1
|
Sato S, Kefalov VJ. Characterization of zebrafish rod and cone photoresponses. Sci Rep 2025; 15:13413. [PMID: 40251282 PMCID: PMC12008237 DOI: 10.1038/s41598-025-96058-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/25/2025] [Indexed: 04/20/2025] Open
Abstract
Zebrafish is a popular species widely used in vision research. The zebrafish retina has one rod and four cone subtypes (UV-, blue-, green-, and red-sensitive) with 40%-rod 60%-cone ratio, making it suitable for comparable studies of rods and cones in health and disease. However, the basic photoresponse properties of the four zebrafish cone subtypes have not been described yet. Here, we established a method for collecting flash photoresponses from zebrafish rods and cones by recording membrane current with a suction electrode. Photoreceptor subtypes could be distinguished based on their characteristic morphology and spectral sensitivity. Rods showed 40-220-fold higher photosensitivity than cones. In the four cone subtypes, green-sensitive cones showed the highest sensitivity, 5.5-fold higher than that of red cones. Unexpectedly, rods produced smaller flash responses than cones despite their larger outer segments. Dim flash response analysis showed the quickest response kinetics in blue- and red-sensitive cones, with responses about 2-fold faster than the responses of UV- and green-sensitive cones, and 6.6-fold faster than the rod responses. We also obtained pharmacologically isolated photoreceptor voltage responses (a-wave) from isolated zebrafish retinas using ex vivo electroretinography (ERG). Dim flashes evoked rod-only responses, while bright flashes evoked two-component responses with a slow rod component and a fast cone component. Red- and green-sensitive cones were the dominant sources of the overall cone response. These studies provide a foundation for the use of zebrafish rods and cones to study the fundamental mechanisms that modulate the function of vertebrate photoreceptors in health and disease.
Collapse
Affiliation(s)
- Shinya Sato
- Department of Ophthalmology, Gavin Herbert Eye Institute-Center for Translational Vision Research, University of California, Irvine, Irvine, USA.
| | - Vladimir J Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute-Center for Translational Vision Research, University of California, Irvine, Irvine, USA.
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
2
|
Sato S, Kefalov V. Characterization of zebrafish rod and cone photoresponses. RESEARCH SQUARE 2025:rs.3.rs-5984163. [PMID: 40162217 PMCID: PMC11952657 DOI: 10.21203/rs.3.rs-5984163/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Zebrafish is a popular species widely used in vision research. The zebrafish retina has one rod and four cone subtypes (UV-, blue-, green-, and red-sensitive) with 40%-rod 60%-cone ratio, making it suitable for comparable studies of rods and cones in health and disease. However, the basic photoresponse properties of the four zebrafish cone subtypes have not been described yet. Here, we established a method for collecting flash photoresponses from zebrafish rods and cones by recording membrane current with a suction electrode. Photoreceptor subtypes could be distinguished based on their characteristic morphology and spectral sensitivity. Rods showed 40-220-fold higher photosensitivity than cones. In the four cone subtypes, green-sensitive cones showed the highest sensitivity, 5.5-fold higher than that of red cones. Unexpectedly, rods produced smaller flash responses than cones despite their larger outer segments. Dim flash response analysis showed the quickest response kinetics in blue- and red-sensitive cones, with responses about 2-fold faster than the responses of UV- and green-sensitive cones, and 6.6-fold faster than the rod responses. We also obtained pharmacologically isolated photoreceptor voltage responses (a-wave) from isolated zebrafish retinas using ex vivo electroretinography (ERG). Dim flashes evoked rod-only responses, while bright flashes evoked two-component responses with a slow rod component and a fast cone component. Red- and green-sensitive cones were the dominant sources of the overall cone response. These studies provide a foundation for the use of zebrafish rods and cones to study the fundamental mechanisms that modulate the function of vertebrate photoreceptors in health and disease.
Collapse
|
3
|
Hossain MS, Wang A, Anika S, Zhang Z, Mozhdehi D. Genetically Engineered Liposwitch-Based Nanomaterials. Biomacromolecules 2024; 25:8058-8068. [PMID: 39495202 PMCID: PMC11632658 DOI: 10.1021/acs.biomac.4c01388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Fusion of intrinsically disordered and globular proteins is a powerful strategy to create functional nanomaterials. However, the immutable nature of genetic encoding restricts the dynamic adaptability of nanostructures postexpression. To address this, we envisioned using a myristoyl switch, a protein that combines allostery and post-translational modifications─two strategies that modify protein properties without altering their sequence─to regulate intrinsically disordered protein (IDP)-driven nanoassembly. A typical myristoyl switch, allosterically activated by a stimulus, reveals a sequestered lipid for membrane association. We hypothesize that this conditional exposure of lipids can regulate the assembly of fusion proteins, a concept we term "liposwitching". We tested this by fusing recoverin, a calcium-dependent myristoyl switch, with elastin-like polypeptide, a thermoresponsive model IDP. Biophysical analyses confirmed recoverin's myristoyl-switch functionality, while dynamic light scattering and cryo-transmission electron microscopy showed distinct calcium- and lipidation-dependent phase separation and assembly. This study highlights liposwitching as a viable strategy for controlling DP-driven nanoassembly, enabling applications in synthetic biology and cellular engineering.
Collapse
Affiliation(s)
| | - Alex Wang
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Salma Anika
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Zhe Zhang
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| | - Davoud Mozhdehi
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, New York 13244, United States
| |
Collapse
|
4
|
Kuo BL, Muste JC, Russell MW, Wu AK, Valentim CCS, Singh RP. Evidence for the Hepato-Retinal Axis: A Systematic Review. Ophthalmic Surg Lasers Imaging Retina 2024; 55:587-596. [PMID: 39037358 DOI: 10.3928/23258160-20240524-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
BACKGROUND AND OBJECTIVE Liver health has been reported to be associated with retinal pathology in various ways. These include deposition of retino-toxins, neovascular drive, and disruption of the blood-retina barrier. Extrahepatic synthesis of implicated molecules and hemodynamic changes in liver dysfunction are also considered. The objective was to review the current evidence for and against a hepato-retinal axis that may guide further areas of preclinical and clinical investigation. METHODS This was a systematic review. PubMed and Cochrane were queried for English language studies examining the connection between hepatic dysfunction and retinal pathology. RESULTS Fourteen studies were included and examined out of 604 candidate publications. The studies selected include preclinical studies as well as clinical case series and studies. CONCLUSIONS Several liver pathologies may be linked to retinal pathology as mediated by hepatically synthesized molecules. The hepato-retinal axis may be present and further, targeted studies of the axis are warranted. [Ophthalmic Surg Lasers Imaging Retina 2024;55:587-596.].
Collapse
|
5
|
Belousov A, Maslov I, Orekhov P, Khorn P, Kuzmichev P, Baleeva N, Motov V, Bogorodskiy A, Krasnova S, Mineev K, Zinchenko D, Zernii E, Ivanovich V, Permyakov S, Hofkens J, Hendrix J, Cherezov V, Gensch T, Mishin A, Baranov M, Mishin A, Borshchevskiy V. Monitoring GPCR conformation with GFP-inspired dyes. iScience 2024; 27:110466. [PMID: 39156645 PMCID: PMC11326922 DOI: 10.1016/j.isci.2024.110466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/15/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024] Open
Abstract
Solvatochromic compounds have emerged as valuable environment-sensitive probes for biological research. Here we used thiol-reactive solvatochromic analogs of the green fluorescent protein (GFP) chromophore to track conformational changes in two proteins, recoverin and the A2A adenosine receptor (A2AAR). Two dyes showed Ca2+-induced fluorescence changes when attached to recoverin. Our best-performing dye, DyeC, exhibited agonist-induced changes in both intensity and shape of its fluorescence spectrum when attached to A2AAR; none of these effects were observed with other common environment-sensitive dyes. Molecular dynamics simulations showed that activation of the A2AAR led to a more confined and hydrophilic environment for DyeC. Additionally, an allosteric modulator of A2AAR induced distinct fluorescence changes in the DyeC spectrum, indicating a unique receptor conformation. Our study demonstrated that GFP-inspired dyes are effective for detecting structural changes in G protein-coupled receptors (GPCRs), offering advantages such as intensity-based and ratiometric tracking, redshifted fluorescence spectra, and sensitivity to allosteric modulation.
