1
|
Xiao W, Cheng J, Zhang L, Li X, Zhang J, Ning G. Deficiency of psmb6 suppresses proteasome activity and promotes apoptosis in larval zebrafish brain. Biochem Biophys Res Commun 2025; 755:151574. [PMID: 40048760 DOI: 10.1016/j.bbrc.2025.151574] [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: 02/19/2025] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
In humans, the abnormal accumulation of proteins is strongly correlated with neurodegenerative diseases. PSMB6 is a member of the proteasome family and encodes the 20S subunit beta 6 which performs caspase-like proteasome activity. However, the biological roles of PSMB6 in neurodevelopment are poorly defined. In this study, we utilized zebrafish to construct a psmb6 knockout model. We show that the deficiency of psmb6 leads to early embryonic death, with proteasome inactivity identified as the cause of neuronal apoptosis. Although the inactivation of p53 cannot rescue the defects observed in psmb6 mutants, it delays the onset of the defective phenotypes. Thus, psmb6 plays a crucial role in early embryonic development.
Collapse
Affiliation(s)
- Wei Xiao
- Zhanjiang Key Laboratory of Zebrafish Model for Development and Disease, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jie Cheng
- Zhanjiang Key Laboratory of Zebrafish Model for Development and Disease, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Liang Zhang
- Research Center of Translational Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, China
| | - Xiaoyu Li
- Zhanjiang Key Laboratory of Zebrafish Model for Development and Disease, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jingjing Zhang
- Zhanjiang Key Laboratory of Zebrafish Model for Development and Disease, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Guozhu Ning
- Zhanjiang Key Laboratory of Zebrafish Model for Development and Disease, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
2
|
Azam M, Jastrzebska B. Mechanisms of Rhodopsin-Related Inherited Retinal Degeneration and Pharmacological Treatment Strategies. Cells 2025; 14:49. [PMID: 39791750 PMCID: PMC11720364 DOI: 10.3390/cells14010049] [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: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary disease characterized by progressive vision loss ultimately leading to blindness. This condition is initiated by mutations in genes expressed in retinal cells, resulting in the degeneration of rod photoreceptors, which is subsequently followed by the loss of cone photoreceptors. Mutations in various genes expressed in the retina are associated with RP. Among them, mutations in the rhodopsin gene (RHO) are the most common cause of this condition. Due to the involvement of numerous genes and multiple mutations in a single gene, RP is a highly heterogeneous disease making the development of effective treatments particularly challenging. The progression of this disease involves complex cellular responses to restore cellular homeostasis, including the unfolded protein response (UPR) signaling, autophagy, and various cell death pathways. These mechanisms, however, often fail to prevent photoreceptor cell degradation and instead contribute to cell death under certain conditions. Current research focuses on the pharmacological modulation of the components of these pathways and the direct stabilization of mutated receptors as potential treatment strategies. Despite these efforts, the intricate interplay between these mechanisms and the diverse causative mutations involved has hindered the development of effective treatments. Advancing our understanding of the interactions between photoreceptor cell death mechanisms and the specific genetic mutations driving RP is critical to accelerate the discovery and development of therapeutic strategies for this currently incurable disease.
Collapse
Affiliation(s)
- Maria Azam
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
3
|
Dion W, Tao Y, Chambers M, Zhao S, Arbuckle RK, Sun M, Kubra S, Jamal I, Nie Y, Ye M, Larsen MB, Camarco D, Ickes E, DuPont C, Wang H, Wang B, Liu S, Pi S, Chen BB, Chen Y, Chen X, Zhu B. SON-dependent nuclear speckle rejuvenation alleviates proteinopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590103. [PMID: 38659924 PMCID: PMC11042303 DOI: 10.1101/2024.04.18.590103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Current treatments targeting individual protein quality control have limited efficacy in alleviating proteinopathies, highlighting the prerequisite for a common upstream druggable target capable of global proteostasis modulation. Building on our prior research establishing nuclear speckles as a pivotal membrane-less organelle responsible for global proteostasis transcriptional control, we aim to alleviate proteinopathies through nuclear speckle rejuvenation. We identified pyrvinium pamoate as a small-molecule nuclear speckle rejuvenator that enhances protein quality control while suppressing YAP1 signaling via decreasing the surface/interfacial tension of nuclear speckle condensates through interaction with the intrinsically disordered region of nuclear speckle scaffold protein SON. In pre-clinical models, nanomolar pyrvinium pamoate alleviated retina degeneration and reduced tauopathy by promoting autophagy and ubiquitin-proteasome system in a SON-dependent manner without causing cellular stress. Aberrant nuclear speckle morphology, reduced protein quality control and increased YAP1 activity were also observed in human tauopathies. Our study uncovers novel therapeutic targets for tackling protein misfolding disorders within an expanded proteostasis framework encompassing nuclear speckles and YAP1.
Collapse
Affiliation(s)
- William Dion
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Yuren Tao
- Department of Neuroscience, School of Medicine, University of California, San Diego, CA, U.S.A
| | - Maci Chambers
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Shanshan Zhao
- Department of Neuroscience, School of Medicine, University of California, San Diego, CA, U.S.A
| | - Riley K. Arbuckle
- Department of Ophthalmology, University of Pittsburgh School of Medicine, PA, U.S.A
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Michelle Sun
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Syeda Kubra
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Imran Jamal
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Yuhang Nie
- Department of Neuroscience, School of Medicine, University of California, San Diego, CA, U.S.A
| | - Megan Ye
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Mads B. Larsen
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Daniel Camarco
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Eleanor Ickes
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Claire DuPont
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Haokun Wang
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Bingjie Wang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, PA, U.S.A
| | - Silvia Liu
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Shaohua Pi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, PA, U.S.A
| | - Bill B Chen
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh School of Medicine, PA, U.S.A
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA, U.S.A
| | - Xu Chen
- Department of Neuroscience, School of Medicine, University of California, San Diego, CA, U.S.A
| | - Bokai Zhu
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, U.S.A
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, U.S.A
| |
Collapse
|
4
|
Brooks C, Kolson D, Sechrest E, Chuah J, Schupp J, Billington N, Deng WT, Smith D, Sokolov M. Therapeutic potential of archaeal unfoldase PANet and the gateless T20S proteasome in P23H rhodopsin retinitis pigmentosa mice. PLoS One 2024; 19:e0308058. [PMID: 39361629 PMCID: PMC11449290 DOI: 10.1371/journal.pone.0308058] [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: 07/16/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
Neurodegenerative diseases are characterized by the presence of misfolded and aggregated proteins which are thought to contribute to the development of the disease. In one form of inherited blinding disease, retinitis pigmentosa, a P23H mutation in the light-sensing receptor, rhodopsin causes rhodopsin misfolding resulting in complete vision loss. We investigated whether a xenogeneic protein-unfolding ATPase (unfoldase) from thermophilic Archaea, termed PANet, could counteract the proteotoxicity of P23H rhodopsin. We found that PANet increased the number of surviving photoreceptors in P23H rhodopsin mice and recognized rhodopsin as a substate in vitro. This data supports the feasibility and efficacy of using a xenogeneic unfoldase as a therapeutic approach in mouse models of human neurodegenerative diseases. We also showed that an archaeal proteasome, called the T20S can degrade rhodopsin in vitro and demonstrated that it is feasible and safe to express gateless T20S proteasomes in vivo in mouse rod photoreceptors. Expression of archaeal proteasomes may be an effective therapeutic approach to stimulate protein degradation in retinopathies and neurodegenerative diseases with protein-misfolding etiology.
Collapse
Affiliation(s)
- Celine Brooks
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia, United States of America
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Douglas Kolson
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia, United States of America
| | - Emily Sechrest
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia, United States of America
| | - Janelle Chuah
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Jane Schupp
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Neil Billington
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Wen-Tao Deng
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia, United States of America
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - David Smith
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, United States of America
| | - Maxim Sokolov
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia, United States of America
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|
5
|
Yang F, Ma H, Boye SL, Boye SE, Ding XQ. Promotion of endoplasmic reticulum retrotranslocation by overexpression of E3 ubiquitin-protein ligase synoviolin 1 reduces endoplasmic reticulum stress and preserves cone photoreceptors in cyclic nucleotide-gated channel deficiency. FASEB J 2024; 38:e70021. [PMID: 39215566 PMCID: PMC11419579 DOI: 10.1096/fj.202400198r] [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: 01/26/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Cone photoreceptor cyclic nucleotide-gated (CNG) channels play an essential role in phototransduction and cellular Ca2+ homeostasis. Mutations in genes encoding the channel subunits CNGA3 and CNGB3 are associated with achromatopsia, progressive cone dystrophy, and early-onset macular degeneration. Cone loss in patients with achromatopsia and cone dystrophy associated with CNG channel mutations has been documented by optical coherence tomography and in mouse models of CNG channel deficiency. Cone death in CNG channel-deficient retinas involves endoplasmic reticulum (ER) stress-associated apoptosis, dysregulation of cellular/ER Ca2+ homeostasis, impaired protein folding/processing, and impaired ER-associated degradation (ERAD). The E3 ubiquitin-protein ligase synoviolin 1 (SYVN1) is the primary component of the SYVN1/SEL1L ER retrotranslocon responsible for ERAD. Previous studies have shown that manipulations that protect cones and reduce ER stress/cone death in CNG channel deficiency, such as increasing ER Ca2+ preservation or treatment with an ER chaperone, increase the expression of SYVN1 and other components of the ER retrotranslocon. The present work investigated the effects of SYVN1 overexpression. Intraocular injection of AAV5-IRBP/GNAT2-Syvn1 resulted in overexpression of SYVN1 in cones of CNG channel-deficient mice. Following treatment, cone density in Cnga3-/- mice was significantly increased, compared with untreated controls, outer segment localization of cone opsin was improved, and ER stress/apoptotic cell death was reduced. Overexpression of SYVN1 also led to increased expression levels of the retrotranslocon components, degradation in ER protein 1 (DERL1), ERAD E3 ligase adaptor subunit (SEL1L), and homocysteine inducible ER protein with ubiquitin-like domain 1 (HERPUD1). Moreover, overexpression of SYVN1 likely enhanced protein ubiquitination/proteasome degradation in CNG channel-deficient retinas. This study demonstrates the role of SYVN1/ERAD in cone preservation in CNG channel deficiency and supports the strategy of promoting ERAD for cone protection.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Hongwei Ma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Sanford L. Boye
- Powell Gene Therapy Center, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Shannon E. Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Xi-Qin Ding
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
6
|
Wang Y, Becker S, Finkelstein S, Dyka FM, Liu H, Eminhizer M, Hao Y, Brush RS, Spencer WJ, Arshavsky VY, Ash JD, Du J, Agbaga MP, Vinberg F, Ellis JM, Lobanova ES. Acyl-CoA synthetase 6 controls rod photoreceptor function and survival by shaping the phospholipid composition of retinal membranes. Commun Biol 2024; 7:1027. [PMID: 39169121 PMCID: PMC11339274 DOI: 10.1038/s42003-024-06691-8] [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: 11/14/2023] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
The retina is light-sensitive neuronal tissue in the back of the eye. The phospholipid composition of the retina is unique and highly enriched in polyunsaturated fatty acids, including docosahexaenoic fatty acid (DHA). While it is generally accepted that a high DHA content is important for vision, surprisingly little is known about the mechanisms of DHA enrichment in the retina. Furthermore, the biological processes controlled by DHA in the eye remain poorly defined as well. Here, we combined genetic manipulations with lipidomic analysis in mice to demonstrate that acyl-CoA synthetase 6 (Acsl6) serves as a regulator of the unique composition of retinal membranes. Inactivation of Acsl6 reduced the levels of DHA-containing phospholipids, led to progressive loss of light-sensitive rod photoreceptor neurons, attenuated the light responses of these cells, and evoked distinct transcriptional response in the retina involving the Srebf1/2 (sterol regulatory element binding transcription factors 1/2) pathway. This study identifies one of the major enzymes responsible for DHA enrichment in the retinal membranes and introduces a model allowing an evaluation of rod functioning and pathology caused by impaired DHA incorporation/retention in the retina.
