 mutation. Homozygous mutant mice, developing retinal degeneration, were examined for morphological and functional consequences of the mutation. In the family, we identified a rare CNGA1 variant (NM_001379270.1) c.1525 G > A; (p.Gly509Arg), which co-segregated among the affected family members. Homozygous Cnga1 mice harboring a (ENSMUST00000087213.12) c.1526 A > G (p.Tyr509Cys) mutation showed progressive degeneration in the retinal photoreceptors from 8 weeks on. This study supports a role for CNGA1 as a disease gene for arRP and provides new insights on the pathobiology of cGMP-binding domain mutations in CNGA1-RP.
Collapse
|
8
|
Yan J, Günter A, Das S, Mühlfriedel R, Michalakis S, Jiao K, Seeliger MW, Paquet-Durand F. Inherited Retinal Degeneration: PARP-Dependent Activation of Calpain Requires CNG Channel Activity. Biomolecules 2022; 12:biom12030455. [PMID: 35327647 PMCID: PMC8946186 DOI: 10.3390/biom12030455] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 01/27/2023] Open
Abstract
Inherited retinal degenerations (IRDs) are a group of blinding diseases, typically involving a progressive loss of photoreceptors. The IRD pathology is often based on an accumulation of cGMP in photoreceptors and associated with the excessive activation of calpain and poly (ADP-ribose) polymerase (PARP). Inhibitors of calpain or PARP have shown promise in preventing photoreceptor cell death, yet the relationship between these enzymes remains unclear. To explore this further, organotypic retinal explant cultures derived from wild-type and IRD-mutant mice were treated with inhibitors specific for calpain, PARP, and voltage-gated Ca2+ channels (VGCCs). The outcomes were assessed using in situ activity assays for calpain and PARP and immunostaining for activated calpain-2, poly (ADP-ribose), and cGMP, as well as the TUNEL assay for cell death detection. The IRD models included the Pde6b-mutant rd1 mouse and rd1*Cngb1−/− double-mutant mice, which lack the beta subunit of the rod cyclic nucleotide-gated (CNG) channel and are partially protected from rd1 degeneration. We confirmed that an inhibition of either calpain or PARP reduces photoreceptor cell death in rd1 retina. However, while the activity of calpain was decreased by the inhibition of PARP, calpain inhibition did not alter the PARP activity. A combination treatment with calpain and PARP inhibitors did not synergistically reduce cell death. In the slow degeneration of rd1*Cngb1−/− double mutant, VGCC inhibition delayed photoreceptor cell death, while PARP inhibition did not. Our results indicate that PARP acts upstream of calpain and that both are part of the same degenerative pathway in Pde6b-dependent photoreceptor degeneration. While PARP activation may be associated with CNG channel activity, calpain activation is linked to VGCC opening. Overall, our data highlights PARP as a target for therapeutic interventions in IRD-type diseases.
Collapse
Affiliation(s)
- Jie Yan
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (J.Y.); (S.D.)
- Graduate Training Centre of Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Alexander Günter
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (A.G.); (R.M.)
| | - Soumyaparna Das
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (J.Y.); (S.D.)
| | - Regine Mühlfriedel
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (A.G.); (R.M.)
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, 80539 München, Germany;
| | - Kangwei Jiao
- Key Laboratory of Yunnan Province, Affiliated Hospital of Yunnan University, Kunming 650051, China;
| | - Mathias W. Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (A.G.); (R.M.)
- Correspondence: (M.W.S.); (F.P.-D.)
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, 72076 Tübingen, Germany; (J.Y.); (S.D.)
- Correspondence: (M.W.S.); (F.P.-D.)
| |
Collapse
|
9
|
Wagner JE, Zobel L, Gerhardt MJ, O'Riordan CR, Frederick A, Petersen-Jones SM, Biel M, Michalakis S. In vivo potency testing of subretinal rAAV5.hCNGB1 gene therapy in the Cngb1 knockout mouse model of retinitis pigmentosa. Hum Gene Ther 2021; 32:1158-1170. [PMID: 34376057 DOI: 10.1089/hum.2021.121] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Retinitis pigmentosa type 45 (RP45) is an autosomal-recessively inherited blinding disease caused by mutations in the cyclic nucleotide gated channel subunit beta 1 (CNGB1) gene. In this study, we developed and tested a novel gene supplementation therapy suitable for clinical translation. To this end, we designed a recombinant adeno-associated virus (rAAV) vector carrying a genome that features a novel human rhodopsin promoter (hRHO194) driving rod-specific expression of full-length human CNGB1 (rAAV5.hCNGB1). rAAV5.hCNGB1 was evaluated for efficacy in the Cngb1 knockout (Cngb1-/-) mouse model of RP45. In particular, increasing doses of rAAV5.hCNGB1 were delivered via single subretinal injection in 4-week-old Cngb1-/- mice and the treatment effect was assessed over a follow-up period of 9 months at the level of (i) retinal morphology, (ii) retinal function, (iii) vision-guided behavior, and (iv) transgene expression. We found that subretinal treatment with rAAV5.hCNGB1 resulted in efficient expression of the human CNGB1 protein in mouse rods and was able to normalize the expression of the endogenous mouse CNGA1 subunit, which together with CNGB1 forms the native heterotetrameric cGMP-gated cation channel in rod photoreceptors. The treatment led to a dose-dependent recovery of rod photoreceptor-driven function and preservation of retinal morphology in Cngb1-/- mice. In summary, these results demonstrate the efficacy of hCNGB1 gene supplementation therapy in the Cngb1-/- mouse model of RP45 and support the translation of this approach towards future clinical application.