Collapse
Affiliation(s)
- Anatoliy Belousov
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Ivan Maslov
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Dynamic Bioimaging Lab, Advanced Optical Microscopy Centre, Biomedical Research Institute, Agoralaan C (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium
- Laboratory for Photochemistry and Spectroscopy, Division for Molecular Imaging and Photonics, Department of Chemistry, KU Leuven, 3001 Leuven, Belgium
| | - Philipp Orekhov
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China
- Sechenov University, Moscow 119146, Russia
| | - Polina Khorn
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Pavel Kuzmichev
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Nadezhda Baleeva
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Vladislav Motov
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | | | - Svetlana Krasnova
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- National Research University Higher School of Economics, Moscow 101000, Russia
| | - Konstantin Mineev
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry Zinchenko
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Evgeni Zernii
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | | | - Sergei Permyakov
- Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino 142292, Russia
| | - Johan Hofkens
- Laboratory for Photochemistry and Spectroscopy, Division for Molecular Imaging and Photonics, Department of Chemistry, KU Leuven, 3001 Leuven, Belgium
- Max Plank Institute for Polymer Research, Mainz, Germany
| | - Jelle Hendrix
- Dynamic Bioimaging Lab, Advanced Optical Microscopy Centre, Biomedical Research Institute, Agoralaan C (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium
- Laboratory for Photochemistry and Spectroscopy, Division for Molecular Imaging and Photonics, Department of Chemistry, KU Leuven, 3001 Leuven, Belgium
| | - Vadim Cherezov
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Thomas Gensch
- Laboratory for Photochemistry and Spectroscopy, Division for Molecular Imaging and Photonics, Department of Chemistry, KU Leuven, 3001 Leuven, Belgium
| | - Alexander Mishin
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Mikhail Baranov
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Alexey Mishin
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Valentin Borshchevskiy
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Joint Institute for Nuclear Research, Dubna 141980, Russian Federation
| |
Collapse
|
6
|
Zaher A, Petronek MS, Allen BG, Mapuskar KA. Balanced Duality: H 2O 2-Based Therapy in Cancer and Its Protective Effects on Non-Malignant Tissues. Int J Mol Sci 2024; 25:8885. [PMID: 39201571 PMCID: PMC11354297 DOI: 10.3390/ijms25168885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
Conventional cancer therapy strategies, although centered around killing tumor cells, often lead to severe side effects on surrounding normal tissues, thus compromising the chronic quality of life in cancer survivors. Hydrogen peroxide (H2O2) is a secondary signaling molecule that has an array of functions in both tumor and normal cells, including the promotion of cell survival pathways and immune cell modulation in the tumor microenvironment. H2O2 is a reactive oxygen species (ROS) crucial in cellular homeostasis and signaling (at concentrations maintained under nM levels), with increased steady-state levels in tumors relative to their normal tissue counterparts. Increased steady-state levels of H2O2 in tumor cells, make them vulnerable to oxidative stress and ultimately, cell death. Recently, H2O2-producing therapies-namely, pharmacological ascorbate and superoxide dismutase mimetics-have emerged as compelling complementary treatment strategies in cancer. Both pharmacological ascorbate and superoxide dismutase mimetics can generate excess H2O2 to overwhelm the impaired H2O2 removal capacity of cancer cells. This review presents an overview of H2O2 metabolism in the physiological and malignant states, in addition to discussing the anti-tumor and normal tissue-sparing mechanism(s) of, and clinical evidence for, two H2O2-based therapies, pharmacological ascorbate and superoxide dismutase mimetics.
Collapse
Affiliation(s)
| | | | | | - Kranti A. Mapuskar
- Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA; (A.Z.); (M.S.P.); (B.G.A.)
| |
Collapse
|
7
|
Montaldo P, Burgod C, Herberg JA, Kaforou M, Cunnington AJ, Mejias A, Cirillo G, Miraglia Del Giudice E, Capristo C, Bandiya P, Kamalaratnam CN, Chandramohan R, Manerkar S, Rodrigo R, Sumanasena S, Krishnan V, Pant S, Shankaran S, Thayyil S. Whole-Blood Gene Expression Profile After Hypoxic-Ischemic Encephalopathy. JAMA Netw Open 2024; 7:e2354433. [PMID: 38306098 PMCID: PMC10837749 DOI: 10.1001/jamanetworkopen.2023.54433] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/06/2023] [Indexed: 02/03/2024] Open
Abstract
Importance Induced hypothermia, the standard treatment for hypoxic-ischemic encephalopathy (HIE) in high-income countries (HICs), is less effective in the low-income populations in South Asia, who have the highest disease burden. Objective To investigate the differences in blood genome expression profiles of neonates with HIE from an HIC vs neonates with HIE from South Asia. Design, Setting, and Participants This case-control study analyzed data from (1) a prospective observational study involving neonates with moderate or severe HIE who underwent whole-body hypothermia between January 2017 and June 2019 and age-matched term healthy controls in Italy and (2) a randomized clinical trial involving neonates with moderate or severe HIE in India, Sri Lanka, and Bangladesh recruited between August 2015 and February 2019. Data were analyzed between October 2020 and August 2023. Exposure Whole-blood RNA that underwent next-generation sequencing. Main Outcome and Measures The primary outcomes were whole-blood genome expression profile at birth associated with adverse outcome (death or disability at 18 months) after HIE in the HIC and South Asia cohorts and changes in whole-genome expression profile during the first 72 hours after birth in neonates with HIE and healthy controls from the HIC cohort. Blood samples for RNA extraction were collected before whole-body hypothermia at 4 time points (6, 24, 48, and 72 hours after birth) for the HIC cohort. Only 1 blood sample was drawn within 6 hours after birth for the South Asia cohort. Results The HIC cohort was composed of 35 neonates (21 females [60.0%]) with a median (IQR) birth weight of 3.3 (3.0-3.6) kg and gestational age of 40.0 (39.0-40.6) weeks. The South Asia cohort consisted of 99 neonates (57 males [57.6%]) with a median (IQR) birth weight of 2.9 (2.7-3.3) kg and gestational age of 39.0 (38.0-40.0) weeks. Healthy controls included 14 neonates (9 females [64.3%]) with a median (IQR) birth weight of 3.4 (3.2-3.7) kg and gestational age of 39.2 (38.9-40.4) weeks. A total of 1793 significant genes in the HIC cohort and 99 significant genes in the South Asia cohort were associated with adverse outcome (false discovery rate <0.05). Only 11 of these genes were in common, and all had opposite direction in fold change. The most significant pathways associated with adverse outcome were downregulation of eukaryotic translation initiation factor 2 signaling in the HIC cohort (z score = -4.56; P < .001) and aldosterone signaling in epithelial cells in the South Asia cohort (z score = null; P < .001). The genome expression profile of neonates with HIE (n = 35) at birth, 24 hours, 48 hours, and 72 hours remained significantly different from that of age-matched healthy controls in the HIC cohort (n = 14). Conclusions and Relevance This case-control study found that disease mechanisms underlying HIE were primarily associated with acute hypoxia in the HIC cohort and nonacute hypoxia in the South Asia cohort. This finding might explain the lack of hypothermic neuroprotection.
