1
|
Song HB, Campello L, Mondal A, Chen HY, English MA, Glen M, Vanlandingham P, Farjo R, Swaroop A. Sex-specific attenuation of photoreceptor degeneration by reserpine in a rhodopsin P23H rat model of autosomal dominant retinitis pigmentosa. eLife 2025; 14:RP103888. [PMID: 40231721 PMCID: PMC11999695 DOI: 10.7554/elife.103888] [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] [Indexed: 04/16/2025] Open
Abstract
Inherited retinal degenerations (IRDs) constitute a group of clinically and genetically diverse vision-impairing disorders. Retinitis pigmentosa (RP), the most common form of IRD, is characterized by gradual dysfunction and degeneration of rod photoreceptors, followed by the loss of cone photoreceptors. Recently, we identified reserpine as a lead molecule for maintaining rod survival in mouse and human retinal organoids as well as in the rd16 mouse, which phenocopy Leber congenital amaurosis caused by mutations in the cilia-centrosomal gene CEP290 (Chen et al., 2023). Here, we show the therapeutic potential of reserpine in a rhodopsin P23H rat model of autosomal dominant RP. At postnatal day (P) 68, when males and females are analyzed together, the reserpine-treated rats exhibit higher rod-derived scotopic b-wave amplitudes compared to the controls with little or no change in scotopic a-wave or cone-derived photopic b-wave. Interestingly, the reserpine-treated female rats display enhanced scotopic a- and b-waves and photopic b-wave responses at P68, along with a better contrast threshold and increased outer nuclear layer thickness. The female rats demonstrate better preservation of both rod and cone photoreceptors following reserpine treatment. Retinal transcriptome analysis reveals sex-specific responses to reserpine, with significant upregulation of phototransduction genes and proteostasis-related pathways, and notably, genes associated with stress response. This study builds upon our previously reported results reaffirming the potential of reserpine for gene-agnostic treatment of IRDs and emphasizes the importance of biological sex in retinal disease research and therapy development.
Collapse
Affiliation(s)
- Hyun Beom Song
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Laura Campello
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Anupam Mondal
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Holly Y Chen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Milton A English
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Michael Glen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | | | | | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
2
|
Xia L, Wang H, Du G, Cheng X, Zhang R, Yu H, Cheng M, Chen Y, Qin S, Leng W. Receptor accessory protein 6, a novel ferroptosis suppressor, drives oral squamous cell carcinoma by maintaining endoplasmic reticulum hemostasis. Int J Biol Macromol 2024; 283:137565. [PMID: 39566754 DOI: 10.1016/j.ijbiomac.2024.137565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
Increasing evidence suggests a close association between endoplasmic-reticulum (ER) stress and ferroptosis. Receptor accessory protein 6 (REEP6) is known to play a crucial role in maintaining ER homeostasis. However, its involvement in ferroptosis remains unknown. In this study, we found that REEP6 was overexpressed, and its overexpression showed a significant association with tumor size and poor survival in OSCC patients. Besides, in vitro and in vivo assays together showed that REEP6 plays an oncogenic role in OSCC progression. The GO/KEGG, and GSEA analysis showed that REEP6 overexpression leads to the inactivation of ferroptosis signaling in OSCC. Moreover, REEP6 overexpression conferred resistance to RSL3, a ferroptosis inducer, whereas REEP6 knockdown sensitized OSCC cells to RSL3. Overexpression of REEP6 decrease the accumulation of iron ions, ROS production, but increase the number of mitochondrial cristae in OSCC cells. More importantly, we confirmed that REEP6 inhibited ferroptosis in OSCC cells by maintaining ER homeostasis via regulating ACSL4 expression. In addition, we identified promoter DNA hypomethylation as the underlying cause of REEP6 overexpression in OSCC. Taken together, REEP6 acts as a novel suppressor of ferroptosis, with its overexpression driven by promoter hypomethylation contributing to OSCC progression by ER stress-mediated ferroptosis via ACSL4.
Collapse
Affiliation(s)
- Lingyun Xia
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Hongbing Wang
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Gao Du
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Xiaobo Cheng
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Rui Zhang
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Hedong Yu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China
| | - Mumo Cheng
- Department of General Practice, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch, Fudan University), Shanghai 200940, China
| | - Yongji Chen
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China.
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China.
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China; Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, China.
| |
Collapse
|
3
|
Dey H, Perez-Hurtado M, Heidelberger R. Syntaxin 3B: A SNARE Protein Required for Vision. Int J Mol Sci 2024; 25:10665. [PMID: 39408994 PMCID: PMC11476516 DOI: 10.3390/ijms251910665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Syntaxin 3 is a member of a large protein family of syntaxin proteins that mediate fusion between vesicles and their target membranes. Mutations in the ubiquitously expressed syntaxin 3A splice form give rise to a serious gastrointestinal disorder in humans called microvillus inclusion disorder, while mutations that additionally involve syntaxin 3B, a splice form that is expressed primarily in retinal photoreceptors and bipolar cells, additionally give rise to an early onset severe retinal dystrophy. In this review, we discuss recent studies elucidating the roles of syntaxin 3B and the regulation of syntaxin 3B functionality in membrane fusion and neurotransmitter release in the vertebrate retina.
Collapse
Affiliation(s)
| | | | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (H.D.)