Collapse
Affiliation(s)
- Johanna E Wagner
- Ludwig-Maximilians-Universität München, 9183, Department of Pharmacy - Center for Drug Research, Munich, Bayern, Germany;
| | - Lena Zobel
- Ludwig-Maximilians-Universität München, 9183, Department of Pharmacy - Center for Drug Research, Munich, Bayern, Germany.,Ludwig-Maximilians-Universität München, 9183, University Hospital - Department of Ophthalmology, Munich, Bayern, Germany;
| | - Maximilian Joachim Gerhardt
- Ludwig-Maximilians-Universität München, 9183, University Hospital - Department of Ophthalmology, Munich, Bayern, Germany;
| | - Catherine R O'Riordan
- Sanofi Genzyme, 2194, Gene Therapy, Rare Diseases, Framingham, Massachusetts, United States; Catherine.O'
| | - Amy Frederick
- Sanofi Genzyme, 2194, Gene Therapy, Rare Diseases, Framingham, Massachusetts, United States;
| | - Simon M Petersen-Jones
- Michigan State University, Veterinary Medical Center, East Lansing, Michigan, United States;
| | - Martin Biel
- Ludwig-Maximilians-Universität München, 9183, Department of Pharmacy - Center for Drug Research, Munich, Bayern, Germany;
| | - Stylianos Michalakis
- Ludwig-Maximilians-Universität München, 9183, Department of Pharmacy - Center for Drug Research, Munich, Bayern, Germany.,Ludwig-Maximilians-Universität München, 9183, University Hospital - Department of Ophthalmology, Munich, Bayern, Germany;
| |
Collapse
|
10
|
Barravecchia I, Demontis GC. HCN1 channels: A versatile tool for signal processing by primary sensory neurons. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:133-146. [PMID: 34197835 DOI: 10.1016/j.pbiomolbio.2021.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/11/2021] [Accepted: 06/25/2021] [Indexed: 12/20/2022]
Abstract
Most primary sensory neurons (PSNs) generate a slowly-activating inward current in response to membrane hyperpolarization (Ih) and express HCN1 along with additional isoforms coding for hyperpolarization-activated channels (HCN). Changes in HCN expression may affect the excitability and firing patterns of PSNs, but retinal and inner ear PSNs do not fire action potentials, suggesting HCN channel roles may extend beyond excitability and cell firing control. In patients taking Ih blockers, photopsia triggered in response to abrupt changes in luminance correlates with impaired visual signal processing via parallel rod and cone pathways. Furthermore, in a mouse model of inherited retinal degeneration, HCN blockers or Hcn1 genetic ablation may worsen photoreceptors' demise. PSN's use of HCN channels to adjust either their firing rate or process signals generated by sensory transduction in non-spiking PSNs indicates HCN1 channels as a versatile tool with a novel role in sensory processing beyond firing control.
Collapse
Affiliation(s)
- Ivana Barravecchia
- Department of Pharmacy, Università di Pisa, Italy, Via Bonanno, 6, 56126, Pisa, Italy; Istitute of Life Science, Scuola Superiore Sant' Anna, 56127, Pisa, Italy.
| | - Gian Carlo Demontis
- Department of Pharmacy, Università di Pisa, Italy, Via Bonanno, 6, 56126, Pisa, Italy.
| |
Collapse
|
11
|
Pavlou M, Schön C, Occelli LM, Rossi A, Meumann N, Boyd RF, Bartoe JT, Siedlecki J, Gerhardt MJ, Babutzka S, Bogedein J, Wagner JE, Priglinger SG, Biel M, Petersen‐Jones SM, Büning H, Michalakis S. Novel AAV capsids for intravitreal gene therapy of photoreceptor disorders. EMBO Mol Med 2021; 13:e13392. [PMID: 33616280 PMCID: PMC8033523 DOI: 10.15252/emmm.202013392] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022] Open
Abstract
Gene therapy using recombinant adeno-associated virus (rAAV) vectors to treat blinding retinal dystrophies has become clinical reality. Therapeutically impactful targeting of photoreceptors still relies on subretinal vector delivery, which detaches the retina and harbours substantial risks of collateral damage, often without achieving widespread photoreceptor transduction. Herein, we report the development of novel engineered rAAV vectors that enable efficient targeting of photoreceptors via less invasive intravitreal administration. A unique in vivo selection procedure was performed, where an AAV2-based peptide-display library was intravenously administered in mice, followed by isolation of vector DNA from target cells after only 24 h. This stringent selection yielded novel vectors, termed AAV2.GL and AAV2.NN, which mediate widespread and high-level retinal transduction after intravitreal injection in mice, dogs and non-human primates. Importantly, both vectors efficiently transduce photoreceptors in human retinal explant cultures. As proof-of-concept, intravitreal Cnga3 delivery using AAV2.GL lead to cone-specific expression of Cnga3 protein and rescued photopic cone responses in the Cnga3-/- mouse model of achromatopsia. These novel rAAV vectors expand the clinical applicability of gene therapy for blinding human retinal dystrophies.