Collapse
Affiliation(s)
- Paolo Montaldo
- Centre for Perinatal Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
- Department of Women's and Children's Health and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Constance Burgod
- Centre for Perinatal Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jethro A. Herberg
- Section of Paediatric Infectious Disease and Centre for Paediatrics and Child Health, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Myrsini Kaforou
- Section of Paediatric Infectious Disease and Centre for Paediatrics and Child Health, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Aubrey J. Cunnington
- Section of Paediatric Infectious Disease and Centre for Paediatrics and Child Health, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Asuncion Mejias
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Grazia Cirillo
- Department of Women's and Children's Health and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Emanuele Miraglia Del Giudice
- Department of Women's and Children's Health and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Carlo Capristo
- Department of Women's and Children's Health and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Prathik Bandiya
- Department of Neonatology, Indira Gandhi Institute of Child Health, Bengaluru, India
| | | | - Rema Chandramohan
- Institute of Child Health, Department of Neonatology, Madras Medical College, Chennai, India
| | - Swati Manerkar
- Department of Neonatology, Lokmanya Tilak Municipal Medical College, Mumbai, India
| | - Ranmali Rodrigo
- Department of Pediatrics, University of Kelaniya, Colombo, Sri Lanka
| | | | - Vaisakh Krishnan
- Centre for Perinatal Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Stuti Pant
- Centre for Perinatal Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Seetha Shankaran
- Neonatal-Perinatal Medicine, Wayne State University, Detroit, Michigan
| | - Sudhin Thayyil
- Centre for Perinatal Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
8
|
Margo TE, Chen FS, Chen YJ, Chen CK. Grk1 Missense Mutations in Type II Oguchi Disease: A Literature Review. ANNALS OF BIOMEDICAL RESEARCH 2024; 5:1-7. [PMID: 39906762 PMCID: PMC11793915 DOI: 10.61545/abr-5-128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Oguchi disease is a rare form of congenital stationary night blindness resulting from arrestin-1 (SAG) or rhodopsin kinase (GRK1) loss-of-function mutations. Unlike other congenital nyctalopias, patients with Oguchi disease can reach the dark-adapted state, albeit only after several hours of sustained darkness exposure. The mechanism underlying rhodopsin kinase dysfunction in Oguchi disease remains understudied. Previous research utilized the Grk1 knockout mice to reveal its role in phototransduction, the process that transduces light into neuronal signals in rod and cone photoreceptors. By studying Grk1 missense mutations via a knock-in approach, a more complete picture of the Oguchi disease mechanism involving GRK1 may be readily harvested. We summarize here the current knowledge on the Type II Oguchi disease with Grk1 missense mutations by focusing on the interaction of GRK1 with other proteins, and how these interactions influence dark adaptation. We call for more detailed analyses of GRK1 missense mutations in animal models, particularly V380D and L157P, to reveal novel disease mechanisms to gain further insight onto GRK1's action and function.
Collapse
Affiliation(s)
- Theodore Edward Margo
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Frank Sungping Chen
- Division of Otolaryngology, PeaceHealth Medical Group, Eugene, Oregan, OR 97401, USA
| | - Yu-Jiun Chen
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ching-Kang Chen
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Ophthalmology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
9
|
Zheng Y, Sun C, Zhang X, Ruzycki PA, Chen S. Missense mutations in CRX homeodomain cause dominant retinopathies through two distinct mechanisms. eLife 2023; 12:RP87147. [PMID: 37963072 PMCID: PMC10645426 DOI: 10.7554/elife.87147] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
Homeodomain transcription factors (HD TFs) are instrumental to vertebrate development. Mutations in HD TFs have been linked to human diseases, but their pathogenic mechanisms remain elusive. Here, we use Cone-Rod Homeobox (CRX) as a model to decipher the disease-causing mechanisms of two HD mutations, p.E80A and p.K88N, that produce severe dominant retinopathies. Through integrated analysis of molecular and functional evidence in vitro and in knock-in mouse models, we uncover two novel gain-of-function mechanisms: p.E80A increases CRX-mediated transactivation of canonical CRX target genes in developing photoreceptors; p.K88N alters CRX DNA-binding specificity resulting in binding at ectopic sites and severe perturbation of CRX target gene expression. Both mechanisms produce novel retinal morphological defects and hinder photoreceptor maturation distinct from loss-of-function models. This study reveals the distinct roles of E80 and K88 residues in CRX HD regulatory functions and emphasizes the importance of transcriptional precision in normal development.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetic and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St LouisSaint LouisUnited States
- Department of Ophthalmology and Visual Sciences, Washington University in St LouisSaint LouisUnited States
| | - Chi Sun
- Molecular Genetic and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St LouisSaint LouisUnited States
- Department of Ophthalmology and Visual Sciences, Washington University in St LouisSaint LouisUnited States
| | - Xiaodong Zhang
- Department of Ophthalmology and Visual Sciences, Washington University in St LouisSaint LouisUnited States
| | - Philip A Ruzycki
- Department of Ophthalmology and Visual Sciences, Washington University in St LouisSaint LouisUnited States
- Department of Genetics, Washington University in St LouisSaint LouisUnited States
| | - Shiming Chen
- Molecular Genetic and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St LouisSaint LouisUnited States
- Department of Ophthalmology and Visual Sciences, Washington University in St LouisSaint LouisUnited States
- Department of Developmental Biology, Washington University in St LouisSaint LouisUnited States
| |
Collapse
|
10
|
Zheng Y, Sun C, Zhang X, Ruzycki PA, Chen S. Missense mutations in CRX homeodomain cause dominant retinopathies through two distinct mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526652. [PMID: 36778408 PMCID: PMC9915647 DOI: 10.1101/2023.02.01.526652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Homeodomain transcription factors (HD TFs) are instrumental to vertebrate development. Mutations in HD TFs have been linked to human diseases, but their pathogenic mechanisms remain elusive. Here we use Cone-Rod Homeobox (CRX) as a model to decipher the disease-causing mechanisms of two HD mutations, p.E80A and p.K88N, that produce severe dominant retinopathies. Through integrated analysis of molecular and functional evidence in vitro and in knock-in mouse models, we uncover two novel gain-of-function mechanisms: p.E80A increases CRX-mediated transactivation of canonical CRX target genes in developing photoreceptors; p.K88N alters CRX DNA-binding specificity resulting in binding at ectopic sites and severe perturbation of CRX target gene expression. Both mechanisms produce novel retinal morphological defects and hinder photoreceptor maturation distinct from loss-of-function models. This study reveals the distinct roles of E80 and K88 residues in CRX HD regulatory functions and emphasizes the importance of transcriptional precision in normal development.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetic and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St Louis, Saint Louis, Missouri, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, USA
| | - Chi Sun
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, USA
| | - Xiaodong Zhang
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, USA
| | - Philip A. Ruzycki
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, USA
- Department of Genetics, Washington University in St Louis, Saint Louis, Missouri, USA
| | - Shiming Chen
- Molecular Genetic and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St Louis, Saint Louis, Missouri, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, USA
- Department of Developmental Biology, Washington University in St Louis, Saint Louis, Missouri, USA
| |
Collapse
|
11
|
Prokai L, Zaman K, Prokai-Tatrai K. Mass spectrometry-based retina proteomics. MASS SPECTROMETRY REVIEWS 2023; 42:1032-1062. [PMID: 35670041 PMCID: PMC9730434 DOI: 10.1002/mas.21786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
A subfield of neuroproteomics, retina proteomics has experienced a transformative growth since its inception due to methodological advances in enabling chemical, biochemical, and molecular biology techniques. This review focuses on mass spectrometry's contributions to facilitate mammalian and avian retina proteomics to catalog and quantify retinal protein expressions, determine their posttranslational modifications, as well as its applications to study the proteome of the retina in the context of biology, health and diseases, and therapy developments.
Collapse
Affiliation(s)
- Laszlo Prokai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Khadiza Zaman
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Katalin Prokai-Tatrai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
12
|
Harris N, Bates SG, Zhuang Z, Bernstein M, Stonemetz JM, Hill TJ, Yu YV, Calarco JA, Sengupta P. Molecular encoding of stimulus features in a single sensory neuron type enables neuronal and behavioral plasticity. Curr Biol 2023; 33:1487-1501.e7. [PMID: 36977417 PMCID: PMC10133190 DOI: 10.1016/j.cub.2023.02.073] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Neurons modify their transcriptomes in response to an animal's experience. How specific experiences are transduced to modulate gene expression and precisely tune neuronal functions are not fully defined. Here, we describe the molecular profile of a thermosensory neuron pair in C. elegans experiencing different temperature stimuli. We find that distinct salient features of the temperature stimulus, including its duration, magnitude of change, and absolute value, are encoded in the gene expression program in this single neuron type, and we identify a novel transmembrane protein and a transcription factor whose specific transcriptional dynamics are essential to drive neuronal, behavioral, and developmental plasticity. Expression changes are driven by broadly expressed activity-dependent transcription factors and corresponding cis-regulatory elements that nevertheless direct neuron- and stimulus-specific gene expression programs. Our results indicate that coupling of defined stimulus characteristics to the gene regulatory logic in individual specialized neuron types can customize neuronal properties to drive precise behavioral adaptation.