| |
Collapse
|
4
|
Kim SY, Park CH, Moon BH, Seabold GK. Murine Retina Outer Plexiform Layer Development and Transcriptome Analysis of Pre-Synapses in Photoreceptors. Life (Basel) 2024; 14:1103. [PMID: 39337887 PMCID: PMC11433150 DOI: 10.3390/life14091103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Photoreceptors in the mammalian retina convert light signals into electrical and molecular signals through phototransduction and transfer the visual inputs to second-order neurons via specialized ribbon synapses. Two kinds of photoreceptors, rods and cones, possess distinct morphology and function. Currently, we have limited knowledge about rod versus (vs.) cone synapse development and the associated genes. The transcription factor neural retina leucine zipper (NRL) determines the rod vs. cone photoreceptor cell fate and is critical for rod differentiation. Nrl knockout mice fail to form rods, generating all cone or S-cone-like (SCL) photoreceptors in the retina, whereas ectopic expression of Nrl using a cone-rod homeobox (Crx) promoter (CrxpNrl) forms all rods. Here, we examined rod and cone pre-synapse development, including axonal elongation, terminal shaping, and synaptic lamination in the outer plexiform layer (OPL) in the presence or absence of Nrl. We show that NRL loss and knockdown result in delayed OPL maturation and plasticity with aberrant dendrites of bipolar neurons. The integrated analyses of the transcriptome in developing rods and SCLs with NRL CUT&RUN and synaptic gene ontology analyses identified G protein subunit beta (Gnb) 1 and p21 (RAC1) activated kinase 5 (Pak5 or Pak7) transcripts were upregulated in developing rods and down-regulated in developing SCLs. Notably, Gnb1 and Gnb5 are rod dominant, and Gnb3 is enriched in cones. NRL binds to the genes of Gnb1, Gnb3, and Gnb5. NRL also regulates pre-synapse ribbon genes, and their expression is altered in rods and SCLs. Our study of histological and gene analyses provides new insights into the morphogenesis of photoreceptor pre-synapse development and regulation of associated genes in the developing retina.
Collapse
Affiliation(s)
- Soo-Young Kim
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine Haewon Park
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bo-Hyun Moon
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Gail K Seabold
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
5
|
Sundaresan Y, Yacoub S, Kodati B, Amankwa CE, Raola A, Zode G. Therapeutic applications of CRISPR/Cas9 gene editing technology for the treatment of ocular diseases. FEBS J 2023; 290:5248-5269. [PMID: 36877952 PMCID: PMC10480348 DOI: 10.1111/febs.16771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 02/04/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
Ocular diseases are a highly heterogeneous group of phenotypes, caused by a spectrum of genetic variants and environmental factors that exhibit diverse clinical symptoms. As a result of its anatomical location, structure and immune privilege, the eye is an ideal system to assess and validate novel genetic therapies. Advances in genome editing have revolutionized the field of biomedical science, enabling researchers to understand the biology behind disease mechanisms and allow the treatment of several health conditions, including ocular pathologies. The advent of clustered regularly interspaced short palindromic repeats (CRISPR)-based gene editing facilitates efficient and specific genetic modifications in the nucleic acid sequence, resulting in permanent changes at the genomic level. This approach has advantages over other treatment strategies and is promising for the treatment of various genetic and non-genetic ocular conditions. This review provides an overview of the CRISPR/CRISPR-associated protein 9 (Cas9) system and summarizes recent advances in the therapeutic application of CRISPR/Cas9 for the treatment of various ocular pathologies, as well as future challenges.
Collapse
Affiliation(s)
| | | | - Bindu Kodati
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Charles E. Amankwa
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Akash Raola
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Gulab Zode
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| |
Collapse
|
6
|
Heymann JB, Vijayasarathy C, Fariss RN, Sieving PA. Advances in understanding the molecular structure of retinoschisin while questions remain of biological function. Prog Retin Eye Res 2023; 95:101147. [PMID: 36402656 PMCID: PMC10185713 DOI: 10.1016/j.preteyeres.2022.101147] [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: 07/28/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022]
Abstract
Retinoschisin (RS1) is a secreted protein that is essential for maintaining integrity of the retina. Numerous mutations in RS1 cause X-linked retinoschisis (XLRS), a progressive degeneration of the retina that leads to vision loss in young males. A key manifestation of XLRS is the formation of cavities (cysts) in the retina and separation of the layers (schisis), disrupting synaptic transmission. There are currently no approved treatments for patients with XLRS. Strategies using adeno-associated viral (AAV) vectors to deliver functional copies of RS1 as a form of gene augmentation therapy, are under clinical evaluation. To improve therapeutic strategies for treating XLRS, it is critical to better understand the secretion of RS1 and its molecular function. Immunofluorescence and immunoelectron microscopy show that RS1 is located on the surfaces of the photoreceptor inner segments and bipolar cells. Sequence homology indicates a discoidin domain fold, similar to many other proteins with demonstrated adhesion functions. Recent structural studies revealed the tertiary structure of RS1 as two back-to-back octameric rings, each cross-linked by disulfides. The observation of higher order structures in vitro suggests the formation of an adhesive matrix spanning the distance between cells (∼100 nm). Several studies indicated that RS1 readily binds to other proteins such as the sodium-potassium ATPase (NaK-ATPase) and extracellular matrix proteins. Alternatively, RS1 may influence fluid regulation via interaction with membrane proteins such as the NaK-ATPase, largely inferred from the use of carbonic anhydrase inhibitors to shrink the typical intra-retinal cysts in XLRS. We discuss these models in light of RS1 structure and address the difficulty in understanding the function of RS1.
Collapse
Affiliation(s)
- J Bernard Heymann
- National Cryo-EM Program, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, 21701, USA.
| | - Camasamudram Vijayasarathy
- Section on Translational Research for Retinal and Macular Degeneration, NIDCD, NIH, Bethesda, MD, 20892, USA
| | - Robert N Fariss
- Biological Imaging Core Facility, NEI, NIH, Bethesda, MD, 20892, USA
| | - Paul A Sieving
- Center for Ocular Regenerative Therapy, Ophthalmology, U C Davis Health, Sacramento, CA, 95817, USA
| |
Collapse
|
7
|
Tseng CC, Hung CC, Shu CW, Lee CH, Chen CF, Kuo MS, Kao YY, Chen CL, Ger LP, Liu PF. The Clinical and Biological Effects of Receptor Expression-Enhancing Protein 6 in Tongue Squamous Cell Carcinoma. Biomedicines 2023; 11:biomedicines11051270. [PMID: 37238941 DOI: 10.3390/biomedicines11051270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/07/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
There are currently no effective biomarkers for the diagnosis and treatment of tongue squamous cell carcinoma (TSCC), which causes a poor 5-year overall survival rate. Thus, it is crucial to identify more effective diagnostic/prognostic biomarkers and therapeutic targets for TSCC patients. The receptor expression-enhancing protein 6 (REEP6), a transmembrane endoplasmic reticulum resident protein, controls the expression or transport of a subset of proteins or receptors. Although it was reported that REEP6 plays a role in lung and colon cancers, its clinical impact and biological role in TSCC are still unknown. The present study aimed to identify a novel effective biomarker and therapeutic target for TSCC patients. Expression levels of REEP6 in specimens from TSCC patients were determined with immunohistochemistry. Gene knockdown was used to evaluate the effects of REEP6 in cancer malignancy (colony/tumorsphere formation, cell cycle regulation, migration, drug resistance and cancer stemness) of TSCC cells. The clinical impact of REEP6 expression and gene co-expression on prognosis were analyzed in oral cancer patients including TSCC patients from The Cancer Genome Atlas database. Tumor tissues had higher levels of REEP6 compared to normal tissues in TSCC patients. Higher REEP6 expression was related to shorter disease-free survival (DFS) in oral cancer patients with poorly differentiated tumor cells. REEP6-knocked-down TSCC cells showed diminished colony/tumorsphere formation, and they also caused G1 arrest and decreased migration, drug resistance and cancer stemness. A high co-expression of REEP6/epithelial-mesenchymal transition or cancer stemness markers also resulted in poor DFS in oral cancer patients. Thus, REEP6 is involved in the malignancy of TSCC and might serve as a potential diagnostic/prognostic biomarker and therapeutic target for TSCC patients.