Collapse
Affiliation(s)
- Yixiao Wang
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Silke Becker
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, USA
| | | | - Frank M Dyka
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Eminhizer
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Ying Hao
- Department of Ophthalmology, Duke University, Durham, NC, USA
| | - Richard S Brush
- Department of Ophthalmology, University of Oklahoma Health Sciences Center and Dean McGee Eye Institute, Oklahoma City, OK, USA
| | - William J Spencer
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - John D Ash
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianhai Du
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Martin-Paul Agbaga
- Department of Ophthalmology, University of Oklahoma Health Sciences Center and Dean McGee Eye Institute, Oklahoma City, OK, USA
| | - Frans Vinberg
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, USA
| | | | | |
Collapse
|
7
|
Lewis TR, Castillo CM, Klementieva NV, Hsu Y, Hao Y, Spencer WJ, Drack AV, Pazour GJ, Arshavsky VY. Contribution of intraflagellar transport to compartmentalization and maintenance of the photoreceptor cell. Proc Natl Acad Sci U S A 2024; 121:e2408551121. [PMID: 39145934 PMCID: PMC11348033 DOI: 10.1073/pnas.2408551121] [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: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
The first steps of vision take place in the ciliary outer segment compartment of photoreceptor cells. The protein composition of outer segments is uniquely suited to perform this function. The most abundant among these proteins is the visual pigment, rhodopsin, whose outer segment trafficking involves intraflagellar transport (IFT). Here, we report three major findings from the analysis of mice in which ciliary transport was acutely impaired by conditional knockouts of IFT-B subunits. First, we demonstrate the existence of a sorting mechanism whereby mislocalized rhodopsin is recruited to and concentrated in extracellular vesicles prior to their release, presumably to protect the cell from adverse effects of protein mislocalization. Second, reducing rhodopsin expression significantly delays photoreceptor degeneration caused by IFT disruption, suggesting that controlling rhodopsin levels may be an effective therapy for some cases of retinal degenerative disease. Last, the loss of IFT-B subunits does not recapitulate a phenotype observed in mutants of the BBSome (another ciliary transport protein complex relying on IFT) in which non-ciliary proteins accumulate in the outer segment. Whereas it is widely thought that the role of the BBSome is to primarily participate in ciliary transport, our data suggest that the BBSome has another major function independent of IFT and possibly related to maintaining the diffusion barrier of the ciliary transition zone.
Collapse
Affiliation(s)
- Tylor R. Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | - Carson M. Castillo
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | | | - Ying Hsu
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA52242
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | - William J. Spencer
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | - Arlene V. Drack
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA52242
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA01605
| | - Vadim Y. Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
8
|
Vasudevan S, Senapati S, Pendergast M, Park PSH. Aggregation of rhodopsin mutants in mouse models of autosomal dominant retinitis pigmentosa. Nat Commun 2024; 15:1451. [PMID: 38365903 PMCID: PMC10873427 DOI: 10.1038/s41467-024-45748-4] [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: 04/07/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
Mutations in rhodopsin can cause it to misfold and lead to retinal degeneration. A distinguishing feature of these mutants in vitro is that they mislocalize and aggregate. It is unclear whether or not these features contribute to retinal degeneration observed in vivo. The effect of P23H and G188R misfolding mutations were examined in a heterologous expression system and knockin mouse models, including a mouse model generated here expressing the G188R rhodopsin mutant. In vitro characterizations demonstrate that both mutants aggregate, with the G188R mutant exhibiting a more severe aggregation profile compared to the P23H mutant. The potential for rhodopsin mutants to aggregate in vivo was assessed by PROTEOSTAT, a dye that labels aggregated proteins. Both mutants mislocalize in photoreceptor cells and PROTEOSTAT staining was detected surrounding the nuclei of photoreceptor cells. The G188R mutant promotes a more severe retinal degeneration phenotype and greater PROTEOSTAT staining compared to that promoted by the P23H mutant. Here, we show that the level of PROTEOSTAT positive cells mirrors the progression and level of photoreceptor cell death, which suggests a potential role for rhodopsin aggregation in retinal degeneration.
Collapse
Affiliation(s)
- Sreelakshmi Vasudevan
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Subhadip Senapati
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
- Prayoga Institute of Education Research, Bengaluru, KA, 560116, India
| | - Maryanne Pendergast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Paul S-H Park
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
9
|
Zegeye Y, Aredo B, Yuksel S, Kirman DC, Kumar A, Chen B, Turpin E, Shresta S, He YG, Gautron L, Tang M, Li X, DiCesare SM, Hulleman JD, Xing C, Ludwig S, Moresco EMY, Beutler BA, Ufret-Vincenty RL. E3 ubiquitin ligase Herc3 deficiency leads to accumulation of subretinal microglia and retinal neurodegeneration. Sci Rep 2024; 14:3010. [PMID: 38321224 PMCID: PMC10847449 DOI: 10.1038/s41598-024-53731-8] [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: 08/01/2023] [Accepted: 02/04/2024] [Indexed: 02/08/2024] Open
Abstract
Activated microglia have been implicated in the pathogenesis of age-related macular degeneration (AMD), diabetic retinopathy, and other neurodegenerative and neuroinflammatory disorders, but our understanding of the mechanisms behind their activation is in infant stages. With the goal of identifying novel genes associated with microglial activation in the retina, we applied a semiquantitative fundus spot scoring scale to an unbiased, state-of-the-science mouse forward genetics pipeline. A mutation in the gene encoding the E3 ubiquitin ligase Herc3 led to prominent accumulation of fundus spots. CRISPR mutagenesis was used to generate Herc3-/- mice, which developed prominent accumulation of fundus spots and corresponding activated Iba1 + /CD16 + subretinal microglia, retinal thinning on OCT and histology, and functional deficits by Optomotory and electrophysiology. Bulk RNA sequencing identified activation of inflammatory pathways and differentially expressed genes involved in the modulation of microglial activation. Thus, despite the known expression of multiple E3 ubiquitin ligases in the retina, we identified a non-redundant role for Herc3 in retinal homeostasis. Our findings are significant given that a dysregulated ubiquitin-proteasome system (UPS) is important in prevalent retinal diseases, in which activated microglia appear to play a role. This association between Herc3 deficiency, retinal microglial activation and retinal degeneration merits further study.
Collapse
Affiliation(s)
- Yeshumenesh Zegeye
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bogale Aredo
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Seher Yuksel
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Dogan Can Kirman
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ashwani Kumar
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bo Chen
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Emily Turpin
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sangita Shresta
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yu-Guang He
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Gautron
- Center for Hypothalamic Research and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Miao Tang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaohong Li
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sophia M DiCesare
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - John D Hulleman
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sara Ludwig
- Center for Hypothalamic Research and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Eva Marie Y Moresco
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruce A Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | |
Collapse
|
10
|
Lewis TR, Makia MS, Castillo CM, Hao Y, Al-Ubaidi MR, Skiba NP, Conley SM, Arshavsky VY, Naash MI. ROM1 is redundant to PRPH2 as a molecular building block of photoreceptor disc rims. eLife 2023; 12:RP89444. [PMID: 37991486 PMCID: PMC10665016 DOI: 10.7554/elife.89444] [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: 11/23/2023] Open
Abstract
Visual signal transduction takes place within a stack of flattened membranous 'discs' enclosed within the light-sensitive photoreceptor outer segment. The highly curved rims of these discs, formed in the process of disc enclosure, are fortified by large hetero-oligomeric complexes of two homologous tetraspanin proteins, PRPH2 (a.k.a. peripherin-2 or rds) and ROM1. While mutations in PRPH2 affect the formation of disc rims, the role of ROM1 remains poorly understood. In this study, we found that the knockout of ROM1 causes a compensatory increase in the disc content of PRPH2. Despite this increase, discs of ROM1 knockout mice displayed a delay in disc enclosure associated with a large diameter and lack of incisures in mature discs. Strikingly, further increasing the level of PRPH2 rescued these morphological defects. We next showed that disc rims are still formed in a knockin mouse in which the tetraspanin body of PRPH2 was replaced with that of ROM1. Together, these results demonstrate that, despite its contribution to the formation of disc rims, ROM1 can be replaced by an excess of PRPH2 for timely enclosure of newly forming discs and establishing normal outer segment structure.
Collapse
Affiliation(s)
- Tylor R Lewis
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Mustafa S Makia
- Department of Biomedical Engineering, University of HoustonHoustonUnited States
| | - Carson M Castillo
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of HoustonHoustonUnited States
- College of Optometry, University of HoustonHoustonUnited States
| | - Nikolai P Skiba
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical CenterDurhamUnited States
- Department of Pharmacology and Cancer Biology, Duke University Medical CenterDurhamUnited States
| | - Muna I Naash
- Department of Biomedical Engineering, University of HoustonHoustonUnited States
- College of Optometry, University of HoustonHoustonUnited States
| |
Collapse
|
11
|
Douglas VP, Douglas KAA, Iannaccone A. Microbiome and Inherited Retinal Degenerations. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1669-1674. [PMID: 37024045 DOI: 10.1016/j.ajpath.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/10/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023]
Abstract
Inherited retinal degenerations (IRDs) represent a genetically and clinically heterogeneous group of progressive and visually debilitating disorders that can lead to irreversible visual loss. Our understanding of IRD pathogenesis at both the genetic and cellular levels has increased tremendously over the past two decades, but the exact pathogenic mechanisms remain incompletely understood. Enhanced understanding of the pathophysiology of these diseases can result in new treatment targets. Alterations in the human gut microbiome play a key role in the pathogenesis of many ocular and nonocular diseases, such as age-related macular degeneration, neurologic and metabolic disorders, and autoimmune conditions. The gut microbiome regulates the susceptibility of mice to develop experimental autoimmune uveitis, a model for autoimmune disease of the posterior portion of the eye elicited by the systemic response to retinal antigens. Because of the mounting evidence in favor of a role for local and systemic inflammatory and autoimmune-mediated components to IRD pathogenesis, this review presents the current knowledge of gut microbiome in IRDs and discusses the association between possible changes in gut microbiome and pathogenesis of these diseases, with special attention to their possible contribution to the inflammatory underpinnings of IRDs.