Collapse
Affiliation(s)
- Marina Pavlou
- Department of OphthalmologyLudwig‐Maximilians‐UniversityMunichGermany
- Centre for Integrated Protein Science Munich (CIPSM) at the Department of PharmacyLudwig‐Maximilians‐UniversityMunichGermany
| | - Christian Schön
- Centre for Integrated Protein Science Munich (CIPSM) at the Department of PharmacyLudwig‐Maximilians‐UniversityMunichGermany
| | - Laurence M Occelli
- Department of Small Animal Clinical SciencesMichigan State UniversityEast LansingMIUSA
| | - Axel Rossi
- Laboratory for Infection Biology and Gene TransferInstitute of Experimental HaematologyHannover Medical SchoolHannoverGermany
| | - Nadja Meumann
- Laboratory for Infection Biology and Gene TransferInstitute of Experimental HaematologyHannover Medical SchoolHannoverGermany
- REBIRTH Research Centre for Translational Regenerative MedicineHannover Medical SchoolHannoverGermany
| | - Ryan F Boyd
- Ophthalmology ServicesCharles River LaboratoriesMattawanMIUSA
| | - Joshua T Bartoe
- Ophthalmology ServicesCharles River LaboratoriesMattawanMIUSA
| | - Jakob Siedlecki
- Department of OphthalmologyLudwig‐Maximilians‐UniversityMunichGermany
| | | | - Sabrina Babutzka
- Department of OphthalmologyLudwig‐Maximilians‐UniversityMunichGermany
- Centre for Integrated Protein Science Munich (CIPSM) at the Department of PharmacyLudwig‐Maximilians‐UniversityMunichGermany
| | - Jacqueline Bogedein
- Department of OphthalmologyLudwig‐Maximilians‐UniversityMunichGermany
- Centre for Integrated Protein Science Munich (CIPSM) at the Department of PharmacyLudwig‐Maximilians‐UniversityMunichGermany
| | - Johanna E Wagner
- Centre for Integrated Protein Science Munich (CIPSM) at the Department of PharmacyLudwig‐Maximilians‐UniversityMunichGermany
| | | | - Martin Biel
- Centre for Integrated Protein Science Munich (CIPSM) at the Department of PharmacyLudwig‐Maximilians‐UniversityMunichGermany
| | | | - Hildegard Büning
- Laboratory for Infection Biology and Gene TransferInstitute of Experimental HaematologyHannover Medical SchoolHannoverGermany
- REBIRTH Research Centre for Translational Regenerative MedicineHannover Medical SchoolHannoverGermany
| | - Stylianos Michalakis
- Department of OphthalmologyLudwig‐Maximilians‐UniversityMunichGermany
- Centre for Integrated Protein Science Munich (CIPSM) at the Department of PharmacyLudwig‐Maximilians‐UniversityMunichGermany
| |
Collapse
|
12
|
Light responses of mammalian cones. Pflugers Arch 2021; 473:1555-1568. [PMID: 33742309 DOI: 10.1007/s00424-021-02551-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 12/24/2022]
Abstract
Cone photoreceptors provide the foundation of most of human visual experience, but because they are smaller and less numerous than rods in most mammalian retinas, much less is known about their physiology. We describe new techniques and approaches which are helping to provide a better understanding of cone function. We focus on several outstanding issues, including the identification of the features of the phototransduction cascade that are responsible for the more rapid kinetics and decreased sensitivity of the cone response, the roles of inner-segment voltage-gated and Ca2+-activated channels, the means by which cones remain responsive even in the brightest illumination, mechanisms of cone visual pigment regeneration in constant light, and energy consumption of cones in comparison to that of rods.