Collapse
Affiliation(s)
- Chung-Chih Tseng
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Department of Dentistry, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 81342, Taiwan
| | - Chung-Ching Hung
- Department of Otolaryngology, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 81342, Taiwan
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Cheng-Hsin Lee
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chun-Feng Chen
- Department of Stomatology, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Mei-Shu Kuo
- Department of Biotechnology, Chia Nan University, Tainan 71710, Taiwan
| | - Yu-Ying Kao
- Department of Biotechnology, Chia Nan University, Tainan 71710, Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Luo-Ping Ger
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| |
Collapse
|
8
|
Fan S, Liu H, Li L. The REEP family of proteins: molecular targets and role in pathophysiology. Pharmacol Res 2022; 185:106477. [PMID: 36191880 DOI: 10.1016/j.phrs.2022.106477] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/18/2022]
Abstract
Receptor expression-enhancing proteins (REEPs) are an evolutionarily conserved protein family that is pivotal to the structure and function of the endoplasmic reticulum (ER). The REEP family can be classified into two major subfamilies in higher species, the REEP1-4 and REEP5-6 subfamilies. Within the REEP1-4 subfamily, REEP1 and REEP2 are closely related, and REEP3 and REEP4 are similarly related. The REEP family is widely distributed in various tissues. Recent studies indicate that the REEP family is involved in many pathological and physiological processes, such as ER morphogenesis and remodeling, microtubule cytoskeleton regulation, and the trafficking and expression of G protein-coupled receptors (GPCRs). Moreover, the REEP family plays crucial roles in the occurrence and development of many diseases, including neurological diseases, diabetes, retinal diseases, cardiac diseases, infertility, obesity, oligoarticular juvenile idiopathic arthritis (OJIA), COVID-19, and cancer. In the present review, we describe the distribution and structure of the REEP family. Furthermore, we summarize the functions and the associated diseases of this family. Based on the pleiotropic actions of the REEP family, the study of its family members is crucial to understanding the relevant pathophysiological processes and developing strategies to modulate and control these related diseases. AVAILABILITY OF DATA AND MATERIAL: The datasets used or analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Sisi Fan
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Huimei Liu
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of tumor microenvironment responsive drug research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
9
|
Angelotti T. Exploring the eukaryotic Yip and REEP/Yop superfamily of membrane-shaping adapter proteins (MSAPs): A cacophony or harmony of structure and function? Front Mol Biosci 2022; 9:912848. [PMID: 36060263 PMCID: PMC9437294 DOI: 10.3389/fmolb.2022.912848] [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: 04/04/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Polytopic cargo proteins are synthesized and exported along the secretory pathway from the endoplasmic reticulum (ER), through the Golgi apparatus, with eventual insertion into the plasma membrane (PM). While searching for proteins that could enhance cell surface expression of olfactory receptors, a new family of proteins termed “receptor expression-enhancing proteins” or REEPs were identified. These membrane-shaping hairpin proteins serve as adapters, interacting with intracellular transport machinery, to regulate cargo protein trafficking. However, REEPs belong to a larger family of proteins, the Yip (Ypt-interacting protein) family, conserved in yeast and higher eukaryotes. To date, eighteen mammalian Yip family members, divided into four subfamilies (Yipf, REEP, Yif, and PRAF), have been identified. Yeast research has revealed many intriguing aspects of yeast Yip function, functions that have not completely been explored with mammalian Yip family members. This review and analysis will clarify the different Yip family nomenclature that have encumbered prior comparisons between yeast, plants, and eukaryotic family members, to provide a more complete understanding of their interacting proteins, membrane topology, organelle localization, and role as regulators of cargo trafficking and localization. In addition, the biological role of membrane shaping and sensing hairpin and amphipathic helical domains of various Yip proteins and their potential cellular functions will be described. Lastly, this review will discuss the concept of Yip proteins as members of a larger superfamily of membrane-shaping adapter proteins (MSAPs), proteins that both shape membranes via membrane-sensing and hairpin insertion, and well as act as adapters for protein-protein interactions. MSAPs are defined by their localization to specific membranes, ability to alter membrane structure, interactions with other proteins via specific domains, and specific interactions/effects on cargo proteins.
Collapse
|
10
|
Young BK, Ramakrishnan C, Ganjawala T, Wang P, Deisseroth K, Tian N. An uncommon neuronal class conveys visual signals from rods and cones to retinal ganglion cells. Proc Natl Acad Sci U S A 2021; 118:e2104884118. [PMID: 34702737 PMCID: PMC8612366 DOI: 10.1073/pnas.2104884118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 01/01/2023] Open
Abstract
Neurons in the central nervous system (CNS) are distinguished by the neurotransmitter types they release, their synaptic connections, morphology, and genetic profiles. To fully understand how the CNS works, it is critical to identify all neuronal classes and reveal their synaptic connections. The retina has been extensively used to study neuronal development and circuit formation. Here, we describe a previously unidentified interneuron in mammalian retina. This interneuron shares some morphological, physiological, and molecular features with retinal bipolar cells, such as receiving input from photoreceptors and relaying visual signals to retinal ganglion cells. It also shares some features with amacrine cells (ACs), particularly Aii-ACs, such as their neurite morphology in the inner plexiform layer, the expression of some AC-specific markers, and possibly the release of the inhibitory neurotransmitter glycine. Thus, we unveil an uncommon interneuron, which may play an atypical role in vision.