Collapse
Affiliation(s)
- Vivian P Douglas
- Department of Ophthalmology, Athens Naval Hospital, Athens, Greece
| | - Konstantinos A A Douglas
- 1st Department of Ophthalmology, "G. Gennimatas" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Alessandro Iannaccone
- Department of Ophthalmology, Duke University School of Medicine, Duke Eye Center, Durham, North Carolina.
| |
Collapse
|
12
|
Kerschensteiner D. Losing, preserving, and restoring vision from neurodegeneration in the eye. Curr Biol 2023; 33:R1019-R1036. [PMID: 37816323 PMCID: PMC10575673 DOI: 10.1016/j.cub.2023.08.044] [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] [Indexed: 10/12/2023]
Abstract
The retina is a part of the brain that sits at the back of the eye, looking out onto the world. The first neurons of the retina are the rod and cone photoreceptors, which convert changes in photon flux into electrical signals that are the basis of vision. Rods and cones are frequent targets of heritable neurodegenerative diseases that cause visual impairment, including blindness, in millions of people worldwide. This review summarizes the diverse genetic causes of inherited retinal degenerations (IRDs) and their convergence onto common pathogenic mechanisms of vision loss. Currently, there are few effective treatments for IRDs, but recent advances in disparate areas of biology and technology (e.g., genome editing, viral engineering, 3D organoids, optogenetics, semiconductor arrays) discussed here enable promising efforts to preserve and restore vision in IRD patients with implications for neurodegeneration in less approachable brain areas.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
13
|
Lewis TR, Makia MS, Castillo CM, Hao Y, Al-Ubaidi MR, Skiba NP, Conley SM, Arshavsky VY, Naash MI. ROM1 is redundant to PRPH2 as a molecular building block of photoreceptor disc rims. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.02.547380. [PMID: 37693615 PMCID: PMC10491102 DOI: 10.1101/2023.07.02.547380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Visual signal transduction takes place within a stack of flattened membranous "discs" enclosed within the light-sensitive photoreceptor outer segment. The highly curved rims of these discs, formed in the process of disc enclosure, are fortified by large hetero-oligomeric complexes of two homologous tetraspanin proteins, PRPH2 (a.k.a. peripherin-2 or rds) and ROM1. While mutations in PRPH2 affect the formation of disc rims, the role of ROM1 remains poorly understood. In this study, we found that the knockout of ROM1 causes a compensatory increase in the disc content of PRPH2. Despite this increase, discs of ROM1 knockout mice displayed a delay in disc enclosure associated with a large diameter and lack of incisures in mature discs. Strikingly, further increasing the level of PRPH2 rescued these morphological defects. We next showed that disc rims are still formed in a knockin mouse in which the tetraspanin body of PRPH2 was replaced with that of ROM1. Together, these results demonstrate that, despite its contribution to the formation of disc rims, ROM1 can be replaced by an excess of PRPH2 for timely enclosure of newly forming discs and establishing normal outer segment structure.
Collapse
Affiliation(s)
- Tylor R. Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Mustafa S. Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA, 77204
| | - Carson M. Castillo
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA, 77204
- College of Optometry, University of Houston, Houston, TX, USA, 77204
| | - Nikolai P. Skiba
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA, 73104
| | - Vadim Y. Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA, 27710
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA, 27710
| | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA, 77204
- College of Optometry, University of Houston, Houston, TX, USA, 77204
| |
Collapse
|
14
|
Wang Y, Snell A, Dyka FM, Colvin ER, Ildefonso C, Ash JD, Lobanova ES. Overexpression of Nfe2l1 increases proteasome activity and delays vision loss in a preclinical model of human blindness. SCIENCE ADVANCES 2023; 9:eadd5479. [PMID: 37450596 PMCID: PMC10348684 DOI: 10.1126/sciadv.add5479] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Proteasomes are the central proteolytic machines that are critical for breaking down most of the damaged and abnormal proteins in human cells. Although universally applicable drugs are not yet available, the stimulation of proteasomal activity is being analyzed as a proof-of-principle strategy to increase cellular resistance to a broad range of proteotoxic stressors. These approaches have included the stimulation of proteasomes through the overexpression of individual proteasome subunits, phosphorylation, or conformational changes induced by small molecules or peptides. In contrast to these approaches, we evaluated a transcription-driven increase in the total proteasome pool to enhance the proteolytic capacity of degenerating retinal neurons. We show that overexpression of nuclear factor erythroid-2-like 1 (Nfe2l1) transcription factor stimulated proteasome biogenesis and activity, improved the clearance of the ubiquitin-proteasomal reporter, and delayed photoreceptor neuron loss in a preclinical mouse model of human blindness caused by misfolded proteins. The findings highlight Nfe2l1 as an emerging therapeutic target to treat neurodegenerative diseases linked to protein misfolding.
Collapse
Affiliation(s)
- Yixiao Wang
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Aaron Snell
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Frank M. Dyka
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Elizabeth R. Colvin
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Cristhian Ildefonso
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - John D. Ash
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ekaterina S. Lobanova
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
15
|
Zhao N, Li N, Wang T. PERK prevents rhodopsin degradation during retinitis pigmentosa by inhibiting IRE1-induced autophagy. J Cell Biol 2023; 222:e202208147. [PMID: 37022709 PMCID: PMC10082367 DOI: 10.1083/jcb.202208147] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/04/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023] Open
Abstract
Chronic endoplasmic reticulum (ER) stress is the underlying cause of many degenerative diseases, including autosomal dominant retinitis pigmentosa (adRP). In adRP, mutant rhodopsins accumulate and cause ER stress. This destabilizes wild-type rhodopsin and triggers photoreceptor cell degeneration. To reveal the mechanisms by which these mutant rhodopsins exert their dominant-negative effects, we established an in vivo fluorescence reporter system to monitor mutant and wild-type rhodopsin in Drosophila. By performing a genome-wide genetic screen, we found that PERK signaling plays a key role in maintaining rhodopsin homeostasis by attenuating IRE1 activities. Degradation of wild-type rhodopsin is mediated by selective autophagy of ER, which is induced by uncontrolled IRE1/XBP1 signaling and insufficient proteasome activities. Moreover, upregulation of PERK signaling prevents autophagy and suppresses retinal degeneration in the adRP model. These findings establish a pathological role for autophagy in this neurodegenerative condition and indicate that promoting PERK activity could be used to treat ER stress-related neuropathies, including adRP.
Collapse
Affiliation(s)
- Ning Zhao
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ning Li
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Tao Wang
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- College of Biological Sciences, China Agricultural University, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
- National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
16
|
Chew LA, Iannaccone A. Gene-agnostic approaches to treating inherited retinal degenerations. Front Cell Dev Biol 2023; 11:1177838. [PMID: 37123404 PMCID: PMC10133473 DOI: 10.3389/fcell.2023.1177838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Most patients with inherited retinal degenerations (IRDs) have been waiting for treatments that are "just around the corner" for decades, with only a handful of seminal breakthroughs happening in recent years. Highlighting the difficulties in the quest for curative therapeutics, Luxturna required 16 years of development before finally obtaining United States Food and Drug Administration (FDA) approval and its international equivalents. IRDs are both genetically and phenotypically heterogeneous. While this diversity offers many opportunities for gene-by-gene precision medicine-based approaches, it also poses a significant challenge. For this reason, alternative (or parallel) strategies to identify more comprehensive, across-the-board therapeutics for the genetically and phenotypically diverse IRD patient population are very appealing. Even when gene-specific approaches may be available and become approved for use, many patients may have reached a disease stage whereby these approaches may no longer be viable. Thus, alternate visual preservation or restoration therapeutic approaches are needed at these stages. In this review, we underscore several gene-agnostic approaches that are being developed as therapeutics for IRDs. From retinal supplementation to stem cell transplantation, optogenetic therapy and retinal prosthetics, these strategies would bypass at least in part the need for treating every individual gene or mutation or provide an invaluable complement to them. By considering the diverse patient population and treatment strategies suited for different stages and patterns of retinal degeneration, gene agnostic approaches are very well poised to impact favorably outcomes and prognosis for IRD patients.
Collapse
Affiliation(s)
- Lindsey A. Chew
- Duke Center for Retinal Degenerations and Ophthalmic Genetic Diseases, Department of Ophthalmology, Duke Eye Center, Duke University School of Medicine, Durham, NC, United States
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Alessandro Iannaccone
- Duke Center for Retinal Degenerations and Ophthalmic Genetic Diseases, Department of Ophthalmology, Duke Eye Center, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
17
|
Wang Y, Lobanova ES. Methods for In Vivo Characterization of Proteostasis in the Mouse Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:383-387. [PMID: 37440061 DOI: 10.1007/978-3-031-27681-1_56] [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] [Indexed: 07/14/2023]
Abstract
An increasing number of studies connect inherited and age-related retinal degenerations with changes in the regulation of proteostasis. Here, we describe technical aspects of existing assays allowing to assess the status of the ubiquitin-proteasome system (UPS), changes in autophagy, and protein translation in mouse retina in vivo. These methods are helpful for the development and testing approaches to modulate proteostasis and delay vision loss.
Collapse
Affiliation(s)
- Yixiao Wang
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Ekaterina S Lobanova
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA.
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
18
|
Barwick SR, Smith SB. Comparison of Mouse Models of Autosomal Dominant Retinitis Pigmentosa Due to the P23H Mutation of Rhodopsin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:341-345. [PMID: 37440054 DOI: 10.1007/978-3-031-27681-1_49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The need for robust and reliable animal models is a crucial step in studying any disease. This certainly applies to inherited retinal degenerative diseases, in which mutations of retinal specific genes result in photoreceptor cell death and subsequent visual loss. Animal models of retinal gene mutations have proven valuable to our understanding of disease mechanisms and as tools to evaluate therapeutic intervention strategies. Notable among these models are mice with a mutation of the rhodopsin gene at amino acid 23 in which proline is substituted for histidine (Rho-P23H). The RHO-P23H mutation is the most common cause of autosomal dominant retinitis pigmentosa. Here, we provide a brief review of the Rho-P23H mouse models currently available for research.
Collapse
Affiliation(s)
- Shannon R Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA.
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
19
|
Chen APF, Chea L, Lee EJ, Lin JH. Lysine Ubiquitylation Drives Rhodopsin Protein Turnover. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:493-498. [PMID: 37440077 DOI: 10.1007/978-3-031-27681-1_72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Rhodopsin is a G-protein-coupled receptor that is specifically and abundantly expressed in rod photoreceptors. Over 150 rhodopsin mutations cause autosomal dominant retinitis pigmentosa (adRP). The most common mutation in the United States is the conversion of proline to histidine at position 23 (P23H) in the N-terminal domain of rhodopsin. We previously found that P23H rhodopsin was misfolded, ubiquitinylated, and rapidly degraded. Here, we investigated the role of lysine residues on P23H rhodopsin ubiquitinylation and turnover. We transfected HEK293 cells with a P23H human rhodopsin construct where all 11 lysine residues were mutated to arginine (K-null P23H). We found that the K-null P23H rhodopsin was significantly less ubiquitylated than intact P23H rhodopsin. We found that K-null P23H protein turnover was significantly slower compared to P23H rhodopsin through cycloheximide chase analysis. Finally, we also generated a wild-type rhodopsin construct where all lysines were converted to arginine and found significantly reduced ubiquitylation. Our findings identify ubiquitinylation of lysine residues as an important posttranslational modification involved in P23H rhodopsin protein degradation.
Collapse
Affiliation(s)
- Allen P F Chen
- Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Leon Chea
- Department of Ophthalmology, Palo Alto, CA, USA
- Department of Pathology, Stanford University, Palo Alto, CA, USA
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Eun-Jin Lee
- Department of Ophthalmology, Palo Alto, CA, USA
- Department of Pathology, Stanford University, Palo Alto, CA, USA
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- USC ROSKI Eye Institute and Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jonathan H Lin
- Department of Ophthalmology, Palo Alto, CA, USA.
- Department of Pathology, Stanford University, Palo Alto, CA, USA.
- VA Palo Alto Healthcare System, Palo Alto, CA, USA.
| |
Collapse
|
20
|
Targeting immunoproteasome in neurodegeneration: A glance to the future. Pharmacol Ther 2023; 241:108329. [PMID: 36526014 DOI: 10.1016/j.pharmthera.2022.108329] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
The immunoproteasome is a specialized form of proteasome equipped with modified catalytic subunits that was initially discovered to play a pivotal role in MHC class I antigen processing and immune system modulation. However, over the last years, this proteolytic complex has been uncovered to serve additional functions unrelated to antigen presentation. Accordingly, it has been proposed that immunoproteasome synergizes with canonical proteasome in different cell types of the nervous system, regulating neurotransmission, metabolic pathways and adaptation of the cells to redox or inflammatory insults. Hence, studying the alterations of immunoproteasome expression and activity is gaining research interest to define the dynamics of neuroinflammation as well as the early and late molecular events that are likely involved in the pathogenesis of a variety of neurological disorders. Furthermore, these novel functions foster the perspective of immunoproteasome as a potential therapeutic target for neurodegeneration. In this review, we provide a brain and retina-wide overview, trying to correlate present knowledge on structure-function relationships of immunoproteasome with the variety of observed neuro-modulatory functions.