Collapse
|
13
|
Koch M, Scheel C, Ma H, Yang F, Stadlmeier M, Glück AF, Murenu E, Traube FR, Carell T, Biel M, Ding XQ, Michalakis S. The cGMP-Dependent Protein Kinase 2 Contributes to Cone Photoreceptor Degeneration in the Cnga3-Deficient Mouse Model of Achromatopsia. Int J Mol Sci 2020; 22:E52. [PMID: 33374621 PMCID: PMC7793084 DOI: 10.3390/ijms22010052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/21/2022] Open
Abstract
Mutations in the CNGA3 gene, which encodes the A subunit of the cyclic guanosine monophosphate (cGMP)-gated cation channel in cone photoreceptor outer segments, cause total colour blindness, also referred to as achromatopsia. Cones lacking this channel protein are non-functional, accumulate high levels of the second messenger cGMP and degenerate over time after induction of ER stress. The cell death mechanisms that lead to loss of affected cones are only partially understood. Here, we explored the disease mechanisms in the Cnga3 knockout (KO) mouse model of achromatopsia. We found that another important effector of cGMP, the cGMP-dependent protein kinase 2 (Prkg2) is crucially involved in cGMP cytotoxicity of cones in Cnga3 KO mice. Virus-mediated knockdown or genetic ablation of Prkg2 in Cnga3 KO mice counteracted degeneration and preserved the number of cones. Analysis of markers of endoplasmic reticulum stress and unfolded protein response confirmed that induction of these processes in Cnga3 KO cones also depends on Prkg2. In conclusion, we identified Prkg2 as a novel key mediator of cone photoreceptor degeneration in achromatopsia. Our data suggest that this cGMP mediator could be a novel pharmacological target for future neuroprotective therapies.
Collapse
Affiliation(s)
- Mirja Koch
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.K.); (C.S.); (E.M.); (M.B.)
| | - Constanze Scheel
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.K.); (C.S.); (E.M.); (M.B.)
| | - Hongwei Ma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (H.M.); (F.Y.); (X.-Q.D.)
| | - Fan Yang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (H.M.); (F.Y.); (X.-Q.D.)
| | - Michael Stadlmeier
- Department of Chemistry, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.S.); (A.F.G.); (F.R.T.); (T.C.)
| | - Andrea F. Glück
- Department of Chemistry, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.S.); (A.F.G.); (F.R.T.); (T.C.)
| | - Elisa Murenu
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.K.); (C.S.); (E.M.); (M.B.)
- Department of Ophthalmology, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Franziska R. Traube
- Department of Chemistry, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.S.); (A.F.G.); (F.R.T.); (T.C.)
| | - Thomas Carell
- Department of Chemistry, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.S.); (A.F.G.); (F.R.T.); (T.C.)
| | - Martin Biel
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.K.); (C.S.); (E.M.); (M.B.)
| | - Xi-Qin Ding
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (H.M.); (F.Y.); (X.-Q.D.)
| | - Stylianos Michalakis
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-University, 81377 Munich, Germany; (M.K.); (C.S.); (E.M.); (M.B.)
- Department of Ophthalmology, Ludwig-Maximilians-University, 80336 Munich, Germany
| |
Collapse
|
14
|
Ropelewski P, Imanishi Y. RPE Cells Engulf Microvesicles Secreted by Degenerating Rod Photoreceptors. eNeuro 2020; 7:ENEURO.0507-19.2020. [PMID: 32376599 PMCID: PMC7242815 DOI: 10.1523/eneuro.0507-19.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/10/2020] [Accepted: 04/24/2020] [Indexed: 01/11/2023] Open
Abstract
Rhodopsin is mislocalized to the inner segment plasma membrane (IS PM) in various blinding disorders including autosomal-dominant retinitis pigmentosa caused by class I rhodopsin mutations. In these disorders, rhodopsin-laden microvesicles are secreted into the extracellular milieu by afflicted photoreceptor cells. Using a Xenopus laevis model expressing class I mutant rhodopsin or Na+/K+-ATPase (NKA) fused to Dendra2, we fluorescently labeled the microvesicles and found retinal pigment epithelial (RPE) cells are capable of engulfing microvesicles containing rhodopsin. A unique sorting mechanism allows class I mutant rhodopsin, but not NKA, to be packaged into the microvesicles. Under normal physiological conditions, NKA is not shed as microvesicles to the extracellular space, but is degraded intracellularly. Those studies provide novel insights into protein homeostasis in the photoreceptor IS PM.
Collapse
Affiliation(s)
- Philip Ropelewski
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| | - Yoshikazu Imanishi
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| |
Collapse
|
15
|
Collin GB, Gogna N, Chang B, Damkham N, Pinkney J, Hyde LF, Stone L, Naggert JK, Nishina PM, Krebs MP. Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss. Cells 2020; 9:E931. [PMID: 32290105 PMCID: PMC7227028 DOI: 10.3390/cells9040931] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated. The progression of PR cell loss with postnatal age was documented in mutant alleles of genes grouped by biological function. As anticipated, a wide range in the onset and rate of cell loss was observed among the reported models. The analysis underscored relationships between RD genes and ciliary function, transcription-coupled DNA damage repair, and cellular chloride homeostasis. Comparing the mouse gene list to human RD genes identified in the RetNet database revealed that mouse models are available for 40% of the known human diseases, suggesting opportunities for future research. This work may provide insight into the molecular players and pathways through which PR degenerative disease occurs and may be useful for planning translational studies.