Collapse
Affiliation(s)
- Brent K Young
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84114
| | | | - Tushar Ganjawala
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202
| | - Ping Wang
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| | - Ning Tian
- Department of Ophthalmology & Visual Sciences, University of Utah, Salt Lake City, UT 84132;
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84114
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84132
- Veterans Affairs Medical Center, Salt Lake City, UT 84148
| |
Collapse
|
11
|
Liang Q, Wu N, Zaneveld S, Liu H, Fu S, Wang K, Bertrand R, Wang J, Li Y, Chen R. Transcript isoforms of Reep6 have distinct functions in the retina. Hum Mol Genet 2021; 30:1907-1918. [PMID: 34104971 DOI: 10.1093/hmg/ddab157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/21/2022] Open
Abstract
Much of the complexity of the eukaryotic cell transcriptome is due to the alternative splicing of mRNA. However, knowledge on how transcriptome complexity is translated into functional complexity remains limited. For example, although different isoforms of a gene may show distinct temporal and spatial expression patterns, it is largely unknown whether these isoforms encode proteins with distinct functions matching their expression pattern. In this report, we investigated the function and relationship of the two isoforms of Reep6, namely Reep6.1 and Reep6.2, in rod photoreceptor cells. These two isoforms result from the alternative splicing of exon 5 and show mutually exclusive expression patterns. Reep6.2 is the canonical isoform that is expressed in non-retinal tissues while Reep6.1 is the only expressed isoform in the adult retina. The Reep6.1 isoform-specific knockout mouse, Reep6E5/E5, is generated by deleting exon 5 and a homozygous deletion phenotypically displayed a rod degeneration phenotype comparable to a Reep6 full knockout mouse, indicating that the Reep6.1 isoform is essential for the rod photoreceptor cell survival. Consistent with the results obtained from a loss-of-function experiment, overexpression of Reep6.2 failed to rescue the rod degeneration phenotype of Reep6 knockout mice while overexpression of Reep6.1 does lead to rescue. These results demonstrate that, consistent with the expression pattern of the isoform, Reep6.1 has rod-specific functions that cannot be substituted by its canonical isoform. Our findings suggested that a strict regulation of splicing is required for the maintenance of photoreceptor cells.
Collapse
Affiliation(s)
- Qingnan Liang
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Nathaniel Wu
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Smriti Zaneveld
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Hehe Liu
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Shangyi Fu
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Keqing Wang
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Renae Bertrand
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Jun Wang
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Yumei Li
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Rui Chen
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, 77030, USA
| |
Collapse
|
12
|
Zhang L, Li Y, Qin L, Wu Y, Lei B. Autosomal Recessive Retinitis Pigmentosa Associated with Three Novel REEP6 Variants in Chinese Population. Genes (Basel) 2021; 12:genes12040537. [PMID: 33917198 PMCID: PMC8068040 DOI: 10.3390/genes12040537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 11/28/2022] Open
Abstract
Retinitis pigmentosa 77 is caused by mutations of REEP6 (MIM: 609346), which encodes a protein for the development of photoreceptors. Our study was to identify disease-causing variants in three Chinese families using targeted next-generation sequencing (NGS). Multiple lines of computational predictions combined with in vitro cellular experiments were applied to evaluate the pathogenicity of the newly found variants. Three novel variants in REEP6, including one missense variant, c.268G>C, one frameshift variant, c.468delC, and one splicing variant, c.598+1G>C, were found, while c.268G>C was detected in all probands. The three variants were classified as likely pathogenic by the American College of Medical Genetics and Genomics (ACMG). REEP6 variant proteins c.268G>C and c.468delC in cultured cells destabilized the REEP6 protein and induced intracellular inclusions. Our data suggested that REEP6 c.268G>C may be a recurrent causative variant in Chinese autosomal recessive retinitis pigmentosa patients.
Collapse
Affiliation(s)
- Lujia Zhang
- Graduate School, Xinxiang Medical University, Xinxiang 453003, China;
- Henan Clinical Research Center for Ophthalmic Diseases, Henan Branch of National Clinical Research Center for Ocular Diseases, Henan Eye Institute, Henan Eye Hospital, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou 450003, China;
| | - Ya Li
- Henan Clinical Research Center for Ophthalmic Diseases, Henan Branch of National Clinical Research Center for Ocular Diseases, Henan Eye Institute, Henan Eye Hospital, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou 450003, China;
| | - Litao Qin
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China;
| | - Yu Wu
- Shanghai Flash Interpretation Biotechnology, Shanghai 201615, China;
| | - Bo Lei
- Henan Clinical Research Center for Ophthalmic Diseases, Henan Branch of National Clinical Research Center for Ocular Diseases, Henan Eye Institute, Henan Eye Hospital, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou 450003, China;
- Correspondence:
| |
Collapse
|
13
|
Syntaxin 3 is essential for photoreceptor outer segment protein trafficking and survival. Proc Natl Acad Sci U S A 2020; 117:20615-20624. [PMID: 32778589 DOI: 10.1073/pnas.2010751117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Trafficking of photoreceptor membrane proteins from their site of synthesis in the inner segment (IS) to the outer segment (OS) is critical for photoreceptor function and vision. Here we evaluate the role of syntaxin 3 (STX3), in trafficking of OS membrane proteins such as peripherin 2 (PRPH2) and rhodopsin. Photoreceptor-specific Stx3 knockouts [Stx3 f/f(iCre75) and Stx3 f/f(CRX-Cre) ] exhibited rapid, early-onset photoreceptor degeneration and functional decline characterized by structural defects in IS, OS, and synaptic terminals. Critically, in the absence of STX3, OS proteins such as PRPH2, the PRPH2 binding partner, rod outer segment membrane protein 1 (ROM1), and rhodopsin were mislocalized along the microtubules to the IS, cell body, and synaptic region. We find that the PRPH2 C-terminal domain interacts with STX3 as well as other photoreceptor SNAREs, and our findings indicate that STX3 is an essential part of the trafficking pathway for both disc (rhodopsin) and rim (PRPH2/ROM1) components of the OS.