Collapse
|
21
|
Kim K, Safarta LA, Chiang WCJ, Coppinger JA, Lee EJ, Lin JH. Network biology analysis of P23H rhodopsin interactome identifies protein and mRNA quality control mechanisms. Sci Rep 2022; 12:17405. [PMID: 36258031 PMCID: PMC9579138 DOI: 10.1038/s41598-022-22316-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/12/2022] [Indexed: 02/05/2023] Open
Abstract
Rhodopsin is essential for phototransduction, and many rhodopsin mutations cause heritable retinal degenerations. The P23H rhodopsin variant generates a misfolded rhodopsin protein that photoreceptors quickly target for degradation by mechanisms that are incompletely understood. To gain insight into how P23H rhodopsin is removed from rods, we used mass spectrometry to identify protein interaction partners of P23H rhodopsin immunopurified from RhoP23H/P23H mice and compared them with protein interaction partners of wild-type rhodopsin from Rho+/+ mice. We identified 286 proteins associated with P23H rhodopsin and 276 proteins associated with wild-type rhodopsin. 113 proteins were shared between wild-type and mutant rhodopsin protein interactomes. In the P23H rhodopsin protein interactome, we saw loss of phototransduction, retinal cycle, and rhodopsin protein trafficking proteins but gain of ubiquitin-related proteins when compared with the wild-type rhodopsin protein interactome. In the P23H rhodopsin protein interactome, we saw enrichment of gene ontology terms related to ER-associated protein degradation, ER stress, and translation. Protein-protein interaction network analysis revealed that translational and ribosomal quality control proteins were significant regulators in the P23H rhodopsin protein interactome. The protein partners identified in our study may provide new insights into how photoreceptors recognize and clear mutant rhodopsin, offering possible novel targets involved in retinal degeneration pathogenesis.
Collapse
Affiliation(s)
- Kyle Kim
- Stanford University School of Medicine, 300 Pasteur Dr. L235, Stanford, CA, 94305, USA
- VA Palo Alto, Palo Alto, CA, 94304, USA
| | - Lance A Safarta
- Stanford University School of Medicine, 300 Pasteur Dr. L235, Stanford, CA, 94305, USA
- VA Palo Alto, Palo Alto, CA, 94304, USA
| | - Wei-Chieh J Chiang
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Judith A Coppinger
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Eun-Jin Lee
- Stanford University School of Medicine, 300 Pasteur Dr. L235, Stanford, CA, 94305, USA
- VA Palo Alto, Palo Alto, CA, 94304, USA
- University of Southern California, Los Angeles, CA, USA
| | - Jonathan H Lin
- Stanford University School of Medicine, 300 Pasteur Dr. L235, Stanford, CA, 94305, USA.
- VA Palo Alto, Palo Alto, CA, 94304, USA.
| |
Collapse
|
22
|
Kolesnikov AV, Lobysheva E, Gnana-Prakasam JP, Kefalov VJ, Kisselev OG. Regulation of rod photoreceptor function by farnesylated G-protein γ-subunits. PLoS One 2022; 17:e0272506. [PMID: 35939447 PMCID: PMC9359561 DOI: 10.1371/journal.pone.0272506] [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: 04/07/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022] Open
Abstract
Heterotrimeric G-protein transducin, Gt, is a key signal transducer and amplifier in retinal rod and cone photoreceptor cells. Despite similar subunit composition, close amino acid identity, and identical posttranslational farnesylation of their Gγ subunits, rods and cones rely on unique Gγ1 (Gngt1) and Gγc (Gngt2) isoforms, respectively. The only other farnesylated G-protein γ-subunit, Gγ11 (Gng11), is expressed in multiple tissues but not retina. To determine whether Gγ1 regulates uniquely rod phototransduction, we generated transgenic rods expressing Gγ1, Gγc, or Gγ11 in Gγ1-deficient mice and analyzed their properties. Immunohistochemistry and Western blotting demonstrated the robust expression of each transgenic Gγ in rod cells and restoration of Gαt1 expression, which is greatly reduced in Gγ1-deficient rods. Electroretinography showed restoration of visual function in all three transgenic Gγ1-deficient lines. Recordings from individual transgenic rods showed that photosensitivity impaired in Gγ1-deficient rods was also fully restored. In all dark-adapted transgenic lines, Gαt1 was targeted to the outer segments, reversing its diffuse localization found in Gγ1-deficient rods. Bright illumination triggered Gαt1 translocation from the rod outer to inner segments in all three transgenic strains. However, Gαt1 translocation in Gγ11 transgenic mice occurred at significantly dimmer background light. Consistent with this, transretinal ERG recordings revealed gradual response recovery in moderate background illumination in Gγ11 transgenic mice but not in Gγ1 controls. Thus, while farnesylated Gγ subunits are functionally active and largely interchangeable in supporting rod phototransduction, replacement of retina-specific Gγ isoforms by the ubiquitous Gγ11 affects the ability of rods to adapt to background light.
Collapse
Affiliation(s)
- Alexander V. Kolesnikov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA, United States of America
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Elena Lobysheva
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jaya P. Gnana-Prakasam
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Vladimir J. Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA, United States of America
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Physiology and Biophysics, University of California, Irvine, CA, United States of America
| | - Oleg G. Kisselev
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
23
|
Robichaux MA, Nguyen V, Chan F, Kailasam L, He F, Wilson JH, Wensel TG. Subcellular localization of mutant P23H rhodopsin in an RFP fusion knock-in mouse model of retinitis pigmentosa. Dis Model Mech 2022; 15:274688. [PMID: 35275162 PMCID: PMC9092655 DOI: 10.1242/dmm.049336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/28/2022] [Indexed: 12/15/2022] Open
Abstract
The P23H mutation in rhodopsin (Rho), the rod visual pigment, is the most common allele associated with autosomal-dominant retinitis pigmentosa (adRP). The fate of misfolded mutant Rho in rod photoreceptors has yet to be elucidated. We generated a new mouse model, in which the P23H-Rho mutant allele is fused to the fluorescent protein Tag-RFP-T (P23HhRhoRFP). In heterozygotes, outer segments formed, and wild-type (WT) rhodopsin was properly localized, but mutant P23H-Rho protein was mislocalized in the inner segments. Heterozygotes exhibited slowly progressing retinal degeneration. Mislocalized P23HhRhoRFP was contained in greatly expanded endoplasmic reticulum (ER) membranes. Quantification of mRNA for markers of ER stress and the unfolded protein response revealed little or no increases. mRNA levels for both the mutant human rhodopsin allele and the WT mouse rhodopsin were reduced, but protein levels revealed selective degradation of the mutant protein. These results suggest that the mutant rods undergo an adaptative process that prolongs survival despite unfolded protein accumulation in the ER. The P23H-Rho-RFP mouse may represent a useful tool for the future study of the pathology and treatment of P23H-Rho and adRP. This article has an associated First Person interview with the first author of the paper. Summary: A mouse line with a knock-in of the human rhodopsin gene altered to contain the P23H mutation and a red fluorescent protein fusion provides a new model for autosomal-dominant retinitis pigmentosa.
Collapse
Affiliation(s)
- Michael A Robichaux
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.,Departments of Ophthalmology and Biochemistry, West Virginia University, 108 Biomedical Road, Morgantown, WV 26506, USA
| | - Vy Nguyen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Fung Chan
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Lavanya Kailasam
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Feng He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - John H Wilson
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
24
|
Peng H, Ramadurgum P, Woodard DR, Daniel S, Nakahara E, Renwick M, Aredo B, Datta S, Chen B, Ufret-Vincenty R, Hulleman JD. Utility of the DHFR-based destabilizing domain across mouse models of retinal degeneration and aging. iScience 2022; 25:104206. [PMID: 35521529 PMCID: PMC9062244 DOI: 10.1016/j.isci.2022.104206] [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: 04/05/2021] [Revised: 02/16/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022] Open
Abstract
The Escherichia coli dihydrofolate reductase (DHFR) destabilizing domain (DD) serves as a promising approach to conditionally regulate protein abundance in a variety of tissues. To test whether this approach could be effectively applied to a wide variety of aged and disease-related ocular mouse models, we evaluated the DHFR DD system in the eyes of aged mice (up to 24 months), a light-induced retinal degeneration (LIRD) model, and two genetic models of retinal degeneration (rd2 and Abca4−/− mice). The DHFR DD was effectively degraded in all model systems, including rd2 mice, which showed significant defects in chymotrypsin proteasomal activity. Moreover, trimethoprim (TMP) administration stabilized the DHFR DD in all mouse models. Thus, the DHFR DD-based approach allows for control of protein abundance in a variety of mouse models, laying the foundation to use this strategy for the conditional control of gene therapies to potentially treat multiple eye diseases. Destabilizing domains (DDs) confer conditional control of ocular protein abundance The DHFR DD is effectively turned over and stabilized in aged mouse’s retina DHFR DDs perform well in environmental and genetic retinal degenerative models
Collapse
|
25
|
Vats A, Xi Y, Feng B, Clinger OD, St Leger AJ, Liu X, Ghosh A, Dermond CD, Lathrop KL, Tochtrop GP, Picaud S, Chen Y. Non-retinoid chaperones improve rhodopsin homeostasis in a mouse model of retinitis pigmentosa. JCI Insight 2022; 7:153717. [PMID: 35472194 PMCID: PMC9220944 DOI: 10.1172/jci.insight.153717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Rhodopsin-associated (RHO-associated) retinitis pigmentosa (RP) is a progressive retinal disease that currently has no cure. RHO protein misfolding leads to disturbed proteostasis and the death of rod photoreceptors, resulting in decreased vision. We previously identified nonretinoid chaperones of RHO, including YC-001 and F5257-0462, by small-molecule high-throughput screening. Here, we profile the chaperone activities of these molecules toward the cell-surface level of 27 RP-causing human RHO mutants in NIH3T3 cells. Furthermore, using retinal explant culture, we show that YC-001 improves retinal proteostasis by supporting RHO homeostasis in RhoP23H/+ mouse retinae, which results in thicker outer nuclear layers (ONL), indicating delayed photoreceptor degeneration. Interestingly, YC-001 ameliorated retinal immune responses and reduced the number of microglia/macrophages in the RhoP23H/+ retinal explants. Similarly, F5257-0462 also protects photoreceptors in RhoP23H/+ retinal explants. In vivo, intravitreal injection of YC-001 or F5257-0462 microparticles in PBS shows that F5257-0462 has a higher efficacy in preserving photoreceptor function and delaying photoreceptor death in RhoP23H/+ mice. Collectively, we provide proof of principle that nonretinoid chaperones are promising drug candidates in treating RHO-associated RP.