Collapse
Affiliation(s)
- Gayle B. Collin
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Navdeep Gogna
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Nattaya Damkham
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jai Pinkney
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lillian F. Hyde
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Lisa Stone
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Jürgen K. Naggert
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Patsy M. Nishina
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| | - Mark P. Krebs
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; (G.B.C.); (N.G.); (B.C.); (N.D.); (J.P.); (L.F.H.); (L.S.); (J.K.N.)
| |
Collapse
|
16
|
Van Hook MJ, Nawy S, Thoreson WB. Voltage- and calcium-gated ion channels of neurons in the vertebrate retina. Prog Retin Eye Res 2019; 72:100760. [PMID: 31078724 PMCID: PMC6739185 DOI: 10.1016/j.preteyeres.2019.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
In this review, we summarize studies investigating the types and distribution of voltage- and calcium-gated ion channels in the different classes of retinal neurons: rods, cones, horizontal cells, bipolar cells, amacrine cells, interplexiform cells, and ganglion cells. We discuss differences among cell subtypes within these major cell classes, as well as differences among species, and consider how different ion channels shape the responses of different neurons. For example, even though second-order bipolar and horizontal cells do not typically generate fast sodium-dependent action potentials, many of these cells nevertheless possess fast sodium currents that can enhance their kinetic response capabilities. Ca2+ channel activity can also shape response kinetics as well as regulating synaptic release. The L-type Ca2+ channel subtype, CaV1.4, expressed in photoreceptor cells exhibits specific properties matching the particular needs of these cells such as limited inactivation which allows sustained channel activity and maintained synaptic release in darkness. The particular properties of K+ and Cl- channels in different retinal neurons shape resting membrane potentials, response kinetics and spiking behavior. A remaining challenge is to characterize the specific distributions of ion channels in the more than 100 individual cell types that have been identified in the retina and to describe how these particular ion channels sculpt neuronal responses to assist in the processing of visual information by the retina.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Scott Nawy
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
17
|
Abstract
Rods and cones are retinal photoreceptor neurons required for our visual sensation. Because of their highly polarized structures and well-characterized processes of G protein-coupled receptor-mediated phototransduction signaling, these photoreceptors have been excellent models for studying the compartmentalization and sorting of proteins. Rods and cones have a modified ciliary compartment called the outer segment (OS) as well as non-OS compartments. The distinct membrane protein compositions between OS and non-OS compartments suggest that the OS is separated from the rest of the cellular compartments by multiple barriers or gates that are selectively permissive to specific cargoes. This review discusses the mechanisms of protein sorting and compartmentalization in photoreceptor neurons. Proper sorting and compartmentalization of membrane proteins are required for signal transduction and transmission. This review also discusses the roles of compartmentalized signaling, which is compromised in various retinal ciliopathies.
Collapse
Affiliation(s)
- Yoshikazu Imanishi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA;
| |
Collapse
|
18
|
Deng WL, Gao ML, Lei XL, Lv JN, Zhao H, He KW, Xia XX, Li LY, Chen YC, Li YP, Pan D, Xue T, Jin ZB. Gene Correction Reverses Ciliopathy and Photoreceptor Loss in iPSC-Derived Retinal Organoids from Retinitis Pigmentosa Patients. Stem Cell Reports 2018. [PMID: 29526738 PMCID: PMC5998840 DOI: 10.1016/j.stemcr.2018.02.003] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Retinitis pigmentosa (RP) is an irreversible, inherited retinopathy in which early-onset nyctalopia is observed. Despite the genetic heterogeneity of RP, RPGR mutations are the most common causes of this disease. Here, we generated induced pluripotent stem cells (iPSCs) from three RP patients with different frameshift mutations in the RPGR gene, which were then differentiated into retinal pigment epithelium (RPE) cells and well-structured retinal organoids possessing electrophysiological properties. We observed significant defects in photoreceptor in terms of morphology, localization, transcriptional profiling, and electrophysiological activity. Furthermore, shorted cilium was found in patient iPSCs, RPE cells, and three-dimensional retinal organoids. CRISPR-Cas9-mediated correction of RPGR mutation rescued photoreceptor structure and electrophysiological property, reversed the observed ciliopathy, and restored gene expression to a level in accordance with that in the control using transcriptome-based analysis. This study recapitulated the pathogenesis of RPGR using patient-specific organoids and achieved targeted gene therapy of RPGR mutations in a dish as proof-of-concept evidence. HiPSC-derived 3D retinae with outer segments and electrophysiological properties RPGR mutation results in diseased photoreceptor in patient iPSC-derived 3D retinae Mutation correction rescues defects in photoreceptor morphology and electrophysiology Ciliogenesis defects appear in RPGR patient-specific iPSCs, iPSC-RPE, and 3D retinae
Collapse
Affiliation(s)
- Wen-Li Deng
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Mei-Ling Gao
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Xin-Lan Lei
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Ji-Neng Lv
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Huan Zhao
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Kai-Wen He
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Xi-Xi Xia
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Ling-Yun Li
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Yu-Chen Chen
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Yan-Ping Li
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Deng Pan
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China
| | - Tian Xue
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Zi-Bing Jin
- Lab for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou 325027, China.