Collapse
|
14
|
Shi X, Hai L, Govindasamy K, Gao J, Coppens I, Hu J, Wang Q, Bhanot P. A Plasmodium homolog of ER tubule-forming proteins is required for parasite virulence. Mol Microbiol 2020; 114:454-467. [PMID: 32432369 DOI: 10.1111/mmi.14526] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 01/27/2023]
Abstract
Reticulon and REEP family of proteins stabilize the high curvature of endoplasmic reticulum (ER) tubules. Plasmodium berghei Yop1 (PbYop1) is a REEP5 homolog in Plasmodium. Here, we characterize its function using a gene-knockout (Pbyop1∆). Pbyop1∆ asexual stage parasites display abnormal ER architecture and an enlarged digestive vacuole. The erythrocytic cycle of Pbyop1∆ parasites is severely attenuated and the incidence of experimental cerebral malaria is significantly decreased in Pbyop1∆-infected mice. Pbyop1∆ sporozoites have reduced speed, are slower to invade host cells but give rise to equal numbers of infected HepG2 cells, as WT sporozoites. We propose that PbYOP1's disruption may lead to defects in trafficking and secretion of a subset of proteins required for parasite development and invasion of erythrocytes. Furthermore, the maintenance of ER morphology in different parasite stages is likely to depend on different proteins.
Collapse
Affiliation(s)
- Xiaoyu Shi
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Lei Hai
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Kavitha Govindasamy
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jian Gao
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Junjie Hu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Qian Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China.,National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Purnima Bhanot
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
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: 59] [Impact Index Per Article: 11.8] [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
|
Lee SH, Hadipour-Lakmehsari S, Murthy HR, Gibb N, Miyake T, Teng ACT, Cosme J, Yu JC, Moon M, Lim S, Wong V, Liu P, Billia F, Fernandez-Gonzalez R, Stagljar I, Sharma P, Kislinger T, Scott IC, Gramolini AO. REEP5 depletion causes sarco-endoplasmic reticulum vacuolization and cardiac functional defects. Nat Commun 2020; 11:965. [PMID: 32075961 PMCID: PMC7031342 DOI: 10.1038/s41467-019-14143-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
The sarco-endoplasmic reticulum (SR/ER) plays an important role in the development and progression of many heart diseases. However, many aspects of its structural organization remain largely unknown, particularly in cells with a highly differentiated SR/ER network. Here, we report a cardiac enriched, SR/ER membrane protein, REEP5 that is centrally involved in regulating SR/ER organization and cellular stress responses in cardiac myocytes. In vitro REEP5 depletion in mouse cardiac myocytes results in SR/ER membrane destabilization and luminal vacuolization along with decreased myocyte contractility and disrupted Ca2+ cycling. Further, in vivo CRISPR/Cas9-mediated REEP5 loss-of-function zebrafish mutants show sensitized cardiac dysfunction upon short-term verapamil treatment. Additionally, in vivo adeno-associated viral (AAV9)-induced REEP5 depletion in the mouse demonstrates cardiac dysfunction. These results demonstrate the critical role of REEP5 in SR/ER organization and function as well as normal heart function and development.
Collapse
Affiliation(s)
- Shin-Haw Lee
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Sina Hadipour-Lakmehsari
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Harsha R Murthy
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Natalie Gibb
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Tetsuaki Miyake
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
- Department of Biology, Faculty of Science, York University, Toronto, ON, M3J1P3, Canada
| | - Allen C T Teng
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Jake Cosme
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Jessica C Yu
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S3G9, Canada
| | - Mark Moon
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
- Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - SangHyun Lim
- Donnelly Centre, University of Toronto, Toronto, ON, M5S1M8, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto Canada, Toronto, Canada
| | - Victoria Wong
- Donnelly Centre, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Peter Liu
- Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Filio Billia
- Toronto General Research Institute, University Health Network, Toronto, ON, M5G2C4, Canada
| | - Rodrigo Fernandez-Gonzalez
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S3G9, Canada
| | - Igor Stagljar
- Donnelly Centre, University of Toronto, Toronto, ON, M5S1M8, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto Canada, Toronto, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Canada
- Mediterranean Institute for Life Sciences, Split, Croatia
| | - Parveen Sharma
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
- Institute of Translational Medicine, University of Liverpool, Liverpool, L693BX, UK
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, Toronto, ON, M5G1L7, Canada
| | - Ian C Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada.
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada.
| |
Collapse
|
17
|
Cataloguing and Selection of mRNAs Localized to Dendrites in Neurons and Regulated by RNA-Binding Proteins in RNA Granules. Biomolecules 2020; 10:biom10020167. [PMID: 31978946 PMCID: PMC7072219 DOI: 10.3390/biom10020167] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022] Open
Abstract
Spatiotemporal translational regulation plays a key role in determining cell fate and function. Specifically, in neurons, local translation in dendrites is essential for synaptic plasticity and long-term memory formation. To achieve local translation, RNA-binding proteins in RNA granules regulate target mRNA stability, localization, and translation. To date, mRNAs localized to dendrites have been identified by comprehensive analyses. In addition, mRNAs associated with and regulated by RNA-binding proteins have been identified using various methods in many studies. However, the results obtained from these numerous studies have not been compiled together. In this review, we have catalogued mRNAs that are localized to dendrites and are associated with and regulated by the RNA-binding proteins fragile X mental retardation protein (FMRP), RNA granule protein 105 (RNG105, also known as Caprin1), Ras-GAP SH3 domain binding protein (G3BP), cytoplasmic polyadenylation element binding protein 1 (CPEB1), and staufen double-stranded RNA binding proteins 1 and 2 (Stau1 and Stau2) in RNA granules. This review provides comprehensive information on dendritic mRNAs, the neuronal functions of mRNA-encoded proteins, the association of dendritic mRNAs with RNA-binding proteins in RNA granules, and the effects of RNA-binding proteins on mRNA regulation. These findings provide insights into the mechanistic basis of protein-synthesis-dependent synaptic plasticity and memory formation and contribute to future efforts to understand the physiological implications of local regulation of dendritic mRNAs in neurons.