Collapse
Affiliation(s)
- Nathan Harris
- Department of Biology, MS008, Brandeis University, 415 South Street, Waltham, MA 02454, USA.
| | - Samuel G Bates
- Department of Biology, MS008, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Zihao Zhuang
- Department of Biology, MS008, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Matthew Bernstein
- Department of Biology, MS008, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Jamie M Stonemetz
- Department of Biology, MS008, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Tyler J Hill
- Department of Biology, MS008, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Yanxun V Yu
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei Province, China
| | - John A Calarco
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord St., Toronto, ON M5S 3G5, Canada
| | - Piali Sengupta
- Department of Biology, MS008, Brandeis University, 415 South Street, Waltham, MA 02454, USA.
| |
Collapse
|
13
|
Barret D, Schuster D, Rodrigues M, Leitner A, Picotti P, Schertler G, Kaupp U, Korkhov V, Marino J. Structural basis of calmodulin modulation of the rod cyclic nucleotide-gated channel. Proc Natl Acad Sci U S A 2023; 120:e2300309120. [PMID: 37011209 PMCID: PMC10104587 DOI: 10.1073/pnas.2300309120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/06/2023] [Indexed: 04/05/2023] Open
Abstract
Calmodulin (CaM) regulates many ion channels to control calcium entry into cells, and mutations that alter this interaction are linked to fatal diseases. The structural basis of CaM regulation remains largely unexplored. In retinal photoreceptors, CaM binds to the CNGB subunit of cyclic nucleotide-gated (CNG) channels and, thereby, adjusts the channel's Cyclic guanosine monophosphate (cGMP) sensitivity in response to changes in ambient light conditions. Here, we provide the structural characterization for CaM regulation of a CNG channel by using a combination of single-particle cryo-electron microscopy and structural proteomics. CaM connects the CNGA and CNGB subunits, resulting in structural changes both in the cytosolic and transmembrane regions of the channel. Cross-linking and limited proteolysis-coupled mass spectrometry mapped the conformational changes induced by CaM in vitro and in the native membrane. We propose that CaM is a constitutive subunit of the rod channel to ensure high sensitivity in dim light. Our mass spectrometry-based approach is generally relevant for studying the effect of CaM on ion channels in tissues of medical interest, where only minute quantities are available.
Collapse
Affiliation(s)
- Diane C. A. Barret
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
| | - Dina Schuster
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049Zürich, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zürich, 8049Zurich, Switzerland
| | - Matthew J. Rodrigues
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
| | - Alexander Leitner
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049Zürich, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, 8049Zürich, Switzerland
| | | | - U. Benjamin Kaupp
- Life and Medical Sciences Institute, University of Bonn, 53115Bonn, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077Göttingen, Germany
| | - Volodymyr M. Korkhov
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
- Institute of Molecular Biology and Biophysics, ETH Zürich, 8049Zurich, Switzerland
| | - Jacopo Marino
- Laboratory of Biomolecular Research, Paul Scherrer Institute, 5232Villigen, Switzerland
| |
Collapse
|
14
|
Harris N, Bates S, Zhuang Z, Bernstein M, Stonemetz J, Hill T, Yu YV, Calarco JA, Sengupta P. Molecular encoding of stimulus features in a single sensory neuron type enables neuronal and behavioral plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525070. [PMID: 36711719 PMCID: PMC9882311 DOI: 10.1101/2023.01.22.525070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Neurons modify their transcriptomes in response to an animal’s experience. How specific experiences are transduced to modulate gene expression and precisely tune neuronal functions are not fully defined. Here, we describe the molecular profile of a thermosensory neuron pair in C. elegans experiencing different temperature stimuli. We find that distinct salient features of the temperature stimulus including its duration, magnitude of change, and absolute value are encoded in the gene expression program in this single neuron, and identify a novel transmembrane protein and a transcription factor whose specific transcriptional dynamics are essential to drive neuronal, behavioral, and developmental plasticity. Expression changes are driven by broadly expressed activity-dependent transcription factors and corresponding cis -regulatory elements that nevertheless direct neuron- and stimulus-specific gene expression programs. Our results indicate that coupling of defined stimulus characteristics to the gene regulatory logic in individual specialized neuron types can customize neuronal properties to drive precise behavioral adaptation.
Collapse
Affiliation(s)
- Nathan Harris
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Samuel Bates
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Zihao Zhuang
- Department of Biology, Brandeis University, Waltham, MA, USA
- Current address: Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | | | - Jamie Stonemetz
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Tyler Hill
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Yanxun V. Yu
- Department of Neurology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - John A. Calarco
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, MA, USA
| |
Collapse
|
15
|
Han X, Zhang L, Tang J, Wang Z, Li S, Yuan L, Qu J. Correlation of photoreceptor damage with anti-retina antibodies level in aqueous humor in macular edema patients. Sci Rep 2022; 12:21212. [PMID: 36481862 PMCID: PMC9732343 DOI: 10.1038/s41598-022-25875-y] [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: 07/26/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
This study aimed to investigate the correlation between the severity of photoreceptor damage and the level of anti-retina antibodies (ARAs) in aqueous humor, including recoverin, CA II and enolase-α IgG antibody of macular edema patients. Aqueous humor samples were collected from macular edema patients and from cataract patients. Patients were divided into three groups according to the severity of discontinuity of ellipsoid zone (EZ) shown on optical coherence tomography (OCT) imaging: cataract patients with intact EZ, macular edema patients with mild EZ damage, and macular edema patients with severe EZ damage. The level of ARAs was determined with enzyme-linked immunosorbent assay (ELISA). The correlation between the level of ARAs and the degree of photoreceptor damage was analyzed. The level of ARAs of the intact EZ group was significantly lower than that in the severely damaged group (P < 0.05). The level of recoverin IgG of the intact EZ group was significantly lower than mildly damaged group (P = 0.030). In a subgroup analysis, the level of recoverin IgG of DME patients was correlated with their central retinal thickness (CRT) (r = 0.462, P = 0.035). The level of ARAs in aqueous humor of patients with DME and RVO-ME was correlated with the degree of photoreceptor damage.
Collapse
Affiliation(s)
- Xinyao Han
- grid.411634.50000 0004 0632 4559Department of Ophthalmology, Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Linqi Zhang
- grid.411634.50000 0004 0632 4559Department of Ophthalmology, Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Jiyang Tang
- grid.411634.50000 0004 0632 4559Department of Ophthalmology, Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Zongyi Wang
- grid.411634.50000 0004 0632 4559Department of Ophthalmology, Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Siying Li
- grid.411634.50000 0004 0632 4559Department of Ophthalmology, Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Li Yuan
- grid.411634.50000 0004 0632 4559Department of Ophthalmology, Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Jinfeng Qu
- grid.411634.50000 0004 0632 4559Department of Ophthalmology, Eye Diseases and Optometry Institute, Peking University People’s Hospital, Beijing, China ,grid.11135.370000 0001 2256 9319Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| |
Collapse
|
16
|
Vladimirov VI, Shchannikova MP, Baldin AV, Kazakov AS, Shevelyova MP, Nazipova AA, Baksheeva VE, Nemashkalova EL, Frolova AS, Tikhomirova NK, Philippov PP, Zamyatnin AA, Permyakov SE, Zinchenko DV, Zernii EY. Redox Regulation of Signaling Complex between Caveolin-1 and Neuronal Calcium Sensor Recoverin. Biomolecules 2022; 12:1698. [PMID: 36421712 PMCID: PMC9687869 DOI: 10.3390/biom12111698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 09/29/2023] Open
Abstract
Caveolin-1 is a cholesterol-binding scaffold protein, which is localized in detergent-resistant membrane (DRM) rafts and interacts with components of signal transduction systems, including visual cascade. Among these components are neuronal calcium sensors (NCSs), some of which are redox-sensitive proteins that respond to calcium signals by modulating the activity of multiple intracellular targets. Here, we report that the formation of the caveolin-1 complex with recoverin, a photoreceptor NCS serving as the membrane-binding regulator of rhodopsin kinase (GRK1), is a redox-dependent process. Biochemical and biophysical in vitro experiments revealed a two-fold decreased affinity of recoverin to caveolin-1 mutant Y14E mimicking its oxidative stress-induced phosphorylation of the scaffold protein. At the same time, wild-type caveolin-1 demonstrated a 5-10-fold increased affinity to disulfide dimer of recoverin (dRec) or its thiol oxidation mimicking the C39D mutant. The formation of dRec in vitro was not affected by caveolin-1 but was significantly potentiated by zinc, the well-known mediator of redox homeostasis. In the MDCK cell model, oxidative stress indeed triggered Y14 phosphorylation of caveolin-1 and disulfide dimerization of recoverin. Notably, oxidative conditions promoted the accumulation of phosphorylated caveolin-1 in the plasma membrane and the recruitment of recoverin to the same sites. Co-localization of these proteins was preserved upon depletion of intracellular calcium, i.e., under conditions reducing membrane affinity of recoverin but favoring its interaction with caveolin-1. Taken together, these data suggest redox regulation of the signaling complex between recoverin and caveolin-1. During oxidative stress, the high-affinity interaction of thiol-oxidized recoverin with caveolin-1/DRMs may disturb the light-induced translocation of the former within photoreceptors and affect rhodopsin desensitization.