| |
Collapse
|
19
|
Blank T, Goldmann T, Koch M, Amann L, Schön C, Bonin M, Pang S, Prinz M, Burnet M, Wagner JE, Biel M, Michalakis S. Early Microglia Activation Precedes Photoreceptor Degeneration in a Mouse Model of CNGB1-Linked Retinitis Pigmentosa. Front Immunol 2018; 8:1930. [PMID: 29354133 PMCID: PMC5760536 DOI: 10.3389/fimmu.2017.01930] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/15/2017] [Indexed: 01/12/2023] Open
Abstract
Retinitis pigmentosa (RP) denotes a family of inherited blinding eye diseases characterized by progressive degeneration of rod and cone photoreceptors in the retina. In most cases, a rod-specific genetic defect results in early functional loss and degeneration of rods, which is followed by degeneration of cones and loss of daylight vision at later stages. Microglial cells, the immune cells of the central nervous system, are activated in retinas of RP patients and in several RP mouse models. However, it is still a matter of debate whether activated microglial cells may be responsible for the amplification of the typical degenerative processes. Here, we used Cngb1−/− mice, which represent a slow degenerative mouse model of RP, to investigate the extent of microglia activation in retinal degeneration. With a combination of FACS analysis, immunohistochemistry and gene expression analysis we established that microglia in the Cngb1−/− retina were already activated in an early, predegenerative stage of the disease. The evidence available so far suggests that early retinal microglia activation represents a first step in RP, which might initiate or accelerate photoreceptor degeneration.
Collapse
Affiliation(s)
- Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Goldmann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,In Vivo Pharmacology, Synovo GmbH, Tübingen, Germany
| | - Mirja Koch
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas Amann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Christian Schön
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Bonin
- Institute for Medical Genetics and Applied Genomics Transcriptomics, University of Tübingen, Tübingen, Germany.,IMGM Laboratories GmbH, Planegg, Germany
| | - Shengru Pang
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | | | - Johanna E Wagner
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
20
|
Barbato S, Marrocco E, Intartaglia D, Pizzo M, Asteriti S, Naso F, Falanga D, Bhat RS, Meola N, Carissimo A, Karali M, Prosser HM, Cangiano L, Surace EM, Banfi S, Conte I. MiR-211 is essential for adult cone photoreceptor maintenance and visual function. Sci Rep 2017; 7:17004. [PMID: 29209045 PMCID: PMC5717140 DOI: 10.1038/s41598-017-17331-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/16/2017] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs) are key post-transcriptional regulators of gene expression that play an important role in the control of fundamental biological processes in both physiological and pathological conditions. Their function in retinal cells is just beginning to be elucidated, and a few have been found to play a role in photoreceptor maintenance and function. MiR-211 is one of the most abundant miRNAs in the developing and adult eye. However, its role in controlling vertebrate visual system development, maintenance and function so far remain incompletely unexplored. Here, by targeted inactivation in a mouse model, we identify a critical role of miR-211 in cone photoreceptor function and survival. MiR-211 knockout (-/-) mice exhibited a progressive cone dystrophy accompanied by significant alterations in visual function. Transcriptome analysis of the retina from miR-211-/- mice during cone degeneration revealed significant alteration of pathways related to cell metabolism. Collectively, this study highlights for the first time the impact of miR-211 function in the retina and significantly contributes to unravelling the role of specific miRNAs in cone photoreceptor function and survival.
Collapse
Affiliation(s)
- Sara Barbato
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Elena Marrocco
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Daniela Intartaglia
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Mariateresa Pizzo
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Sabrina Asteriti
- Department of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, United Kingdom
| | - Federica Naso
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Danila Falanga
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Rajeshwari S Bhat
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Nicola Meola
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
- Aarhus University, Department of Molecular Biology and Genetics, C.F. Møllers Allé 3 building 1130, 422-8000, Aarhus C, Denmark
| | - Annamaria Carissimo
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
| | - Marianthi Karali
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
- Medical Genetics, Department of Biochemistry, Biophysics and General Pathology, University "Luigi Vanvitelli", via Luigi De Crecchio 7, 80138, Naples, Italy
| | - Haydn M Prosser
- The Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Lorenzo Cangiano
- Department of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy
| | - Enrico Maria Surace
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Sandro Banfi
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy.
- Medical Genetics, Department of Biochemistry, Biophysics and General Pathology, University "Luigi Vanvitelli", via Luigi De Crecchio 7, 80138, Naples, Italy.
| | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (Naples), 80078, Italy.