Collapse
|
18
|
Joe MK, Li W, Hiriyanna S, Yu W, Shah SA, Abu-Asab M, Qian H, Wu Z. A Common Outer Retinal Change in Retinal Degeneration by Optical Coherence Tomography Can Be Used to Assess Outcomes of Gene Therapy. Hum Gene Ther 2019; 30:1520-1530. [PMID: 31672061 DOI: 10.1089/hum.2019.162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Identifying early disease hallmarks in animal models with slow disease progression may expedite disease detection and assessment of treatment outcomes. Using optical coherence tomography, a widely applied noninvasive method for monitoring retinal structure changes, we analyzed retinal optical sections from six mouse lines with retinal degeneration caused by mutations in different disease-causing genes. While images from wild-type mice revealed four well-separated hyper-reflective bands in the outer retina (designated as outer retina reflective bands, ORRBs) at all ages, the second band (ORRB2) and the third band (ORRB3) were merged in retinas of five mutant mouse lines at early ages, suggesting the pathological nature of this alteration. This ORRB change appeared to be degenerating photoreceptor related, and occurred before obvious morphological changes that can be identified on both hematoxylin and eosin-stained sections and electron microscopic sections. Importantly, the merging of ORRB2 and ORRB3 was reversed by treatment with adeno-associated viral vector-mediated gene replacement therapies, and this restoration occurred much earlier than measurable functional or structural improvement. Our data suggest that the ORRB change could be a common hallmark of early retinal degeneration and its restoration could be used for rapid and noninvasive assessment of therapeutic effects following gene therapy or other treatment interventions.
Collapse
Affiliation(s)
- Myung Kuk Joe
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Wenbo Li
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Suja Hiriyanna
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Wenhan Yu
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Shreya A Shah
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Mones Abu-Asab
- Histopathology Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Zhijian Wu
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
19
|
Lin Y, Xu CL, Velez G, Yang J, Tanaka AJ, Breazzano MP, Mahajan VB, Sparrow JR, Tsang SH. Novel REEP6 gene mutation associated with autosomal recessive retinitis pigmentosa. Doc Ophthalmol 2019; 140:67-75. [PMID: 31538292 DOI: 10.1007/s10633-019-09719-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/04/2019] [Indexed: 01/06/2023]
Abstract
PURPOSE This study reports the ophthalmic and genetic findings of a Cameroonian patient with autosomal recessive retinitis pigmentosa (arRP) caused by a novel Receptor Expression Enhancing Protein 6 (REEP6) homozygous mutation. PATIENT AND METHODS A 33-year-old man underwent comprehensive ophthalmic examinations, including visual acuity measurements, dilated fundus imaging, electroretinography (ERG), and spectral-domain optical coherence tomography (SD-OCT). Short-wavelength fundus autofluorescence (SW-AF) and near-infrared fundus autofluorescence (NIR-AF) were also evaluated. Whole exome sequencing (WES) was used to identify potential pathogenic variants. RESULTS Fundus examination revealed typical RP findings with additional temporal ten micron yellow dots. SD-OCT imaging revealed cystoid macular edema and perifoveal outer retinal atrophy with centrally preserved inner segment ellipsoid zone (EZ) bands. Hyperreflective spots were seen in the inner retinal layers. On SW-AF images, a hypoautofluorescent area in the perifoveal area was observed. NIR-AF imaging revealed an irregularly shaped hyperautofluorescent ring. His visual acuity was mildly affected. ERG showed undetectable rod responses and intact cone responses. Genetic testing via WES revealed a novel homozygous mutation (c.295G>A, p.Glu99Lys) in the gene encoding REEP6, which is predicted to alter the charge in the transmembrane helix. CONCLUSIONS This report is not only the first description of a Cameroonian patient with arRP associated with a REEP6 mutation, but also this particular genetic alteration. Substitution of p.Glu99Lys in REEP6 likely disrupts the interactions between REEP6 and the ER membrane. NIR-AF imaging may be particularly useful for assessing functional photoreceptor cells and show an "avocado" pattern of hyperautofluorescence in patients with the REEP6 mutation.
Collapse
Affiliation(s)
- Yuchen Lin
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Eye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Christine L Xu
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Gabriel Velez
- Omics Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA.,Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - Jing Yang
- Omics Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Akemi J Tanaka
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Mark P Breazzano
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.,Department of Ophthalmology, New York University School of Medicine, New York, NY, USA
| | - Vinit B Mahajan
- Omics Laboratory, Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Janet R Sparrow
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA. .,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
20
|
Méjécase C, Mohand-Saïd S, El Shamieh S, Antonio A, Condroyer C, Blanchard S, Letexier M, Saraiva JP, Sahel JA, Audo I, Zeitz C. A novel nonsense variant in REEP6 is involved in a sporadic rod-cone dystrophy case. Clin Genet 2019; 93:707-711. [PMID: 29120066 DOI: 10.1111/cge.13171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 12/22/2022]
Abstract
Rod-cone dystrophy (RCD), also called retinitis pigmentosa, is the most common form of progressive inherited retinal disorders secondary to photoreceptor degeneration. It is a genetically heterogeneous disease characterized by night blindness, followed by visual field constriction and, in most severe cases, total blindness. The aim of our study was to identify the underlying gene defect leading to severe RCD in a 60-year-old woman. The patient's DNA was investigated by targeted next generation sequencing followed by whole exome sequencing. A novel nonsense variant, c.267G>A p.(Trp89*), was identified at a homozygous state in the proband in REEP6 gene, recently reported mutated in 7 unrelated families with RCD. Further functional studies will help to understand the physiopathology associated with REEP6 mutations that may be linked to a protein trafficking defect.