Collapse
Affiliation(s)
- Vasiliy I. Vladimirov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Margarita P. Shchannikova
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Alexey V. Baldin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Alexey S. Kazakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Marina P. Shevelyova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Aliya A. Nazipova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Viktoriia E. Baksheeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Ekaterina L. Nemashkalova
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Anastasia S. Frolova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Natalia K. Tikhomirova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Pavel P. Philippov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Andrey A. Zamyatnin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Sergei E. Permyakov
- Institute for Biological Instrumentation, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Pushchino 142290, Russia
| | - Dmitry V. Zinchenko
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290, Russia
| | - Evgeni Yu. Zernii
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| |
Collapse
|
17
|
Rehman S, Gora AH, Varshney S, Dias J, Olsvik PA, Fernandes JMO, Brugman S, Kiron V. Developmental defects and behavioral changes in a diet-induced inflammation model of zebrafish. Front Immunol 2022; 13:1018768. [PMID: 36389790 PMCID: PMC9643868 DOI: 10.3389/fimmu.2022.1018768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022] Open
Abstract
Soybean meal evokes diet-induced intestinal inflammation in certain fishes. Although the molecular aspects of soybean-induced intestinal inflammation in zebrafish are known, the impact of the inflammatory diet on fish behavior remain largely underexplored. We fed zebrafish larvae with three diets - control, soybean meal and soybean meal with β-glucan to gain deeper insight into the behavioral changes associated with the soybean meal-induced inflammation model. We assessed the effect of the diets on the locomotor behavior, morphological development, oxygen consumption and larval transcriptome. Our study revealed that dietary soybean meal can reduce the locomotor activity, induce developmental defects and increase the oxygen demand in zebrafish larvae. Transcriptomic analysis pointed to the suppression of genes linked to visual perception, organ development, phototransduction pathway and activation of genes linked to the steroid biosynthesis pathway. On the contrary, β-glucan, an anti-inflammatory feed additive, counteracted the behavioral and phenotypic changes linked to dietary soybean. Although we did not identify any differentially expressed genes from the soybean meal alone fed group vs soybean meal + β-glucan-fed group comparison, the unique genes from the comparisons of the two groups with the control likely indicate reduction in inflammatory cytokine signaling, inhibition of proteolysis and induction of epigenetic modifications by the dietary glucan. Furthermore, we found that feeding an inflammatory diet at the larval stage can lead to long-lasting developmental defects. In conclusion, our study reveals the extra-intestinal manifestations associated with soybean meal-induced inflammation model.
Collapse
Affiliation(s)
- Saima Rehman
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Adnan H. Gora
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Shubham Varshney
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | - Pål A. Olsvik
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | - Sylvia Brugman
- Department of Animal Sciences, Host Microbe Interactomics, Wageningen University, Wageningen, Netherlands
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
- *Correspondence: Viswanath Kiron,
| |
Collapse
|
18
|
Chrispell JD, Xiong Y, Weiss ER. Grk7 but not Grk1 undergoes cAMP-dependent phosphorylation in zebrafish cone photoreceptors and mediates cone photoresponse recovery to elevated cAMP. J Biol Chem 2022; 298:102636. [PMID: 36273582 PMCID: PMC9692042 DOI: 10.1016/j.jbc.2022.102636] [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: 07/19/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022] Open
Abstract
In the vertebrate retina, phosphorylation of photoactivated visual pigments in rods and cones by G protein-coupled receptor kinases (GRKs) is essential for sustained visual function. Previous in vitro analysis demonstrated that GRK1 and GRK7 are phosphorylated by PKA, resulting in a reduced capacity to phosphorylate rhodopsin. In vivo observations revealed that GRK phosphorylation occurs in the dark and is cAMP dependent. In many vertebrates, including humans and zebrafish, GRK1 is expressed in both rods and cones while GRK7 is expressed only in cones. However, mice express only GRK1 in both rods and cones and lack GRK7. We recently generated a mutation in Grk1 that deletes the phosphorylation site, Ser21. This mutant demonstrated delayed dark adaptation in mouse rods but not in cones in vivo, suggesting GRK1 may serve a different role depending upon the photoreceptor cell type in which it is expressed. Here, zebrafish were selected to evaluate the role of cAMP-dependent GRK phosphorylation in cone photoreceptor recovery. Electroretinogram analyses of larvae treated with forskolin show that elevated intracellular cAMP significantly decreases recovery of the cone photoresponse, which is mediated by Grk7a rather than Grk1b. Using a cone-specific dominant negative PKA transgene, we show for the first time that PKA is required for Grk7a phosphorylation in vivo. Lastly, immunoblot analyses of rod grk1a-/- and cone grk1b-/- zebrafish and Nrl-/- mouse show that cone-expressed Grk1 does not undergo cAMP-dependent phosphorylation in vivo. These results provide a better understanding of the function of Grk phosphorylation relative to cone adaptation and recovery.
Collapse
|
19
|
Barret DC, Kaupp UB, Marino J. The structure of cyclic nucleotide-gated channels in rod and cone photoreceptors. Trends Neurosci 2022; 45:763-776. [DOI: 10.1016/j.tins.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/06/2022] [Accepted: 07/19/2022] [Indexed: 10/16/2022]
|
20
|
Ziaka K, van der Spuy J. The Role of Hsp90 in Retinal Proteostasis and Disease. Biomolecules 2022; 12:biom12070978. [PMID: 35883534 PMCID: PMC9313453 DOI: 10.3390/biom12070978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/24/2022] Open
Abstract
Photoreceptors are sensitive neuronal cells with great metabolic demands, as they are responsible for carrying out visual phototransduction, a complex and multistep process that requires the exquisite coordination of a large number of signalling protein components. Therefore, the viability of photoreceptors relies on mechanisms that ensure a well-balanced and functional proteome that maintains the protein homeostasis, or proteostasis, of the cell. This review explores how the different isoforms of Hsp90, including the cytosolic Hsp90α/β, the mitochondrial TRAP1, and the ER-specific GRP94, are involved in the different proteostatic mechanisms of photoreceptors, and elaborates on Hsp90 function when retinal homeostasis is disturbed. In addition, several studies have shown that chemical manipulation of Hsp90 has significant consequences, both in healthy and degenerating retinae, and this can be partially attributed to the fact that Hsp90 interacts with important photoreceptor-associated client proteins. Here, the interaction of Hsp90 with the retina-specific client proteins PDE6 and GRK1 will be further discussed, providing additional insights for the role of Hsp90 in retinal disease.