| |
Collapse
|
21
|
Petersen-Jones SM, Occelli LM, Winkler PA, Lee W, Sparrow JR, Tsukikawa M, Boye SL, Chiodo V, Capasso JE, Becirovic E, Schön C, Seeliger MW, Levin AV, Michalakis S, Hauswirth WW, Tsang SH. Patients and animal models of CNGβ1-deficient retinitis pigmentosa support gene augmentation approach. J Clin Invest 2017; 128:190-206. [PMID: 29202463 PMCID: PMC5749539 DOI: 10.1172/jci95161] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/10/2017] [Indexed: 01/07/2023] Open
Abstract
Retinitis pigmentosa (RP) is a major cause of blindness that affects 1.5 million people worldwide. Mutations in cyclic nucleotide-gated channel β 1 (CNGB1) cause approximately 4% of autosomal recessive RP. Gene augmentation therapy shows promise for treating inherited retinal degenerations; however, relevant animal models and biomarkers of progression in patients with RP are needed to assess therapeutic outcomes. Here, we evaluated RP patients with CNGB1 mutations for potential biomarkers of progression and compared human phenotypes with those of mouse and dog models of the disease. Additionally, we used gene augmentation therapy in a CNGβ1-deficient dog model to evaluate potential translation to patients. CNGB1-deficient RP patients and mouse and dog models had a similar phenotype characterized by early loss of rod function and slow rod photoreceptor loss with a secondary decline in cone function. Advanced imaging showed promise for evaluating RP progression in human patients, and gene augmentation using adeno-associated virus vectors robustly sustained the rescue of rod function and preserved retinal structure in the dog model. Together, our results reveal an early loss of rod function in CNGB1-deficient patients and a wide window for therapeutic intervention. Moreover, the identification of potential biomarkers of outcome measures, availability of relevant animal models, and robust functional rescue from gene augmentation therapy support future work to move CNGB1-RP therapies toward clinical trials.
Collapse
Affiliation(s)
- Simon M Petersen-Jones
- Department of Small Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, USA
| | - Laurence M Occelli
- Department of Small Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, USA
| | - Paige A Winkler
- Department of Small Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, USA
| | - Winston Lee
- Department of Ophthalmology Pathology & Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Janet R Sparrow
- Department of Ophthalmology Pathology & Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Mai Tsukikawa
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Vince Chiodo
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Jenina E Capasso
- Ocular Genetics, Wills Eye Hospital (WEH), Philadelphia, Pennsylvania, USA
| | - Elvir Becirovic
- Center for Integrated Protein Science Munich (CIPSM), Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Schön
- Center for Integrated Protein Science Munich (CIPSM), Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mathias W Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Alex V Levin
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Ocular Genetics, Wills Eye Hospital (WEH), Philadelphia, Pennsylvania, USA
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich (CIPSM), Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Stephen H Tsang
- Department of Ophthalmology Pathology & Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University Medical Center (CUMC), Edward S. Harkness Eye Institute, New York, New York, USA
| |
Collapse
|
22
|
Sartiani L, Mannaioni G, Masi A, Novella Romanelli M, Cerbai E. The Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels: from Biophysics to Pharmacology of a Unique Family of Ion Channels. Pharmacol Rev 2017; 69:354-395. [PMID: 28878030 DOI: 10.1124/pr.117.014035] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels are important members of the voltage-gated pore loop channels family. They show unique features: they open at hyperpolarizing potential, carry a mixed Na/K current, and are regulated by cyclic nucleotides. Four different isoforms have been cloned (HCN1-4) that can assemble to form homo- or heterotetramers, characterized by different biophysical properties. These proteins are widely distributed throughout the body and involved in different physiologic processes, the most important being the generation of spontaneous electrical activity in the heart and the regulation of synaptic transmission in the brain. Their role in heart rate, neuronal pacemaking, dendritic integration, learning and memory, and visual and pain perceptions has been extensively studied; these channels have been found also in some peripheral tissues, where their functions still need to be fully elucidated. Genetic defects and altered expression of HCN channels are linked to several pathologies, which makes these proteins attractive targets for translational research; at the moment only one drug (ivabradine), which specifically blocks the hyperpolarization-activated current, is clinically available. This review discusses current knowledge about HCN channels, starting from their biophysical properties, origin, and developmental features, to (patho)physiologic role in different tissues and pharmacological modulation, ending with their present and future relevance as drug targets.
Collapse
Affiliation(s)
- Laura Sartiani
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| |
Collapse
|
23
|
Peripherin-2 and Rom-1 have opposing effects on rod outer segment targeting of retinitis pigmentosa-linked peripherin-2 mutants. Sci Rep 2017; 7:2321. [PMID: 28539581 PMCID: PMC5443838 DOI: 10.1038/s41598-017-02514-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/12/2017] [Indexed: 12/30/2022] Open
Abstract
Mutations in the photoreceptor outer segment (OS) specific peripherin-2 lead to autosomal dominant retinitis pigmentosa (adRP). By contrast, mutations in the peripherin-2 homolog Rom-1 cause digenic RP in combination with certain heterozygous mutations in peripherin-2. The mechanisms underlying the differential role of peripherin-2 and Rom-1 in RP pathophysiology remained elusive so far. Here, focusing on two adRP-linked peripherin-2 mutants, P210L and C214S, we analyzed the binding characteristics, protein assembly, and rod OS targeting of wild type (perWT), mutant peripherin-2 (perMT), or Rom-1 complexes, which can be formed in patients heterozygous for peripherin-2 mutations. Both mutants are misfolded and lead to decreased binding to perWT and Rom-1. Furthermore, both mutants are preferentially forming non-covalent perMT-perMT, perWT-perMT, and Rom-1-perMT dimers. However, only perWT-perMT, but not perMT-perMT or Rom-1-perMT complexes could be targeted to murine rod OS. Our study provides first evidence that non-covalent perWT-perMT dimers can be targeted to rod OS. Finally, our study unravels unexpected opposing roles of perWT and Rom-1 in rod OS targeting of adRP-linked peripherin-2 mutants and suggests a new treatment strategy for the affected individuals.