Collapse
Affiliation(s)
- C Méjécase
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - S Mohand-Saïd
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC 1423, Paris, France
| | - S El Shamieh
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - A Antonio
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC 1423, Paris, France
| | - C Condroyer
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - S Blanchard
- IntegraGen SA, Genopole Campus, Evry, France
| | - M Letexier
- IntegraGen SA, Genopole Campus, Evry, France
| | - J-P Saraiva
- IntegraGen SA, Genopole Campus, Evry, France
| | - J-A Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC 1423, Paris, France.,Institute of Ophthalmology, University College of London, London, UK.,Fondation Ophtalmologique Adolphe de Rothschild, Paris, France.,Academie des Sciences, Institut de France, Paris, France.,Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburg, Pennsylvania, USA
| | - I Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC 1423, Paris, France.,Institute of Ophthalmology, University College of London, London, UK
| | - C Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
21
|
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
|
22
|
Hitti RJ, Oliver JAC, Schofield EC, Bauer A, Kaukonen M, Forman OP, Leeb T, Lohi H, Burmeister LM, Sargan D, Mellersh CS. Whole Genome Sequencing of Giant Schnauzer Dogs with Progressive Retinal Atrophy Establishes NECAP1 as a Novel Candidate Gene for Retinal Degeneration. Genes (Basel) 2019; 10:genes10050385. [PMID: 31117272 PMCID: PMC6562617 DOI: 10.3390/genes10050385] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/08/2019] [Accepted: 05/17/2019] [Indexed: 12/30/2022] Open
Abstract
Canine progressive retinal atrophies (PRA) are genetically heterogeneous diseases characterized by retinal degeneration and subsequent blindness. PRAs are untreatable and affect multiple dog breeds, significantly impacting welfare. Three out of seven Giant Schnauzer (GS) littermates presented with PRA around four years of age. We sought to identify the causal variant to improve our understanding of the aetiology of this form of PRA and to enable development of a DNA test. Whole genome sequencing of two PRA-affected full-siblings and both unaffected parents was performed. Variants were filtered based on those segregating appropriately for an autosomal recessive disorder and predicted to be deleterious. Successive filtering against 568 canine genomes identified a single nucleotide variant in the gene encoding NECAP endocytosis associated 1 (NECAP1): c.544G>A (p.Gly182Arg). Five thousand one hundred and thirty canids of 175 breeds, 10 cross-breeds and 3 wolves were genotyped for c.544G>A. Only the three PRA-affected GS were homozygous (allele frequency in GS, excluding proband family = 0.015). In addition, we identified heterozygotes belonging to Spitz and Dachshund varieties, demonstrating c.544G>A segregates in other breeds of German origin. This study, in parallel with the known retinal expression and role of NECAP1 in clathrin mediated endocytosis (CME) in synapses, presents NECAP1 as a novel candidate gene for retinal degeneration in dogs and other species.
Collapse
Affiliation(s)
- Rebekkah J Hitti
- Kennel Club Genetics Centre, Animal Health Trust, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK.
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK.
| | - James A C Oliver
- Kennel Club Genetics Centre, Animal Health Trust, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK.
| | - Ellen C Schofield
- Kennel Club Genetics Centre, Animal Health Trust, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK.
| | - Anina Bauer
- Institute of Genetics, University of Bern, 3001 Bern, Switzerland.
| | - Maria Kaukonen
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland.
- Department of Medical Genetics, University of Helsinki, 00014 Helsinki, Finland.
- Folkhälsan Research Center, 00290 Helsinki, Finland.
| | - Oliver P Forman
- Wisdom Health, Waltham-on-the-Wolds, Leicestershire LE14 4RS, UK.
| | - Tosso Leeb
- Institute of Genetics, University of Bern, 3001 Bern, Switzerland.
| | - Hannes Lohi
- Department of Veterinary Biosciences, University of Helsinki, 00014 Helsinki, Finland.
- Department of Medical Genetics, University of Helsinki, 00014 Helsinki, Finland.
- Folkhälsan Research Center, 00290 Helsinki, Finland.
| | - Louise M Burmeister
- Kennel Club Genetics Centre, Animal Health Trust, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK.
| | - David Sargan
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK.
| | - Cathryn S Mellersh
- Kennel Club Genetics Centre, Animal Health Trust, Lanwades Park, Newmarket, Suffolk CB8 7UU, UK.
| |
Collapse
|
23
|
Assawachananont J, Kim SY, Kaya KD, Fariss R, Roger JE, Swaroop A. Cone-rod homeobox CRX controls presynaptic active zone formation in photoreceptors of mammalian retina. Hum Mol Genet 2019; 27:3555-3567. [PMID: 30084954 DOI: 10.1093/hmg/ddy272] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
In the mammalian retina, rod and cone photoreceptors transmit the visual information to bipolar neurons through highly specialized ribbon synapses. We have limited understanding of regulatory pathways that guide morphogenesis and organization of photoreceptor presynaptic architecture in the developing retina. While neural retina leucine zipper (NRL) transcription factor determines rod cell fate and function, cone-rod homeobox (CRX) controls the expression of both rod- and cone-specific genes and is critical for terminal differentiation of photoreceptors. A comprehensive immunohistochemical evaluation of Crx-/- (null), CrxRip/+ and CrxRip/Rip (models of dominant congenital blindness) mouse retinas revealed abnormal photoreceptor synapses, with atypical ribbon shape, number and length. Integrated analysis of retinal transcriptomes of Crx-mutants with CRX- and NRL-ChIP-Seq data identified a subset of differentially expressed CRX target genes that encode presynaptic proteins associated with the cytomatrix active zone (CAZ) and synaptic vesicles. Immunohistochemistry of Crx-mutant retina validated aberrant expression of REEP6, PSD95, MPP4, UNC119, UNC13, RGS7 and RGS11, with some reduction in Ribeye and no significant change in immunostaining of RIMS1, RIMS2, Bassoon and Pikachurin. Our studies demonstrate that CRX controls the establishment of CAZ and anchoring of ribbons, but not the formation of ribbon itself, in photoreceptor presynaptic terminals.