Collapse
Affiliation(s)
- Abhishek Vats
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Yibo Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Bing Feng
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Owen D Clinger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Anthony J St Leger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Xujie Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Archisha Ghosh
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Chase D Dermond
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Kira L Lathrop
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, United States of America
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| |
Collapse
|
26
|
Wang Y, Punzo C, Ash JD, Lobanova ES. Tsc2 knockout counteracts ubiquitin-proteasome system insufficiency and delays photoreceptor loss in retinitis pigmentosa. Proc Natl Acad Sci U S A 2022; 119:e2118479119. [PMID: 35275792 PMCID: PMC8931319 DOI: 10.1073/pnas.2118479119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/12/2022] [Indexed: 01/18/2023] Open
Abstract
SignificanceStudies in multiple experimental systems have demonstrated that an increase in proteolytic capacity of post-mitotic cells improves cellular resistance to a variety of stressors, delays cellular aging and senescence. Therefore, approaches to increase the ability of cells to degrade misfolded proteins could potentially be applied to the treatment of a broad spectrum of human disorders. An example would be retinal degenerations, which cause irreversible loss of vision and are linked to impaired protein degradation. This study suggests that chronic activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway in degenerating photoreceptor neurons could stimulate the degradation of ubiquitinated proteins and enhance proteasomal activity through phosphorylation.
Collapse
Affiliation(s)
- Yixiao Wang
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610
| | - Claudio Punzo
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Medical School, Worcester, MA 01655
| | - John D. Ash
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610
| | - Ekaterina S. Lobanova
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610
| |
Collapse
|
27
|
Xi Y, Chen Y. Pharmacological strategies for treating misfolded rhodopsin-associated autosomal dominant retinitis pigmentosa. Neural Regen Res 2022; 17:110-112. [PMID: 34100444 PMCID: PMC8451548 DOI: 10.4103/1673-5374.314306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Yibo Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuanyuan Chen
- Department of Pharmacology and Chemical Biology; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
28
|
Adamus G. Importance of Autoimmune Responses in Progression of Retinal Degeneration Initiated by Gene Mutations. Front Med (Lausanne) 2021; 8:672444. [PMID: 34926479 PMCID: PMC8674421 DOI: 10.3389/fmed.2021.672444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Inherited retinal diseases (IRDs) are clinically and genetically heterogeneous rare disorders associated with retinal dysfunction and death of retinal photoreceptor cells, leading to blindness. Among the most frequent and severe forms of those retinopathies is retinitis pigmentosa (RP) that affects 1:4,000 individuals worldwide. The genes that have been implicated in RP are associated with the proteins present in photoreceptor cells or retinal pigment epithelium (RPE). Asymmetric presentation or sudden progression in retinal disease suggests that a gene mutation alone might not be responsible for retinal degeneration. Immune responses could directly target the retina or be site effect of immunity as a bystander deterioration. Autoantibodies against retinal autoantigens have been found in RP, which led to a hypothesis that autoimmunity could be responsible for the progression of photoreceptor cell death initiated by a genetic mutation. The other contributory factor to retinal degeneration is inflammation that activates the innate immune mechanisms, such as complement. If autoimmune responses contribute to the progression of retinopathy, this could have an implication on treatment, such as gene replacement therapy. In this review, we provide a perspective on the current role of autoimmunity/immunity in RP pathophysiology.
Collapse
Affiliation(s)
- Grazyna Adamus
- Ocular Immunology Laboratory, Casey Eye Institute, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
29
|
Frayssinhes JYA, Cerruti F, Laulin J, Cattaneo A, Bachi A, Apcher S, Coux O, Cascio P. PA28γ-20S proteasome is a proteolytic complex committed to degrade unfolded proteins. Cell Mol Life Sci 2021; 79:45. [PMID: 34913092 PMCID: PMC11071804 DOI: 10.1007/s00018-021-04045-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023]
Abstract
PA28γ is a nuclear activator of the 20S proteasome that, unlike the 19S regulatory particle, stimulates hydrolysis of several substrates in an ATP- and ubiquitin-independent manner and whose exact biological functions and molecular mechanism of action still remain elusive. In an effort to shed light on these important issues, we investigated the stimulatory effect of PA28γ on the hydrolysis of different fluorogenic peptides and folded or denatured full-length proteins by the 20S proteasome. Importantly, PA28γ was found to dramatically enhance breakdown rates by 20S proteasomes of several naturally or artificially unstructured proteins, but not of their native, folded counterparts. Furthermore, these data were corroborated by experiments in cell lines with a nucleus-tagged myelin basic protein. Finally, mass spectrometry analysis of the products generated during proteasomal degradation of two proteins demonstrated that PA28γ does not increase, but rather decreases, the variability of peptides that are potentially suitable for MHC class I antigen presentation. These unexpected findings indicate that global stimulation of the degradation of unfolded proteins may represent a more general feature of PA28γ and suggests that this proteasomal activator might play a broader role in the pathway of protein degradation than previously believed.
Collapse
Affiliation(s)
| | - Fulvia Cerruti
- Department of Veterinary Sciences, University of Turin, Largo P. Braccini 2, 10095, Grugliasco, Turin, Italy
| | - Justine Laulin
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie Des Tumeurs et Immunothérapie, Villejuif, France
| | | | - Angela Bachi
- The FIRC Institute of Molecular Oncology (IFOM), 20139, Milan, Italy
| | - Sebastien Apcher
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie Des Tumeurs et Immunothérapie, Villejuif, France
| | - Olivier Coux
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), CNRS UMR 5237, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Paolo Cascio
- Department of Veterinary Sciences, University of Turin, Largo P. Braccini 2, 10095, Grugliasco, Turin, Italy.
| |
Collapse
|
30
|
Retinal ganglion cell loss in an ex vivo mouse model of optic nerve cut is prevented by curcumin treatment. Cell Death Discov 2021; 7:394. [PMID: 34911931 PMCID: PMC8674341 DOI: 10.1038/s41420-021-00760-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/27/2021] [Accepted: 11/09/2021] [Indexed: 11/08/2022] Open
Abstract
Retinal ganglion cell (RGC) loss is a pathologic feature common to several retinopathies associated to optic nerve damage, leading to visual loss and blindness. Although several scientific efforts have been spent to understand the molecular and cellular changes occurring in retinal degeneration, an effective therapy to counteract the retinal damage is still not available. Here we show that eyeballs, enucleated with the concomitant optic nerve cut (ONC), when kept in PBS for 24 h showed retinal and optic nerve degeneration. Examining retinas and optic nerves at different time points in a temporal window of 24 h, we found a thinning of some retinal layers especially RGC's layer, observing a powerful RGC loss after 24 h correlated with an apoptotic, MAPKs and degradative pathways dysfunctions. Specifically, we detected a time-dependent increase of Caspase-3, -9 and pro-apoptotic marker levels, associated with a strong reduction of BRN3A and NeuN levels. Importantly, a powerful activation of JNK, c-Jun, and ERK signaling (MAPKs) were observed, correlated with a significant augmented SUMO-1 and UBC9 protein levels. The degradation signaling pathways was also altered, causing a significant decrease of ubiquitination level and an increased LC3B activation. Notably, it was also detected an augmented Tau protein level. Curcumin, a powerful antioxidant natural compound, prevented the alterations of apoptotic cascade, MAPKs, and SUMO-1 pathways and the degradation system, preserving the RGC survival and the retinal layer thickness. This ex vivo retinal degeneration model could be a useful method to study, in a short time window, the effect of neuroprotective tools like curcumin that could represent a potential treatment to contrast retinal cell death.
Collapse
|
31
|
Ye Q, Wang J, Liu X, Liu Z, BaZong L, Ma J, Shen R, Ye W, Zhang W, Wang D. The Role of RAD6B and PEDF in Retinal Degeneration. Neuroscience 2021; 480:19-31. [PMID: 34774969 DOI: 10.1016/j.neuroscience.2021.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022]
Abstract
RAD6B is an E2 ubiquitin-conjugating enzyme, playing an important role in DNA damage repair, gene expression, senescence, apoptosis and protein degradation. However, the specific mechanism between ubiquitin and retinal degeneration requires more investigation. Pigment epithelium-derived factor (PEDF) has a potent neurotrophic effect on the retina, protecting retinal neurons and photoreceptors from cell death caused by pathological damage. In this study, we found that loss of RAD6B leads to retinal degeneration in mice, especially in old age. Affymetrix microarray analysis showed that the PEDF signal was changed in RAD6B deficient groups. The expression of γ-H2AX, β-Gal, P53, Caspase-3, P21 and P16 was increased significantly in retinas of RAD6B knockout (KO) mice. Our studies suggest that RAD6B and PEDF play an important role in the health of retina, whereas the absence of RAD6B accelerates the degeneration.
Collapse
Affiliation(s)
- Qiang Ye
- Institute of Human Anatomy and Histoembryology, Basic Medical College, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China; Department of Ophthalmology, The Second Hospital of Lanzhou University, 82 Cuiying Door, Lanzhou 730000, China
| | - Jiaqi Wang
- Institute of Human Anatomy and Histoembryology, Basic Medical College, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China
| | - Xiangwen Liu
- Institute of Human Anatomy and Histoembryology, Basic Medical College, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China
| | - Zihua Liu
- Institute of Human Anatomy and Histoembryology, Basic Medical College, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China
| | - LuoSong BaZong
- Department of Ophthalmology, The Second Hospital of Lanzhou University, 82 Cuiying Door, Lanzhou 730000, China
| | - Jinhai Ma
- Department of Ophthalmology, The Second Hospital of Lanzhou University, 82 Cuiying Door, Lanzhou 730000, China
| | - Rong Shen
- Institute of Human Anatomy and Histoembryology, Basic Medical College, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China.
| | - Weichun Ye
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou 730000, China.
| | - Wenfang Zhang
- Department of Ophthalmology, The Second Hospital of Lanzhou University, 82 Cuiying Door, Lanzhou 730000, China.
| | - Degui Wang
- Institute of Human Anatomy and Histoembryology, Basic Medical College, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, China.
| |
Collapse
|
32
|
Marchesi N, Fahmideh F, Boschi F, Pascale A, Barbieri A. Ocular Neurodegenerative Diseases: Interconnection between Retina and Cortical Areas. Cells 2021; 10:2394. [PMID: 34572041 PMCID: PMC8469605 DOI: 10.3390/cells10092394] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
The possible interconnection between the eye and central nervous system (CNS) has been a topic of discussion for several years just based on fact that the eye is properly considered an extension of the brain. Both organs consist of neurons and derived from a neural tube. The visual process involves photoreceptors that receive light stimulus from the external environment and send it to retinal ganglionic cells (RGC), one of the cell types of which the retina is composed. The retina, the internal visual membrane of the eye, processes the visual stimuli in electric stimuli to transfer it to the brain, through the optic nerve. Retinal chronic progressive neurodegeneration, which may occur among the elderly, can lead to different disorders of the eye such as glaucoma, age-related macular degeneration (AMD), and diabetic retinopathy (DR). Mainly in the elderly population, but also among younger people, such ocular pathologies are the cause of irreversible blindness or impaired, reduced vision. Typical neurodegenerative diseases of the CSN are a group of pathologies with common characteristics and etiology not fully understood; some risk factors have been identified, but they are not enough to justify all the cases observed. Furthermore, several studies have shown that also ocular disorders present characteristics of neurodegenerative diseases and, on the other hand, CNS pathologies, i.e., Alzheimer disease (AD) and Parkinson disease (PD), which are causes of morbidity and mortality worldwide, show peculiar alterations at the ocular level. The knowledge of possible correlations could help to understand the mechanisms of onset. Moreover, the underlying mechanisms of these heterogeneous disorders are still debated. This review discusses the characteristics of the ocular illnesses, focusing on the relationship between the eye and the brain. A better comprehension could help in future new therapies, thus reducing or avoiding loss of vision and improve quality of life.