Collapse
|
21
|
Zinc Modulation of Neuronal Calcium Sensor Proteins: Three Modes of Interaction with Different Structural Outcomes. Biomolecules 2022; 12:biom12070956. [PMID: 35883512 PMCID: PMC9312857 DOI: 10.3390/biom12070956] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/06/2023] Open
Abstract
Neuronal calcium sensors (NCSs) are the family of EF-hand proteins mediating Ca2+-dependent signaling pathways in healthy neurons and neurodegenerative diseases. It was hypothesized that the calcium sensor activity of NCSs can be complemented by sensing fluctuation of intracellular zinc, which could further diversify their function. Here, using a set of biophysical techniques, we analyzed the Zn2+-binding properties of five proteins belonging to three different subgroups of the NCS family, namely, VILIP1 and neurocalcin-δ/NCLD (subgroup B), recoverin (subgroup C), as well as GCAP1 and GCAP2 (subgroup D). We demonstrate that each of these proteins is capable of coordinating Zn2+ with a different affinity, stoichiometry, and structural outcome. In the absence of calcium, recoverin and VILIP1 bind two zinc ions with submicromolar affinity, and the binding induces pronounced conformational changes and regulates the dimeric state of these proteins without significant destabilization of their structure. In the presence of calcium, recoverin binds zinc with slightly decreased affinity and moderate conformational outcome, whereas VILIP1 becomes insensitive to Zn2+. NCALD binds Zn2+ with micromolar affinity, but the binding induces dramatic destabilization and aggregation of the protein. In contrast, both GCAPs demonstrate low-affinity binding of zinc independent of calcium, remaining relatively stable even at submillimolar Zn2+ concentrations. Based on these data, and the results of structural bioinformatics analysis, NCSs can be divided into three categories: (1) physiological Ca2+/Zn2+ sensor proteins capable of binding exchangeable (signaling) zinc (recoverin and VILIP1), (2) pathological Ca2+/Zn2+ sensors responding only to aberrantly high free zinc concentrations by denaturation and aggregation (NCALD), and (3) Zn2+-resistant, Ca2+ sensor proteins (GCAP1, GCAP2). We suggest that NCS proteins may therefore govern the interconnection between Ca2+-dependent and Zn2+-dependent signaling pathways in healthy neurons and zinc cytotoxicity-related neurodegenerative diseases, such as Alzheimer’s disease and glaucoma.
Collapse
|
22
|
Zang J, Gesemann M, Keim J, Samardzija M, Grimm C, Neuhauss SCF. Circadian regulation of vertebrate cone photoreceptor function. eLife 2021; 10:e68903. [PMID: 34550876 PMCID: PMC8494479 DOI: 10.7554/elife.68903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
Eukaryotes generally display a circadian rhythm as an adaption to the reoccurring day/night cycle. This is particularly true for visual physiology that is directly affected by changing light conditions. Here we investigate the influence of the circadian rhythm on the expression and function of visual transduction cascade regulators in diurnal zebrafish and nocturnal mice. We focused on regulators of shut-off kinetics such as Recoverins, Arrestins, Opsin kinases, and Regulator of G-protein signaling that have direct effects on temporal vision. Transcript as well as protein levels of most analyzed genes show a robust circadian rhythm-dependent regulation, which correlates with changes in photoresponse kinetics. Electroretinography demonstrates that photoresponse recovery in zebrafish is delayed in the evening and accelerated in the morning. Functional rhythmicity persists in continuous darkness, and it is reversed by an inverted light cycle and disrupted by constant light. This is in line with our finding that orthologous gene transcripts from diurnal zebrafish and nocturnal mice are often expressed in an anti-phasic daily rhythm.
Collapse
Affiliation(s)
- Jingjing Zang
- University of Zurich, Department of Molecular Life SciencesZurichSwitzerland
| | - Matthias Gesemann
- University of Zurich, Department of Molecular Life SciencesZurichSwitzerland
| | - Jennifer Keim
- University of Zurich, Department of Molecular Life SciencesZurichSwitzerland
| | - Marijana Samardzija
- Lab for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of ZurichZurichSwitzerland
| | - Christian Grimm
- Lab for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of ZurichZurichSwitzerland
| | - Stephan CF Neuhauss
- University of Zurich, Department of Molecular Life SciencesZurichSwitzerland
| |
Collapse
|
23
|
Ahrens N, Aeissen E, Lippe A, Janssen-Bienhold U, Christoffers J, Koch KW. Farnesylation of Zebrafish G-Protein-Coupled Receptor Kinase Using Bio-orthogonal Labeling. ACS Chem Neurosci 2021; 12:1824-1832. [PMID: 33945258 DOI: 10.1021/acschemneuro.1c00155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
G-protein-coupled receptors are deactivated or desensitized by phosphorylation by respective G-protein-coupled receptor kinases (GRKs). In zebrafish rod and cone photoreceptor cells, four orthologous GRKs are expressed participating in the deactivation of rod and cone opsins. An important feature of GRKs in general is the consensus sites for lipid modification, which would allow the posttranslational attachment of isoprenoids facilitating membrane association and enzymatic performance. Because direct proof is missing for isoprenoid modification of zebrafish GRKs, we used a semichemical approach to study the incorporation of a farnesyl moiety into a GRK and its cellular consequences. The approach involves organic synthesis of a functionalized farnesyl derivative that is suitable for a subsequent alkyne-azide cycloaddition (click reaction). For this purpose, zebrafish GRK was expressed in HEK293 cells and modified in situ with the synthetic farnesyl moiety. Successful farnesylation by an endogenous farnesyltransferase was detected by immunoblotting and immunocytochemistry using a biotin-streptavidin-coupled assay and ligation with a fluorescence dye, respectively. Immunocytochemical detection of farnesylated GRK in different cell compartments indicates the applicability of the approach for studying the transport of cellular components.
Collapse
Affiliation(s)
- Nicole Ahrens
- Division of Biochemistry, Department of Neurosciences, Carl von Ossietzky Universität Oldenburg, D-26111 Oldenburg, Germany
| | - Enno Aeissen
- Institut für Chemie, Carl von Ossietzky Universität Oldenburg, D-26111 Oldenburg, Germany
| | - Anka Lippe
- Institut für Chemie, Carl von Ossietzky Universität Oldenburg, D-26111 Oldenburg, Germany
| | - Ulrike Janssen-Bienhold
- Division of Neurobiology, Department of Neurosciences, Carl von Ossietzky Universität Oldenburg, D-26111 Oldenburg, Germany
| | - Jens Christoffers
- Institut für Chemie, Carl von Ossietzky Universität Oldenburg, D-26111 Oldenburg, Germany
| | - Karl-Wilhelm Koch
- Division of Biochemistry, Department of Neurosciences, Carl von Ossietzky Universität Oldenburg, D-26111 Oldenburg, Germany
| |
Collapse
|
24
|
Functional modulation of phosphodiesterase-6 by calcium in mouse rod photoreceptors. Sci Rep 2021; 11:8938. [PMID: 33903621 PMCID: PMC8076185 DOI: 10.1038/s41598-021-88140-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/07/2021] [Indexed: 02/02/2023] Open
Abstract
Phosphodiesterase-6 (PDE6) is a key protein in the G-protein cascade converting photon information to bioelectrical signals in vertebrate photoreceptor cells. Here, we demonstrate that PDE6 is regulated by calcium, contrary to the common view that PDE1 is the unique PDE class whose activity is modulated by intracellular Ca2+. To broaden the operating range of photoreceptors, mammalian rod photoresponse recovery is accelerated mainly by two calcium sensor proteins: recoverin, modulating the lifetime of activated rhodopsin, and guanylate cyclase-activating proteins (GCAPs), regulating the cGMP synthesis. We found that decreasing rod intracellular Ca2+ concentration accelerates the flash response recovery and increases the basal PDE6 activity (βdark) maximally by ~ 30% when recording local electroretinography across the rod outer segment layer from GCAPs-/- recoverin-/- mice. Our modeling shows that a similar elevation in βdark can fully explain the observed acceleration of flash response recovery in low Ca2+. Additionally, a reduction of the free Ca2+ in GCAPs-/- recoverin-/- rods shifted the inhibition constants of competitive PDE inhibitor 3-isobutyl-1-methylxanthine (IBMX) against the thermally activated and light-activated forms of PDE6 to opposite directions, indicating a complex interaction between IBMX, PDE6, and calcium. The discovered regulation of PDE6 is a previously unknown mechanism in the Ca2+-mediated modulation of rod light sensitivity.
Collapse
|
25
|
Migocka-Patrzałek M, Elias M. Muscle Glycogen Phosphorylase and Its Functional Partners in Health and Disease. Cells 2021; 10:cells10040883. [PMID: 33924466 PMCID: PMC8070155 DOI: 10.3390/cells10040883] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 02/07/2023] Open
Abstract
Glycogen phosphorylase (PG) is a key enzyme taking part in the first step of glycogenolysis. Muscle glycogen phosphorylase (PYGM) differs from other PG isoforms in expression pattern and biochemical properties. The main role of PYGM is providing sufficient energy for muscle contraction. However, it is expressed in tissues other than muscle, such as the brain, lymphoid tissues, and blood. PYGM is important not only in glycogen metabolism, but also in such diverse processes as the insulin and glucagon signaling pathway, insulin resistance, necroptosis, immune response, and phototransduction. PYGM is implicated in several pathological states, such as muscle glycogen phosphorylase deficiency (McArdle disease), schizophrenia, and cancer. Here we attempt to analyze the available data regarding the protein partners of PYGM to shed light on its possible interactions and functions. We also underline the potential for zebrafish to become a convenient and applicable model to study PYGM functions, especially because of its unique features that can complement data obtained from other approaches.