Collapse
|
24
|
Schön C, Paquet-Durand F, Michalakis S. Cav1.4 L-Type Calcium Channels Contribute to Calpain Activation in Degenerating Photoreceptors of rd1 Mice. PLoS One 2016; 11:e0156974. [PMID: 27270916 PMCID: PMC4896623 DOI: 10.1371/journal.pone.0156974] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/23/2016] [Indexed: 12/01/2022] Open
Abstract
Retinitis pigmentosa is an inherited blinding disorder characterized by progressive degeneration and loss of photoreceptors. The exact mechanism of degeneration and cell death of photoreceptors is not known, but is thought to involve disturbed Ca2+—signaling. Ca2+ can enter the photoreceptor cell via outer segment cyclic nucleotide-gated (CNG) channels or synaptic Cav1.4 L-type voltage-gated calcium channels (VGCC). Previously, we have shown that genetic ablation of the Cngb1 gene encoding the B subunit of the rod CNG channel delays the fast progressing degeneration in the rd1 mutant mouse model of retinitis pigmentosa. In this study, we crossbred rd1 mice with the Cacna1f-deficient mouse lacking the Cav1.4 α1 subunit of the L-type VGCC. Longitudinal in vivo examinations of photoreceptor layer thickness by optical coherence tomography revealed a significant, but not sustained delay of retinal degeneration in Cacna1f x rd1 double mutant mice compared to rd1 mice. This was accompanied by a reduction of TUNEL positive cells in the early phase of rod degeneration. Remarkably, Cacna1f x rd1 double mutant mice displayed a strong decrease in the activation of the Ca2+-dependent protease calpain during photoreceptor loss. Our results show that genetic deletion of the synaptic Cav1.4 L-type VGCCs impairs calpain activation and leads to a short-term preservation of photoreceptors in the rd1 mouse.
Collapse
Affiliation(s)
- Christian Schön
- Center for Integrated Protein Science Munich CiPSM at the Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - François Paquet-Durand
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Schleichstr. 4/3, 72076, Tübingen, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich CiPSM at the Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
25
|
Forman OP, Hitti RJ, Boursnell M, Miyadera K, Sargan D, Mellersh C. Canine genome assembly correction facilitates identification of a MAP9 deletion as a potential age of onset modifier for RPGRIP1-associated canine retinal degeneration. Mamm Genome 2016; 27:237-45. [PMID: 27017229 DOI: 10.1007/s00335-016-9627-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/15/2016] [Indexed: 11/25/2022]
Abstract
Retinal degeneration (RD) in the Miniature Long Haired Dachshund (MLHD) is a cone-rod dystrophy resulting in eventual blindness in affected individuals. In a previous study, a 44-nucleotide insertion (ins44) in exon 2 of RPGRIP1 was associated with RD. However, results on an extended population of MLHD revealed a variable RD onset age for ins44 homozygous dogs. Further investigations using a genome-wide association study comparing early onset and late onset RD cases identified an age of onset modifying locus for RD, approximately 30 Mb upstream of RPGRIP1 on chr15. In this investigation, target enriched sequencing identified a MAP9 deletion spanning approximately 22 kb associated with early RD onset. Identification of the deletion required correction to the CanFam3.1 genome build as canine MAP9 is part of a historic tandem duplication, resulting in incomplete assembly of this genome region. The deletion breakpoints were identified in MAP9 intron 10 and in a downstream partial MAP9 pseudogene. The fusion of these two genes, which we have called MAP9 EORD (microtubule-associated protein, early onset retinal degeneration), is in frame and is expressed at the RNA level, with the 3' region containing several predicted deleterious variants. We speculate that MAP9 associates with α-tubulin in the basal body of the cilium. RPGRIP1 is also known to locate to the cilium, where it is closely associated with RPGR. RPGRIP1 mutations also cause redistribution of α-tubulin away from the ciliary region in photoreceptors. Hence, a MAP9 partial deficit is a particularly attractive candidate to synergise with a partial RPGRIP1 deficit to cause a more serious disease.
Collapse
Affiliation(s)
- Oliver P Forman
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, Suffolk, CB8 7UU, UK
| | - Rebekkah J Hitti
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, Suffolk, CB8 7UU, UK.
| | - Mike Boursnell
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, Suffolk, CB8 7UU, UK
| | - Keiko Miyadera
- School of Veterinary Medicine, University of Pennsylvania, 3900 Delancey St, Philadelphia, PA, 19104, USA
| | - David Sargan
- Comparative Genetics Group, Department of Clinical Veterinary Medicine, University of Cambridge, Madingley Rd., Cambridge, CB3 0ES, UK
| | - Cathryn Mellersh
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, Suffolk, CB8 7UU, UK
| |
Collapse
|