Collapse
Affiliation(s)
- Juthaporn Assawachananont
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Soo-Young Kim
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Koray D Kaya
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert Fariss
- Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jerome E Roger
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Centre d'Etude et de Recherches Thérapeutiques en Ophthalmologie, Retina France, Orsay, France.,Paris-Saclay Institute of Neuroscience, CNRS, Univ Paris Sud, Université Paris-Saclay, Orsay, France
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Zaneveld SA, Eblimit A, Liang Q, Bertrand R, Wu N, Liu H, Nguyen Q, Zaneveld J, Wang K, Li Y, Chen R. Gene Therapy Rescues Retinal Degeneration in Receptor Expression-Enhancing Protein 6 Mutant Mice. Hum Gene Ther 2018; 30:302-315. [PMID: 30101608 PMCID: PMC6437630 DOI: 10.1089/hum.2018.078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hereditary retinal dystrophy is clinically defined as a broad group of chronic and progressive disorders that affect visual function by causing photoreceptor degeneration. Previously, we identified mutations in the gene encoding receptor expression-enhancing protein 6 (REEP6), in individuals with autosomal recessive retinitis pigmentosa (RP), the most common form of inherited retinal dystrophy. One individual was molecularly diagnosed with biallelic REEP6 mutations, a missense mutation over a frameshift mutation. In this study, we generated Reep6 compound heterozygous mice, Reep6L135P/-, which mimic the patient genotype and recapitulate the early-onset retinal degeneration phenotypes observed in the individual with RP. To determine the feasibility of rescuing the Reep6 mutant phenotype via gene replacement therapy, we delivered Reep6.1, the mouse retina-specific isoform of REEP6, to photoreceptors of Reep6 mutant mice on postnatal day 20. Evaluation of the therapeutic effects 2 months posttreatment showed improvements in the photoresponse as well as preservation of photoreceptor cells. Importantly, guanylyl cyclase 1 (GC1) expression was also restored to the outer segment after treatment. Furthermore, rAAV8-Reep6.1 single treatment in Reep6 mutant mice 1 year postinjection showed significant improvements in retinal function and morphology, suggesting that the treatment is effective even after a prolonged period. Findings from this study show that gene replacement therapy in the retina with rAAV overexpressing Reep6 is effective, preserving photoreceptor function in Reep6 mutant mice. These findings provide evidence that rAAV8-based gene therapy can prolong survival of photoreceptors in vivo and can be potentially used as a therapeutic modality for treatment of patients with RP.
Collapse
Affiliation(s)
- Smriti Agrawal Zaneveld
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Aiden Eblimit
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Qingnan Liang
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,3 Department of Biochemistry, Baylor College of Medicine, Houston, TX
| | - Renae Bertrand
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,3 Department of Biochemistry, Baylor College of Medicine, Houston, TX
| | - Nathaniel Wu
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Hehe Liu
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Quynh Nguyen
- 3 Department of Biochemistry, Baylor College of Medicine, Houston, TX
| | - Jacques Zaneveld
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Keqing Wang
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Yumei Li
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Rui Chen
- 1 Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX.,2 Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
25
|
Sarin S, Zuniga-Sanchez E, Kurmangaliyev YZ, Cousins H, Patel M, Hernandez J, Zhang KX, Samuel MA, Morey M, Sanes JR, Zipursky SL. Role for Wnt Signaling in Retinal Neuropil Development: Analysis via RNA-Seq and In Vivo Somatic CRISPR Mutagenesis. Neuron 2018; 98:109-126.e8. [PMID: 29576390 DOI: 10.1016/j.neuron.2018.03.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 01/16/2018] [Accepted: 03/02/2018] [Indexed: 12/22/2022]
Abstract
Screens for genes that orchestrate neural circuit formation in mammals have been hindered by practical constraints of germline mutagenesis. To overcome these limitations, we combined RNA-seq with somatic CRISPR mutagenesis to study synapse development in the mouse retina. Here synapses occur between cellular layers, forming two multilayered neuropils. The outer neuropil, the outer plexiform layer (OPL), contains synapses made by rod and cone photoreceptor axons on rod and cone bipolar dendrites, respectively. We used RNA-seq to identify selectively expressed genes encoding cell surface and secreted proteins and CRISPR-Cas9 electroporation with cell-specific promoters to assess their roles in OPL development. Among the genes identified in this way are Wnt5a and Wnt5b. They are produced by rod bipolars and activate a non-canonical signaling pathway in rods to regulate early OPL patterning. The approach we use here can be applied to other parts of the brain.
Collapse
Affiliation(s)
- Sumeet Sarin
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Elizabeth Zuniga-Sanchez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Henry Cousins
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Mili Patel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Jeanette Hernandez
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kelvin X Zhang
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Melanie A Samuel
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA
| | - Marta Morey
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02130, USA.
| | - S Lawrence Zipursky
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
26
|
Hoshino A, Ratnapriya R, Brooks MJ, Chaitankar V, Wilken MS, Zhang C, Starostik MR, Gieser L, La Torre A, Nishio M, Bates O, Walton A, Bermingham-McDonogh O, Glass IA, Wong ROL, Swaroop A, Reh TA. Molecular Anatomy of the Developing Human Retina. Dev Cell 2017; 43:763-779.e4. [PMID: 29233477 DOI: 10.1016/j.devcel.2017.10.029] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/15/2017] [Accepted: 10/26/2017] [Indexed: 02/07/2023]
Abstract
Clinical and genetic heterogeneity associated with retinal diseases makes stem-cell-based therapies an attractive strategy for personalized medicine. However, we have limited understanding of the timing of key events in the developing human retina, and in particular the factors critical for generating the unique architecture of the fovea and surrounding macula. Here we define three key epochs in the transcriptome dynamics of human retina from fetal day (D) 52 to 136. Coincident histological analyses confirmed the cellular basis of transcriptional changes and highlighted the dramatic acceleration of development in the fovea compared with peripheral retina. Human and mouse retinal transcriptomes show remarkable similarity in developmental stages, although morphogenesis was greatly expanded in humans. Integration of DNA accessibility data allowed us to reconstruct transcriptional networks controlling photoreceptor differentiation. Our studies provide insights into human retinal development and serve as a resource for molecular staging of human stem-cell-derived retinal organoids.
Collapse
Affiliation(s)
- Akina Hoshino
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA
| | - Rinki Ratnapriya
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew J Brooks
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vijender Chaitankar
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew S Wilken
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA
| | - Chi Zhang
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA
| | - Margaret R Starostik
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Linn Gieser
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anna La Torre
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA; Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Mario Nishio
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA
| | - Olivia Bates
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA
| | - Ashley Walton
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Ian A Glass
- Department of Pediatrics and Medicine, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|