Collapse
|
26
|
Zang J, Neuhauss SCF. Biochemistry and physiology of zebrafish photoreceptors. Pflugers Arch 2021; 473:1569-1585. [PMID: 33598728 PMCID: PMC8370914 DOI: 10.1007/s00424-021-02528-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
All vertebrates share a canonical retina with light-sensitive photoreceptors in the outer retina. These photoreceptors are of two kinds: rods and cones, adapted to low and bright light conditions, respectively. They both show a peculiar morphology, with long outer segments, comprised of ordered stacks of disc-shaped membranes. These discs host numerous proteins, many of which contribute to the visual transduction cascade. This pathway converts the light stimulus into a biological signal, ultimately modulating synaptic transmission. Recently, the zebrafish (Danio rerio) has gained popularity for studying the function of vertebrate photoreceptors. In this review, we introduce this model system and its contribution to our understanding of photoreception with a focus on the cone visual transduction cascade.
Collapse
Affiliation(s)
- Jingjing Zang
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrase 190, CH - 8057, Zürich, Switzerland
| | - Stephan C F Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrase 190, CH - 8057, Zürich, Switzerland.
| |
Collapse
|
27
|
Regulation of retinal membrane guanylyl cyclase (RetGC) by negative calcium feedback and RD3 protein. Pflugers Arch 2021; 473:1393-1410. [PMID: 33537894 PMCID: PMC8329130 DOI: 10.1007/s00424-021-02523-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 11/07/2022]
Abstract
This article presents a brief overview of the main biochemical and cellular processes involved in regulation of cyclic GMP production in photoreceptors. The main focus is on how the fluctuations of free calcium concentrations in photoreceptors between light and dark regulate the activity of retinal membrane guanylyl cyclase (RetGC) via calcium sensor proteins. The emphasis of the review is on the structure of RetGC and guanylyl cyclase activating proteins (GCAPs) in relation to their functional role in photoreceptors and congenital diseases of photoreceptors. In addition to that, the structure and function of retinal degeneration-3 protein (RD3), which regulates RetGC in a calcium-independent manner, is discussed in detail in connections with its role in photoreceptor biology and inherited retinal blindness.
Collapse
|
28
|
Marino V, Riva M, Zamboni D, Koch KW, Dell'Orco D. Bringing the Ca 2+ sensitivity of myristoylated recoverin into the physiological range. Open Biol 2021; 11:200346. [PMID: 33401992 PMCID: PMC7881174 DOI: 10.1098/rsob.200346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The prototypical Ca2+-sensor protein recoverin (Rec) is thought to regulate the activity of rhodopsin kinase (GRK1) in photoreceptors by switching from a relaxed (R) disc membrane-bound conformation in the dark to a more compact, cytosol-diffusing tense (T) conformation upon cell illumination. However, the apparent affinity for Ca2+ of its physiologically relevant form (myristoylated recoverin) is almost two orders of magnitude too low to support this mechanism in vivo. In this work, we compared the individual and synergistic roles of the myristic moiety, the GRK1 target and the disc membrane in modulating the calcium sensitivity of Rec. We show that the sole presence of the target or the disc membrane alone are not sufficient to achieve a physiological response to changes in intracellular [Ca2+]. Instead, the simultaneous presence of GRK1 and membrane allows the T to R transition to occur in a physiological range of [Ca2+] with high cooperativity via a conformational selection mechanism that drives the structural transitions of Rec in the presence of multiple ligands. Our conclusions may apply to other sensory transduction systems involving protein complexes and biological membranes.
Collapse
Affiliation(s)
- Valerio Marino
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Matteo Riva
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.,Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| | - Davide Zamboni
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.,Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| | - Karl-Wilhelm Koch
- Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| | - Daniele Dell'Orco
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
| |
Collapse
|
29
|
Stockman A, Henning GB, Rider AT. Clinical vision and molecular loss: Integrating visual psychophysics with molecular genetics reveals key details of normal and abnormal visual processing. Prog Retin Eye Res 2020; 83:100937. [PMID: 33388434 DOI: 10.1016/j.preteyeres.2020.100937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 01/08/2023]
Abstract
Over the past two decades we have developed techniques and models to investigate the ways in which known molecular defects affect visual performance. Because molecular defects in retinal signalling invariably alter the speed of visual processing, our strategy has been to measure the resulting changes in flicker sensitivity. Flicker measurements provide not only straightforward clinical assessments of visual performance but also reveal fundamental details about the functioning of both abnormal and normal visual systems. Here, we bring together our past measurements of patients with pathogenic variants in the GNAT2, RGS9, GUCA1A, RPE65, OPA1, KCNV2 and NR2E3 genes and analyse the results using a standard model of visual processing. The model treats flicker sensitivity as the result of the actions of a sequence of simple processing steps, one or more of which is altered by the genetic defect. Our analyses show that most defects slow down the visual response directly, but some speed it up. Crucially, however, other steps in the processing sequence can make compensatory adjustments to offset the abnormality. For example, if the abnormal step slows down the visual response, another step is likely to speed up or attenuate the response to rebalance system performance. Such compensatory adjustments are probably made by steps in the sequence that usually adapt to changing light levels. Our techniques and modelling also allow us to tease apart stationary and progressive effects, and the localised molecular losses help us to unravel and characterise individual steps in the normal and abnormal processing sequences.
Collapse
Affiliation(s)
- Andrew Stockman
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK.
| | - G Bruce Henning
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK
| | - Andrew T Rider
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, England, UK
| |
Collapse
|
30
|
Ahrens N, Elbers D, Greb H, Janssen-Bienhold U, Koch KW. Interaction of G protein-coupled receptor kinases and recoverin isoforms is determined by localization in zebrafish photoreceptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118946. [PMID: 33385424 DOI: 10.1016/j.bbamcr.2020.118946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
The zebrafish retina expresses four recoverin genes (rcv1a, rcv1b, rcv2a and rcv2b) and four opsin kinase genes (grk1a, grk1b, grk7a and grk7b) coding for recoverin and G protein-coupled receptor kinase (opsin kinase) paralogs, respectively. Both protein groups are suggested to form regulatory complexes in rod and cone outer segments, but at present, we lack information about co-localization of recoverin and opsin kinases in zebrafish retinae and which protein-protein interacting pairs form. We analyzed the distribution and co-localization of recoverin and opsin kinase expression in the zebrafish retina. For this purpose, we used custom-tailored monospecific antibodies revealing that the amount of recoverin paralogs in a zebrafish retina can differ by more than one order of magnitude with the highest amount for recoverin 1a and 2b. Further, immunohistochemical labelling showed presence of recoverin 1a in all rod cell compartments, but it only co-localized with opsin kinase 1a in rod outer segments. In contrast, recoverin 2b was only detected in double cones and co-localized with opsin kinases 1b, 7a and 7b. Further, we investigated the interaction between recoverin and opsin kinase variants by surface plasmon resonance spectroscopy indicating interaction of recoverin 1a and recoverin 2b with all opsin kinases. However, binding kinetics for recoverin 1a differed from those observed with recoverin 2b that showed slower association and dissociation processes. Our results indicate diverse recoverin and opsin kinase properties due to differential expression and interaction profiles.