Collapse
Affiliation(s)
| | | | | | | | - Annalisa Barbieri
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Viale Taramelli 14, 27100 Pavia, Italy; (N.M.); (F.F.); (F.B.); (A.P.)
| |
Collapse
|
33
|
Prokai-Tatrai K, Zaman K, Nguyen V, De La Cruz DL, Prokai L. Proteomics-Based Retinal Target Engagement Analysis and Retina-Targeted Delivery of 17β-Estradiol by the DHED Prodrug for Ocular Neurotherapy in Males. Pharmaceutics 2021; 13:1392. [PMID: 34575465 PMCID: PMC8466286 DOI: 10.3390/pharmaceutics13091392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/21/2022] Open
Abstract
We examined the impact of 17β-estradiol (E2) eye drops on the modulation of the proteome profile in the male rat retina. With discovery-driven proteomics, we have identified proteins that were regulated by our treatment. These proteins were assembled to several bioinformatics-based networks implicating E2's beneficial effects on the male rat retina in a broad context of ocular neuroprotection including the maintenance of retinal homeostasis, facilitation of efficient disposal of damaged proteins, and mitochondrial respiratory chain biogenesis. We have also shown for the first time that the hormone's beneficial effects on the male retina can be constrained to this target site by treatment with the bioprecursor prodrug, DHED. A large concentration of E2 was produced after DHED eye drops not only in male rat retinae but also in those of rabbits. However, DHED treatment did not increase circulating E2 levels, thereby ensuring therapeutic safety in males. Targeted proteomics focusing on selected biomarkers of E2's target engagement further confirmed the prodrug's metabolism to E2 in the male retina and indicated that the retinal impact of DHED treatment was identical to that of the direct E2 treatment. Altogether, our study shows the potential of topical DHED therapy for an efficacious and safe protection of the male retina without the unwanted hormonal side-effects associated with current estrogen therapies.
Collapse
Affiliation(s)
- Katalin Prokai-Tatrai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (K.Z.); (V.N.); (D.L.D.L.C.); (L.P.)
| | | | | | | | | |
Collapse
|
34
|
Technological advancements to study cellular signaling pathways in inherited retinal degenerative diseases. Curr Opin Pharmacol 2021; 60:102-110. [PMID: 34388439 DOI: 10.1016/j.coph.2021.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/02/2021] [Indexed: 01/01/2023]
Abstract
Inherited retinal degenerative diseases (IRDs) are rare neurodegenerative disorders with mutations in hundreds of genes leading to vision loss, primarily owing to photoreceptor cell death. This genetic diversity is impeding development of effective treatment options. Gene-based therapies have resulted in the first FDA-approved drug (Luxturna) for RPE65-specific IRD. Although currently explored in clinical trials, genomic medicines are mutation-dependent, hence suitable only for patients harboring a specific mutation. Better understanding of the pathways leading to photoreceptor degeneration may help to determine common targets and develop mutation-independent therapies for larger groups of patients with IRDs. In this review, we discuss the key pathways involved in photoreceptor cell death studied by transcriptomics, proteomics, and metabolomics techniques to identify potential therapeutic targets in IRDs.
Collapse
|
35
|
Yang F, Ma H, Butler MR, Ding XQ. Preservation of endoplasmic reticulum (ER) Ca 2+ stores by deletion of inositol-1,4,5-trisphosphate receptor type 1 promotes ER retrotranslocation, proteostasis, and protein outer segment localization in cyclic nucleotide-gated channel-deficient cone photoreceptors. FASEB J 2021; 35:e21579. [PMID: 33960001 DOI: 10.1096/fj.202002711r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/03/2021] [Accepted: 03/25/2021] [Indexed: 11/11/2022]
Abstract
Endoplasmic reticulum (ER) Ca2+ homeostasis relies on an appropriate balance between efflux- and influx-channel activity responding to dynamic changes of intracellular Ca2+ levels. Dysregulation of this complex signaling network has been shown to contribute to neuronal and photoreceptor death in neuro- and retinal degenerative diseases, respectively. In mice with cone cyclic nucleotide-gated (CNG) channel deficiency, a model of achromatopsia/cone dystrophy, cones display early-onset ER stress-associated apoptosis and protein mislocalization. Cones in these mice also show reduced cytosolic Ca2+ level and subsequent elevation in the ER Ca2+ -efflux-channel activity, specifically the inositol-1,4,5-trisphosphate receptor type 1 (IP3 R1), and deletion of IP3 R1 results in preservation of cones. This work investigated how preservation of ER Ca2+ stores leads to cone protection. We examined the effects of cone specific deletion of IP3 R1 on ER stress responses/cone death, protein localization, and ER proteostasis/ER-associated degradation. We demonstrated that deletion of IP3 R1 improves trafficking of cone-specific proteins M-/S-opsin and phosphodiesterase 6C to cone outer segments and reduces localization to cone inner segments. Consistent with the improved protein localization, deletion of IP3 R1 results in increased ER retrotranslocation protein expression, reduced proteasome subunit expression, reduced ER stress/cone death, and reduced retinal remodeling. We also observed the enhanced ER retrotranslocation in mice that have been treated with a chemical chaperone, supporting the connection between improved ER retrotranslocation/proteostasis and alleviation of ER stress. Findings from this work demonstrate the importance of ER Ca2+ stores in ER proteostasis and protein trafficking/localization in photoreceptors, strengthen the link between dysregulation of ER Ca2+ homeostasis and ER stress/cone degeneration, and support an involvement of improved ER proteostasis in ER Ca2+ preservation-induced cone protection; thereby identifying IP3 R1 as a critical mediator of ER stress and protein mislocalization and as a potential target to preserve cones in CNG channel deficiency.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hongwei Ma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael R Butler
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xi-Qin Ding
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
36
|
Gunawan M, Low C, Neo K, Yeo S, Ho C, Barathi VA, Chan AS, Sharif NA, Kageyama M. The Role of Autophagy in Chemical Proteasome Inhibition Model of Retinal Degeneration. Int J Mol Sci 2021; 22:ijms22147271. [PMID: 34298888 PMCID: PMC8303873 DOI: 10.3390/ijms22147271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 01/27/2023] Open
Abstract
We recently demonstrated that chemical proteasome inhibition induced inner retinal degeneration, supporting the pivotal roles of the ubiquitin–proteasome system in retinal structural integrity maintenance. In this study, using beclin1-heterozygous (Becn1-Het) mice with autophagic dysfunction, we tested our hypothesis that autophagy could be a compensatory retinal protective mechanism for proteasomal impairment. Despite the reduced number of autophagosome, the ocular tissue morphology and intraocular pressure were normal. Surprisingly, Becn1-Het mice experienced the same extent of retinal degeneration as was observed in wild-type mice, following an intravitreal injection of a chemical proteasome inhibitor. Similarly, these mice equally responded to other chemical insults, including endoplasmic reticulum stress inducer, N-methyl-D-aspartate, and lipopolysaccharide. Interestingly, in cultured neuroblastoma cells, we found that the mammalian target of rapamycin-independent autophagy activators, lithium chloride and rilmenidine, rescued these cells against proteasome inhibition-induced death. These results suggest that Becn1-mediated autophagy is not an effective intrinsic protective mechanism for retinal damage induced by insults, including impaired proteasomal activity; furthermore, autophagic activation beyond normal levels is required to alleviate the cytotoxic effect of proteasomal inhibition. Further studies are underway to delineate the precise roles of different forms of autophagy, and investigate the effects of their activation in rescuing retinal neurons under various pathological conditions.
Collapse
Affiliation(s)
- Merry Gunawan
- Santen-SERI Open Innovation Centre, 20 College Road, The Academia, Singapore 169856, Singapore; (M.G.); (C.L.); (K.N.)
| | - Choonbing Low
- Santen-SERI Open Innovation Centre, 20 College Road, The Academia, Singapore 169856, Singapore; (M.G.); (C.L.); (K.N.)
| | - Kurt Neo
- Santen-SERI Open Innovation Centre, 20 College Road, The Academia, Singapore 169856, Singapore; (M.G.); (C.L.); (K.N.)
| | - Siawey Yeo
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, 20 College Road, The Academia, Singapore 169856, Singapore; (S.Y.); (V.A.B.)
| | - Candice Ho
- Singapore Eye Research Institute, 20 College Road, The Academia, Singapore 169856, Singapore; (C.H.); (A.S.C.)
| | - Veluchamy A. Barathi
- Translational Pre-Clinical Model Platform, Singapore Eye Research Institute, 20 College Road, The Academia, Singapore 169856, Singapore; (S.Y.); (V.A.B.)
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
- Academic Clinical Program in Ophthalmology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Anita Sookyee Chan
- Singapore Eye Research Institute, 20 College Road, The Academia, Singapore 169856, Singapore; (C.H.); (A.S.C.)
| | - Najam A. Sharif
- Global Alliance and External Research, Santen Inc., Emeryville, CA 94608, USA;
| | - Masaaki Kageyama
- Santen-SERI Open Innovation Centre, 20 College Road, The Academia, Singapore 169856, Singapore; (M.G.); (C.L.); (K.N.)
- Correspondence:
| |
Collapse
|
37
|
Saltykova IV, Elahi A, Pitale PM, Gorbatyuk OS, Athar M, Gorbatyuk MS. Tribbles homolog 3-mediated targeting the AKT/mTOR axis in mice with retinal degeneration. Cell Death Dis 2021; 12:664. [PMID: 34215725 PMCID: PMC8253859 DOI: 10.1038/s41419-021-03944-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/28/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023]
Abstract
Various retinal degenerative disorders manifest in alterations of the AKT/mTOR axis. Despite this, consensus on the therapeutic targeting of mTOR in degenerating retinas has not yet been achieved. Therefore, we investigated the role of AKT/mTOR signaling in rd16 retinas, in which we restored the AKT/mTOR axis by genetic ablation of pseudokinase TRB3, known to inhibit phosphorylation of AKT and mTOR. First, we found that TRB3 ablation resulted in preservation of photoreceptor function in degenerating retinas. Then, we learned that the mTOR downstream cellular pathways involved in the homeostasis of photoreceptors were also reprogrammed in rd16 TRB3-/- retinas. Thus, the level of inactivated translational repressor p-4E-BP1 was significantly increased in these mice along with the restoration of translational rate. Moreover, in rd16 mice manifesting decline in p-mTOR at P15, we found elevated expression of Beclin-1 and ATG5 autophagy genes. Thus, these mice showed impaired autophagy flux measured as an increase in LC3 conversion and p62 accumulation. In addition, the RFP-EGFP-LC3 transgene expression in rd16 retinas resulted in statistically fewer numbers of red puncta in photoreceptors, suggesting impaired late autophagic vacuoles. In contrast, TRIB3 ablation in these mice resulted in improved autophagy flux. The restoration of translation rate and the boost in autophagosome formation occurred concomitantly with an increase in total Ub and rhodopsin protein levels and the elevation of E3 ligase Parkin1. We propose that TRB3 may retard retinal degeneration and be a promising therapeutic target to treat various retinal degenerative disorders.