Collapse
Affiliation(s)
- Nicole Ahrens
- Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| | - Dana Elbers
- Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| | - Helena Greb
- Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| | - Ulrike Janssen-Bienhold
- Department of Neuroscience, Division of Neurobiology, University of Oldenburg, 26111 Oldenburg, Germany
| | - Karl-Wilhelm Koch
- Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany.
| |
Collapse
|
31
|
Rhodopsin-mediated light-off-induced protein kinase A activation in mouse rod photoreceptor cells. Proc Natl Acad Sci U S A 2020; 117:26996-27003. [PMID: 33046651 DOI: 10.1073/pnas.2009164117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Light-induced extrasynaptic dopamine release in the retina reduces adenosine 3',5'-cyclic monophosphate (cAMP) in rod photoreceptor cells, which is thought to mediate light-dependent desensitization. However, the fine time course of the cAMP dynamics in rods remains elusive due to technical difficulty. Here, we visualized the spatiotemporal regulation of cAMP-dependent protein kinase (PKA) in mouse rods by two-photon live imaging of retinal explants of PKAchu mice, which express a fluorescent biosensor for PKA. Unexpectedly, in addition to the light-on-induced suppression, we observed prominent light-off-induced PKA activation. This activation required photopic light intensity and was confined to the illuminated rods. The estimated maximum spectral sensitivity of 489 nm and loss of the light-off-induced PKA activation in rod-transducin-knockout retinas strongly suggest the involvement of rhodopsin. In support of this notion, rhodopsin-deficient retinal explants showed only the light-on-induced PKA suppression. Taken together, these results suggest that, upon photopic light stimulation, rhodopsin and dopamine signals are integrated to shape the light-off-induced cAMP production and following PKA activation. This may support the dark adaptation of rods.
Collapse
|
32
|
Vladimirov VI, Baksheeva VE, Mikhailova IV, Ismailov RG, Litus EA, Tikhomirova NK, Nazipova AA, Permyakov SE, Zernii EY, Zinchenko DV. A Novel Approach to Bacterial Expression and Purification of Myristoylated Forms of Neuronal Calcium Sensor Proteins. Biomolecules 2020; 10:biom10071025. [PMID: 32664359 PMCID: PMC7407513 DOI: 10.3390/biom10071025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 11/18/2022] Open
Abstract
N-terminal myristoylation is a common co-and post-translational modification of numerous eukaryotic and viral proteins, which affects their interaction with lipids and partner proteins, thereby modulating various cellular processes. Among those are neuronal calcium sensor (NCS) proteins, mediating transduction of calcium signals in a wide range of regulatory cascades, including reception, neurotransmission, neuronal growth and survival. The details of NCSs functioning are of special interest due to their involvement in the progression of ophthalmological and neurodegenerative diseases and their role in cancer. The well-established procedures for preparation of native-like myristoylated forms of recombinant NCSs via their bacterial co-expression with N-myristoyl transferase from Saccharomyces cerevisiae often yield a mixture of the myristoylated and non-myristoylated forms. Here, we report a novel approach to preparation of several NCSs, including recoverin, GCAP1, GCAP2, neurocalcin δ and NCS-1, ensuring their nearly complete N-myristoylation. The optimized bacterial expression and myristoylation of the NCSs is followed by a set of procedures for separation of their myristoylated and non-myristoylated forms using a combination of hydrophobic interaction chromatography steps. We demonstrate that the refolded and further purified myristoylated NCS-1 maintains its Са2+-binding ability and stability of tertiary structure. The developed approach is generally suited for preparation of other myristoylated proteins.
Collapse
Affiliation(s)
- Vasiliy I. Vladimirov
- Laboratory of pharmacokinetics, Department of Biological Testing, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences in Puschino, Pushchino, 142290 Moscow Region, Russia; (V.I.V.); (I.V.M.)
| | - Viktoriia E. Baksheeva
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (N.K.T.); (E.Y.Z.)
| | - Irina V. Mikhailova
- Laboratory of pharmacokinetics, Department of Biological Testing, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences in Puschino, Pushchino, 142290 Moscow Region, Russia; (V.I.V.); (I.V.M.)
- Faculty of BioMedPharmTechnological, Pushchino State Institute of Natural Sciences, Pushchino, 142290 Moscow Region, Russia
| | - Ramis G. Ismailov
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, 142290 Moscow Region, Russia; (R.G.I.); (E.A.L.); (A.A.N.); (S.E.P.)
| | - Ekaterina A. Litus
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, 142290 Moscow Region, Russia; (R.G.I.); (E.A.L.); (A.A.N.); (S.E.P.)
| | - Natalia K. Tikhomirova
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (N.K.T.); (E.Y.Z.)
| | - Aliya A. Nazipova
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, 142290 Moscow Region, Russia; (R.G.I.); (E.A.L.); (A.A.N.); (S.E.P.)
| | - Sergei E. Permyakov
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino, 142290 Moscow Region, Russia; (R.G.I.); (E.A.L.); (A.A.N.); (S.E.P.)
| | - Evgeni Yu. Zernii
- Department of Cell Signaling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia; (V.E.B.); (N.K.T.); (E.Y.Z.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Dmitry V. Zinchenko
- Laboratory of pharmacokinetics, Department of Biological Testing, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences in Puschino, Pushchino, 142290 Moscow Region, Russia; (V.I.V.); (I.V.M.)
- Correspondence:
| |
Collapse
|
33
|
Borsatto A, Marino V, Abrusci G, Lattanzi G, Dell'Orco D. Effects of Membrane and Biological Target on the Structural and Allosteric Properties of Recoverin: A Computational Approach. Int J Mol Sci 2019; 20:ijms20205009. [PMID: 31658639 PMCID: PMC6829511 DOI: 10.3390/ijms20205009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/01/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Recoverin (Rec) is a prototypical calcium sensor protein primarily expressed in the vertebrate retina. The binding of two Ca2+ ions to the functional EF-hand motifs induces the extrusion of a myristoyl group that increases the affinity of Rec for the membrane and leads to the formation of a complex with rhodopsin kinase (GRK1). Here, unbiased all-atom molecular dynamics simulations were performed to monitor the spontaneous insertion of the myristoyl group into a model multicomponent biological membrane for both isolated Rec and for its complex with a peptide from the GRK1 target. It was found that the functional membrane anchoring of the myristoyl group is triggered by persistent electrostatic protein-membrane interactions. In particular, salt bridges between Arg43, Arg46 and polar heads of phosphatidylserine lipids are necessary to enhance the myristoyl hydrophobic packing in the Rec-GRK1 assembly. The long-distance communication between Ca2+-binding EF-hands and residues at the interface with GRK1 is significantly influenced by the presence of the membrane, which leads to dramatic changes in the connectivity of amino acids mediating the highest number of persistent interactions (hubs). In conclusion, specific membrane composition and allosteric interactions are both necessary for the correct assembly and dynamics of functional Rec-GRK1 complex.
Collapse
Affiliation(s)
- Alberto Borsatto
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
- Department of Physics, University of Trento, 38123 Trento, Italy.
| | - Valerio Marino
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56026 Pisa, Italy.
| | - Gianfranco Abrusci
- Department of Physics, University of Trento, 38123 Trento, Italy.
- Trento Institute for Fundamental Physics and Applications (INFN-TIFPA), Via Sommarive 14, Povo, 38123 Trento, Italy.
| | - Gianluca Lattanzi
- Department of Physics, University of Trento, 38123 Trento, Italy.
- Trento Institute for Fundamental Physics and Applications (INFN-TIFPA), Via Sommarive 14, Povo, 38123 Trento, Italy.
| | - Daniele Dell'Orco
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
| |
Collapse
|
34
|
Roy K, Marin EP. Lipid Modifications in Cilia Biology. J Clin Med 2019; 8:jcm8070921. [PMID: 31252577 PMCID: PMC6678300 DOI: 10.3390/jcm8070921] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/17/2022] Open
Abstract
Cilia are specialized cellular structures with distinctive roles in various signaling cascades. Ciliary proteins need to be trafficked to the cilium to function properly; however, it is not completely understood how these proteins are delivered to their final localization. In this review, we will focus on how different lipid modifications are important in ciliary protein trafficking and, consequently, regulation of signaling pathways. Lipid modifications can play a variety of roles, including tethering proteins to the membrane, aiding trafficking through facilitating interactions with transporter proteins, and regulating protein stability and abundance. Future studies focusing on the role of lipid modifications of ciliary proteins will help our understanding of how cilia maintain specific protein pools strictly connected to their functions.
Collapse
Affiliation(s)
- Kasturi Roy
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, PO Box 208029, New Haven, CT 06520-8029, USA.
| | - Ethan P Marin
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, PO Box 208029, New Haven, CT 06520-8029, USA
| |
Collapse
|