Collapse
Affiliation(s)
- Irina V Saltykova
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Asif Elahi
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Priyam M Pitale
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Oleg S Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohammad Athar
- Department of Dermatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marina S Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
38
|
Inhibition of Epigenetic Modifiers LSD1 and HDAC1 Blocks Rod Photoreceptor Death in Mouse Models of Retinitis Pigmentosa. J Neurosci 2021; 41:6775-6792. [PMID: 34193554 DOI: 10.1523/jneurosci.3102-20.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 11/21/2022] Open
Abstract
Epigenetic modifiers are increasingly being investigated as potential therapeutics to modify and overcome disease phenotypes. Diseases of the nervous system present a particular problem as neurons are postmitotic and demonstrate relatively stable gene expression patterns and chromatin organization. We have explored the ability of epigenetic modifiers to prevent degeneration of rod photoreceptors in a mouse model of retinitis pigmentosa (RP), using rd10 mice of both sexes. The histone modification eraser enzymes lysine demethylase 1 (LSD1) and histone deacetylase 1 (HDAC1) are known to have dramatic effects on the development of rod photoreceptors. In the RP mouse model, inhibitors of these enzymes blocked rod degeneration, preserved vision, and affected the expression of multiple genes including maintenance of rod-specific transcripts and downregulation of those involved in inflammation, gliosis, and cell death. The neuroprotective activity of LSD1 inhibitors includes two pathways. First, through targeting histone modifications, they increase accessibility of chromatin and upregulate neuroprotective genes, such as from the Wnt pathway. We propose that this process is going in rod photoreceptors. Second, through nonhistone targets, they inhibit transcription of inflammatory genes and inflammation. This process is going in microglia, and lack of inflammation keeps rod photoreceptors alive.SIGNIFICANCE STATEMENT Retinal degenerations are a leading cause of vision loss. RP is genetically very heterogeneous, and the multiple pathways leading to cell death are one reason for the slow progress in identifying suitable treatments for patients. Here we demonstrate that inhibition of LSD1and HDAC1 in a mouse model of RP leads to preservation of rod photoreceptors and visual function, retaining of expression of rod-specific genes, and with decreased inflammation, cell death, and Müller cell gliosis. We propose that these epigenetic inhibitors cause more open and accessible chromatin, allowing expression of neuroprotective genes. A second mechanism that allows rod photoreceptor survival is suppression of inflammation by epigenetic inhibitors in microglia. Manipulation of epigenetic modifiers is a new strategy to fight neurodegeneration in RP.
Collapse
|
39
|
Adelöf J, Wiseman J, Zetterberg M, Hernebring M. PA28α overexpressing female mice maintain exploratory behavior and capacity to prevent protein aggregation in hippocampus as they age. Aging Cell 2021; 20:e13336. [PMID: 33720528 PMCID: PMC8045925 DOI: 10.1111/acel.13336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/13/2021] [Accepted: 02/09/2021] [Indexed: 01/08/2023] Open
Abstract
With age, protein damage accumulates and increases the risk of age-related diseases. The proteasome activator PA28αβ is involved in protein damage clearance during early embryogenesis and has demonstrated protective effects against proteinopathy. We have recently discovered that adult female mice overexpressing PA28α (PA28αOE) have enhanced learning and memory, and protein extracts from their hippocampi prevent aggregation more efficiently than wild type. In this study, we investigated the effect of overexpressing PA28α on aging using C57BL/6N×BALB/c F2 hybrid mice. We found that the hippocampal anti-aggregation effect was maintained in young adult (7 months) to middle-aged (15 months) and old (22 months) PA28αOE females. While the PA28αOE influence on learning and memory gradually decreased with aging, old PA28αOE females did not display the typical drop in explorative behavior-a behavioral hallmark of aging-but were as explorative as young mice. PA28αOE lowered PA28-dependent proteasome capacity in both heart and hippocampus, and there was no indication of lower protein damage load in PA28αOE. The life span of PA28αOE was also similar to wild type. In both wild type and PA28αOE, PA28-dependent proteasome capacity increased with aging in the heart, while 26S and 20S proteasome capacities were unchanged in the timepoints analyzed. Thus, PA28αOE females exhibit improved hippocampal ability to prevent aggregation throughout life and enhanced cognitive capabilities with different behavioral outcomes dependent on age; improved memory at early age and a youth-like exploration at old age. The cognitive effects of PA28αβ combined with its anti-aggregation molecular effect highlight the therapeutical potential of PA28αβ in combating proteinopathies.
Collapse
Affiliation(s)
- Julia Adelöf
- Department of Clinical Neuroscience Institute of Neuroscience and Physiology Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
- Discovery Biology, Discovery Sciences BioPharmaceuticals R&DAstraZeneca Gothenburg Sweden
| | - John Wiseman
- Discovery Biology, Discovery Sciences BioPharmaceuticals R&DAstraZeneca Gothenburg Sweden
| | - Madeleine Zetterberg
- Department of Clinical Neuroscience Institute of Neuroscience and Physiology Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Malin Hernebring
- Department of Clinical Neuroscience Institute of Neuroscience and Physiology Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
- Discovery Biology, Discovery Sciences BioPharmaceuticals R&DAstraZeneca Gothenburg Sweden
| |
Collapse
|
40
|
Quaranta L, Bruttini C, Micheletti E, Konstas AGP, Michelessi M, Oddone F, Katsanos A, Sbardella D, De Angelis G, Riva I. Glaucoma and neuroinflammation: An overview. Surv Ophthalmol 2021; 66:693-713. [PMID: 33582161 DOI: 10.1016/j.survophthal.2021.02.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
Glaucoma is an optic neuropathy characterized by well-defined optic disc morphological changes (i.e., cup enlargement, neuroretinal border thinning, and notching, papillary vessel modifications) consequent to retinal ganglion cell loss, axonal degeneration, and lamina cribrosa remodeling. These modifications tend to be progressive and are the main cause of functional damage in glaucoma. Despite the latest findings about the pathophysiology of the disease, the exact trigger mechanisms and the mechanism of degeneration of retinal ganglion cells and their axons have not been completely elucidated. Neuroinflammation may play a role in both the development and the progression of the disease as a result of its effects on retinal environment and retinal ganglion cells. We summarize the latest findings about neuroinflammation in glaucoma and examine the connection between risk factors, neuroinflammation, and retinal ganglion cell degeneration.
Collapse
Affiliation(s)
- Luciano Quaranta
- Department of Surgical & Clinical, Diagnostic and Pediatric Sciences, Section of Ophthalmology, University of Pavia - IRCCS Fondazione Policlinico San Matteo, Pavia, Italy.
| | - Carlo Bruttini
- Department of Surgical & Clinical, Diagnostic and Pediatric Sciences, Section of Ophthalmology, University of Pavia - IRCCS Fondazione Policlinico San Matteo, Pavia, Italy
| | - Eleonora Micheletti
- Department of Surgical & Clinical, Diagnostic and Pediatric Sciences, Section of Ophthalmology, University of Pavia - IRCCS Fondazione Policlinico San Matteo, Pavia, Italy
| | - Anastasios G P Konstas
- 1st and 3rd University Departments of Ophthalmology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Andreas Katsanos
- Department of Ophthalmology, University of Ioannina, Ioannina, Greece
| | | | - Giovanni De Angelis
- Department of Surgical & Clinical, Diagnostic and Pediatric Sciences, Section of Ophthalmology, University of Pavia - IRCCS Fondazione Policlinico San Matteo, Pavia, Italy
| | | |
Collapse
|
41
|
Xu J, Zhao H, Wang T. Suppression of retinal degeneration by two novel ERAD ubiquitin E3 ligases SORDD1/2 in Drosophila. PLoS Genet 2020; 16:e1009172. [PMID: 33137101 PMCID: PMC7660902 DOI: 10.1371/journal.pgen.1009172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 11/12/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations in the gene rhodopsin are one of the major causes of autosomal dominant retinitis pigmentosa (adRP). Mutant forms of Rhodopsin frequently accumulate in the endoplasmic reticulum (ER), cause ER stress, and trigger photoreceptor cell degeneration. Here, we performed a genome-wide screen to identify suppressors of retinal degeneration in a Drosophila model of adRP, carrying a point mutation in the major rhodopsin, Rh1 (Rh1G69D). We identified two novel E3 ubiquitin ligases SORDD1 and SORDD2 that effectively suppressed Rh1G69D-induced photoreceptor dysfunction and retinal degeneration. SORDD1/2 promoted the ubiquitination and degradation of Rh1G69D through VCP (valosin containing protein) and independent of processes reliant on the HRD1 (HMG-CoA reductase degradation protein 1)/HRD3 complex. We further demonstrate that SORDD1/2 and HRD1 function in parallel and in a redundant fashion to maintain rhodopsin homeostasis and integrity of photoreceptor cells. These findings identify a new ER-associated protein degradation (ERAD) pathway and suggest that facilitating SORDD1/2 function may be a therapeutic strategy to treat adRP. Misfolded rhodopsins accumulated in endoplasmic reticulum (ER) could disrupt the homeostasis of the ER and cause ER stress. Chronic ER stress would finally lead to photoreceptor cell death and retinal degeneration. To diminish the stress and sustain homeostasis cells develop alternative strategies to clear the misfolded rhodopsins. Previous studies have suggested that ubiquitin E3 ligase HRD1 is involved in the degradation of misfolded rhodopsins. In this study, we define novel ubiquitin E3 ligase SORDD1/2 based on a genetic screen and demonstrate that SORDD1/2 promotes the degradation of misfolded rhodopsins through ER-associated degradation (ERAD) pathway. Furthermore, we demonstrate that SORDD1/2 function independently of HRD1 in misfolded rhodopsins degradation. We also show SORDD1/2 and HRD1 play redundant roles in rhodopsin homeostasis. Finally, we demonstrate that SORDD1 works well in a Drosophila disease model. Our studies identify a novel ERAD pathway that acts in parallel to HRD1, and suggest that SORDD1 is a good candidate therapeutic target.
Collapse
Affiliation(s)
- Jaiwei Xu
- College of Biological Sciences, China Agricultural University, China
- National Institute of Biological Sciences, China
| | - Haifang Zhao
- National Institute of Biological Sciences, China
| | - Tao Wang
- National Institute of Biological Sciences, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, China
- * E-mail:
| |
Collapse
|
42
|
Karlen SJ, Miller EB, Burns ME. Microglia Activation and Inflammation During the Death of Mammalian Photoreceptors. Annu Rev Vis Sci 2020; 6:149-169. [PMID: 32936734 PMCID: PMC10135402 DOI: 10.1146/annurev-vision-121219-081730] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Photoreceptors are highly specialized sensory neurons with unique metabolic and physiological requirements. These requirements are partially met by Müller glia and cells of the retinal pigment epithelium (RPE), which provide essential metabolites, phagocytose waste, and control the composition of the surrounding microenvironment. A third vital supporting cell type, the retinal microglia, can provide photoreceptors with neurotrophic support or exacerbate neuroinflammation and hasten neuronal cell death. Understanding the physiological requirements for photoreceptor homeostasis and the factors that drive microglia to best promote photoreceptor survival has important implications for the treatment and prevention of blinding degenerative diseases like retinitis pigmentosa and age-related macular degeneration.
Collapse
Affiliation(s)
- Sarah J. Karlen
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, California 95616, USA
| | - Eric B. Miller
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA
| | - Marie E. Burns
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, California 95616, USA
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
43
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
44
|
Rod function deficit in retained photoreceptors of patients with class B Rhodopsin mutations. Sci Rep 2020; 10:12552. [PMID: 32724127 PMCID: PMC7387454 DOI: 10.1038/s41598-020-69456-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/09/2020] [Indexed: 12/27/2022] Open
Abstract
A common inherited retinal disease is caused by mutations in RHO expressed in rod photoreceptors that provide vision in dim ambient light. Approximately half of all RHO mutations result in a Class B phenotype where mutant rods are retained in some retinal regions but show severe degeneration in other regions. We determined the natural history of dysfunction and degeneration of retained rods by serially evaluating patients. Even when followed for more than 20 years, rod function and structure at some retinal locations could remain unchanged. Other locations showed loss of both vision and photoreceptors but the rate of rod vision loss was greater than the rate of photoreceptor degeneration. This unexpected divergence in rates with disease progression implied the development of a rod function deficit beyond loss of cells. The divergence of progression rates was also detectable over a short interval of 2 years near the health-disease transition in the superior retina. A model of structure–function relationship supported the existence of a large rod function deficit which was also most prominent near regions of health-disease transition. Our studies support the realistic therapeutic goal of improved night vision for retinal regions specifically preselected for rod function deficit in patients.
Collapse
|
45
|
Sharkey LM, Sandoval-Pistorius SS, Moore SJ, Gerson JE, Komlo R, Fischer S, Negron-Rios KY, Crowley EV, Padron F, Patel R, Murphy GG, Paulson HL. Modeling UBQLN2-mediated neurodegenerative disease in mice: Shared and divergent properties of wild type and mutant UBQLN2 in phase separation, subcellular localization, altered proteostasis pathways, and selective cytotoxicity. Neurobiol Dis 2020; 143:105016. [PMID: 32653673 DOI: 10.1016/j.nbd.2020.105016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/26/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
The ubiquitin-binding proteasomal shuttle protein UBQLN2 is implicated in common neurodegenerative disorders due to its accumulation in disease-specific aggregates and, when mutated, directly causes familial frontotemporal dementia/amyotrophic lateral sclerosis (FTD/ALS). Like other proteins linked to FTD/ALS, UBQLN2 undergoes phase separation to form condensates. The relationship of UBQLN2 phase separation and accumulation to neurodegeneration, however, remains uncertain. Employing biochemical, neuropathological and behavioral assays, we studied the impact of overexpressing WT or mutant UBQLN2 in the CNS of transgenic mice. Expression of UBQLN2 harboring a pathogenic mutation (P506T) elicited profound and widespread intraneuronal inclusion formation and aggregation without prominent neurodegenerative or behavioral changes. Both WT and mutant UBQLN2 formed ubiquitin- and P62-positive inclusions in neurons, supporting the view that UBQLN2 is intrinsically prone to phase separate, with the size, shape and frequency of inclusions depending on expression level and the presence or absence of a pathogenic mutation. Overexpression of WT or mutant UBQLN2 resulted in a dose-dependent decrease in levels of a key interacting chaperone, HSP70, as well as dose-dependent profound degeneration of the retina. We conclude that, at least in mice, robust aggregation of a pathogenic form of UBQLN2 is insufficient to cause neuronal loss recapitulating that of human FTD/ALS. Our results nevertheless support the view that altering the normal cellular balance of UBQLN2, whether wild type or mutant protein, has deleterious effects on cells of the CNS and retina that likely reflect perturbations in ubiquitin-dependent protein homeostasis.
Collapse
Affiliation(s)
- Lisa M Sharkey
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America.
| | - Stephanie S Sandoval-Pistorius
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Shannon J Moore
- Michigan Neuroscience Institute and Department of Physiology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Julia E Gerson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Robert Komlo
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Svetlana Fischer
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Keyshla Y Negron-Rios
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Emily V Crowley
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Francisco Padron
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Ronak Patel
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Geoffrey G Murphy
- Michigan Neuroscience Institute and Department of Physiology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, United States of America.
| |
Collapse
|
46
|
Wang X, Wang H. Priming the Proteasome to Protect against Proteotoxicity. Trends Mol Med 2020; 26:639-648. [PMID: 32589934 PMCID: PMC7321925 DOI: 10.1016/j.molmed.2020.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Increased proteotoxic stress (IPTS) resulting from the increased production or decreased removal of abnormally folded proteins is recognized as an important pathogenic factor for a large group of highly disabling and life-threatening human diseases, such as neurodegenerative disorders and many heart diseases. The proteasome is pivotal to the timely removal of abnormal proteins but its functional capacity often becomes inadequate in the disease conditions; consequently, proteasome functional insufficiency in return exacerbates IPTS. Recent research in proteasome biology reveals that the proteasome can be activated by endogenous protein kinases, making it possible to pharmacologically prime the proteasome for treating diseases with IPTS.
Collapse
Affiliation(s)
- Xuejun Wang
- University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA.
| | - Hongmin Wang
- University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| |
Collapse
|
47
|
Liu X, Feng B, Vats A, Tang H, Seibel W, Swaroop M, Tawa G, Zheng W, Byrne L, Schurdak M, Chen Y. Pharmacological clearance of misfolded rhodopsin for the treatment of RHO-associated retinitis pigmentosa. FASEB J 2020; 34:10146-10167. [PMID: 32536017 DOI: 10.1096/fj.202000282r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 01/20/2023]
Abstract
Rhodopsin mutation and misfolding is a common cause of autosomal dominant retinitis pigmentosa (RP). Using a luciferase reporter assay, we undertook a small-molecule high-throughput screening (HTS) of 68, 979 compounds and identified nine compounds that selectively reduced the misfolded P23H rhodopsin without an effect on the wild type (WT) rhodopsin protein. Further, we found five of these compounds, including methotrexate (MTX), promoted P23H rhodopsin degradation that also cleared out other misfolded rhodopsin mutant proteins. We showed MTX increased P23H rhodopsin degradation via the lysosomal but not the proteasomal pathway. Importantly, one intravitreal injection (IVI) of 25 pmol MTX increased electroretinogram (ERG) response and rhodopsin level in the retinae of RhoP23H/+ knock-in mice at 1 month of age. Additionally, four weekly IVIs increased the photoreceptor cell number in the retinae of RhoP23H/+ mice compared to vehicle control. Our study indicates a therapeutic potential of repurposing MTX for the treatment of rhodopsin-associated RP.
Collapse
Affiliation(s)
- Xujie Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bing Feng
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abhishek Vats
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hong Tang
- Drug Discovery Center, University of Cincinnati, Cincinnati, OH, USA
| | - William Seibel
- Drug Discovery Center, University of Cincinnati, Cincinnati, OH, USA.,Oncology Department, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Manju Swaroop
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Gregory Tawa
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Leah Byrne
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Schurdak
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
48
|
Finkelstein S, Gospe SM, Schuhmann K, Shevchenko A, Arshavsky VY, Lobanova ES. Phosphoinositide Profile of the Mouse Retina. Cells 2020; 9:cells9061417. [PMID: 32517352 PMCID: PMC7349851 DOI: 10.3390/cells9061417] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/24/2022] Open
Abstract
Phosphoinositides are known to play multiple roles in eukaryotic cells. Although dysregulation of phosphoinositide metabolism in the retina has been reported to cause visual dysfunction in animal models and human patients, our understanding of the phosphoinositide composition of the retina is limited. Here, we report a characterization of the phosphoinositide profile of the mouse retina and an analysis of the subcellular localization of major phosphorylated phosphoinositide forms in light-sensitive photoreceptor neurons. Using chromatography of deacylated phosphatidylinositol headgroups, we established PI(4,5)P2 and PI(4)P as two major phosphorylated phosphoinositides in the retina. Using high-resolution mass spectrometry, we revealed 18:0/20:4 and 16:0/20:4 as major fatty-acyl chains of retinal phosphoinositides. Finally, analysis of fluorescent phosphoinositide sensors in rod photoreceptors demonstrated distinct subcellular distribution patterns of major phosphoinositides. The PI(4,5)P2 reporter was enriched in the inner segments and synapses, but was barely detected in the light-sensitive outer segments. The PI(4)P reporter was mostly found in the outer and inner segments and the areas around nuclei, but to a lesser degree in the synaptic region. These findings provide support for future mechanistic studies defining the biological significance of major mono- (PI(4)P) and bisphosphate (PI(4,5)P2) phosphatidylinositols in photoreceptor biology and retinal health.
Collapse
Affiliation(s)
- Stella Finkelstein
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; (S.F.); (S.M.G.III); (V.Y.A.)
| | - Sidney M. Gospe
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; (S.F.); (S.M.G.III); (V.Y.A.)
| | - Kai Schuhmann
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; (K.S.); (A.S.)
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; (K.S.); (A.S.)
| | - Vadim Y. Arshavsky
- Department of Ophthalmology, Duke University, Durham, NC 27710, USA; (S.F.); (S.M.G.III); (V.Y.A.)
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Ekaterina S. Lobanova
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
- Correspondence:
| |
Collapse
|
49
|
Aguilà M, Bellingham J, Athanasiou D, Bevilacqua D, Duran Y, Maswood R, Parfitt DA, Iwawaki T, Spyrou G, Smith AJ, Ali RR, Cheetham ME. AAV-mediated ERdj5 overexpression protects against P23H rhodopsin toxicity. Hum Mol Genet 2020; 29:1310-1318. [PMID: 32196553 PMCID: PMC7254845 DOI: 10.1093/hmg/ddaa049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/17/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023] Open
Abstract
Rhodopsin misfolding caused by the P23H mutation is a major cause of autosomal dominant retinitis pigmentosa (adRP). To date, there are no effective treatments for adRP. The BiP co-chaperone and reductase ERdj5 (DNAJC10) is part of the endoplasmic reticulum (ER) quality control machinery, and previous studies have shown that overexpression of ERdj5 in vitro enhanced the degradation of P23H rhodopsin, whereas knockdown of ERdj5 increased P23H rhodopsin ER retention and aggregation. Here, we investigated the role of ERdj5 in photoreceptor homeostasis in vivo by using an Erdj5 knockout mouse crossed with the P23H knock-in mouse and by adeno-associated viral (AAV) vector-mediated gene augmentation of ERdj5 in P23H-3 rats. Electroretinogram (ERG) and optical coherence tomography of Erdj5-/- and P23H+/-:Erdj5-/- mice showed no effect of ERdj5 ablation on retinal function or photoreceptor survival. Rhodopsin levels and localization were similar to those of control animals at a range of time points. By contrast, when AAV2/8-ERdj5-HA was subretinally injected into P23H-3 rats, analysis of the full-field ERG suggested that overexpression of ERdj5 reduced visual function loss 10 weeks post-injection (PI). This correlated with a significant preservation of photoreceptor cells at 4 and 10 weeks PI. Assessment of the outer nuclear layer (ONL) morphology showed preserved ONL thickness and reduced rhodopsin retention in the ONL in the injected superior retina. Overall, these data suggest that manipulation of the ER quality control and ER-associated degradation factors to promote mutant protein degradation could be beneficial for the treatment of adRP caused by mutant rhodopsin.
Collapse
Affiliation(s)
| | | | | | | | - Yanai Duran
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | - Ryea Maswood
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | | | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada, 920-0293, Japan
| | - Giannis Spyrou
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, 581 83, Sweden
| | | | - Robin R Ali
- UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | | |
Collapse
|
50
|
Lewis TR, Shores CR, Cady MA, Hao Y, Arshavsky VY, Burns ME. The F220C and F45L rhodopsin mutations identified in retinitis pigmentosa patients do not cause pathology in mice. Sci Rep 2020; 10:7538. [PMID: 32371886 PMCID: PMC7200662 DOI: 10.1038/s41598-020-64437-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/18/2020] [Indexed: 11/12/2022] Open
Abstract
Retinitis pigmentosa is a retinal degenerative disease that leads to blindness through photoreceptor loss. Rhodopsin is the most frequently mutated protein in this disease. While many rhodopsin mutations have well-understood consequences that lead to cell death, the disease association of several rhodopsin mutations identified in retinitis pigmentosa patients, including F220C and F45L, has been disputed. In this study, we generated two knockin mouse lines bearing each of these mutations. We did not observe any photoreceptor degeneration in either heterozygous or homozygous animals of either line. F220C mice exhibited minor disruptions of photoreceptor outer segment dimensions without any mislocalization of outer segment proteins, whereas photoreceptors of F45L mice were normal. Suction electrode recordings from individual photoreceptors of both mutant lines showed normal flash sensitivity and photoresponse kinetics. Taken together, these data suggest that neither the F220C nor F45L mutation has pathological consequences in mice and, therefore, may not be causative of retinitis pigmentosa in humans.
Collapse
Affiliation(s)
- Tylor R Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Camilla R Shores
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, United States
| | - Martha A Cady
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, 27710, United States.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, United States
| | - Marie E Burns
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95616, United States. .,Center for Neuroscience and Department of Ophthalmology & Vision Science, University of California, Davis, CA, 95616, United States.
| |
Collapse
|