1
|
Chen Y, Yang X, Mao J. The Neuroprotective Effect of Activation of Sigma-1 Receptor on Neural Injury by Optic Nerve Crush. Invest Ophthalmol Vis Sci 2023; 64:9. [PMID: 37669061 PMCID: PMC10484044 DOI: 10.1167/iovs.64.12.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/09/2023] [Indexed: 09/06/2023] Open
Abstract
Purpose This study aimed to explore the neuroprotective effects of sigma-1 receptor (S1R) on optic nerve crush (ONC) mice by upregulating its expression through intravitreal injection of adeno-associated virus (AAV). Methods The animals were divided into four groups. Mice that underwent ONC were administered an intravitreal injection with blank vector (ONC group), with AAV targeting downregulation of S1R (S1R-sh group), or with AAV targeting overexpression of S1R (S1R-AAV group). Mice in the control group underwent intravitreal injection with blank vector. The thickness of each layer of the retina was measured through optical coherence tomography, and the apoptotic rate of retinal neurons was determined using the TUNEL assay. The expression levels of brain-derived neurotrophic factor (BDNF) and S1R were quantified through western blot. Electroretinogram (ERG) was performed to evaluate the visual function. Results The thickness of the total retina (P = 0.001), ganglion cell layer (P = 0.017), and inner nuclear layer (P = 0.002) in S1R-AAV group was significantly thicker than that of the ONC group. The number of retinal apoptotic cells in the S1R-AAV group was 23% lower than that in the ONC group (P = 0.002). ERG results showed that, compared to the ONC group, the amplitudes of the a- and b-waves were higher in the S1R-AAV group (a-wave, P < 0.001; b-wave, P = 0.007). Western blot showed that the expression of BDNF in the S1R-AAV group was higher than that in the ONC group (P < 0.001). Conclusions Activation of S1R in the retina through intravitreal injection of AAV can effectively maintain the retina structure, promote neuronal cell survival, and protect visual function.
Collapse
Affiliation(s)
- Yao Chen
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Chang Sha, China
| | - Xueli Yang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Chang Sha, China
| | - Junfeng Mao
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Chang Sha, China
| |
Collapse
|
2
|
Wang J, Barwick SR, Xiao H, Smith SB. Evaluation of the role of Sigma 1 receptor and Cullin3 in retinal photoreceptor cells. Free Radic Biol Med 2023; 205:214-223. [PMID: 37328017 PMCID: PMC10527355 DOI: 10.1016/j.freeradbiomed.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/24/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Sigma 1 receptor (Sig1R), a pluripotent modulator of cell survival, is neuroprotective in models of retinal degeneration when activated by the high-affinity, high-specificity ligand (+)-pentazocine ((+)-PTZ). The molecular mechanisms of Sig1R-mediated retinal neuroprotection are under investigation. We previously reported that the antioxidant regulatory transcription factor Nrf2 may be involved in Sig1R-mediated retinal photoreceptor cell (PRC) rescue. Cullin 3 (Cul3) is a component of the Nrf2-Keap1 antioxidant pathway and facilitates Nrf2 ubiquitination. Our earlier transcriptome analysis revealed decreased Cul3 in retinas lacking Sig1R. Here, we asked whether Sig1R activation can modulate Cul3 expression in 661 W cone PRCs. Proximity ligation and co-immunoprecipitation (co-IP) showed that Cul3 resides closely to and co-IPs with Sig1R. Activation of Sig1R using (+)-PTZ significantly increased Cul3 at the gene/protein level; silencing Sig1R decreased Cul3 gene/protein levels. Experiments in which Cul3 was silenced in cells exposed to tBHP resulted in increased oxidative stress, which was not attenuated with Sig1R activation by (+)-PTZ, whereas cells transfected with scrambled siRNA (and incubated with tBHP) responded to (+)-PTZ treatment by decreasing levels of oxidative stress. Assessment of mitochondrial respiration and glycolysis revealed significantly improved maximal respiration, spare capacity and glycolytic capacity in oxidatively-stressed cells transfected with scrambled siRNA and treated with (+)-PTZ, but not in (+)-PTZ treated, oxidatively-stressed cells in which Cul3 had been silenced. The data provide the first evidence that Sig1R co-localizes/interacts with Cul3, a key player in the Nrf2-Keap1 antioxidant pathway. The data suggest that the preservation of mitochondrial respiration/glycolytic function and reduction of oxidative stress observed upon activation of Sig1R occur in part in a Cul3-dependent manner.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Shannon R Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; 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; 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
|
3
|
Wang H, Peng Z, Li Y, Sahn JJ, Hodges TR, Chou TH, Liu Q, Zhou X, Jiao S, Porciatti V, Liebl DJ, Martin SF, Wen R. σ 2R/TMEM97 in retinal ganglion cell degeneration. Sci Rep 2022; 12:20753. [PMID: 36456686 PMCID: PMC9715665 DOI: 10.1038/s41598-022-24537-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
The sigma 2 receptor (σ2R) was recently identified as an endoplasmic reticulum (ER) membrane protein known as transmembrane protein 97 (TMEM97). Studies have shown that σ2R/TMEM97 binding compounds are neuroprotective, suggesting a role of σ2R/TMEM97 in neurodegenerative processes. To understand the function of σ2R/TMEM97 in neurodegeneration pathways, we characterized ischemia-induced retinal ganglion cell (RGC) degeneration in TMEM97-/- mice and found that RGCs in TMEM97-/- mice are resistant to degeneration. In addition, intravitreal injection of a selective σ2R/TMEM97 ligand DKR-1677 significantly protects RGCs from ischemia-induced degeneration in wildtype mice. Our results provide conclusive evidence that σ2R/TMEM97 plays a role to facilitate RGC death following ischemic injury and that inhibiting the function of σ2R/TMEM97 is neuroprotective. This work is a breakthrough toward elucidating the biology and function of σ2R/TMEM97 in RGCs and likely in other σ2R/TMEM97 expressing neurons. Moreover, these findings support future studies to develop new neuroprotective approaches for RGC degenerative diseases by inhibiting σ2R/TMEM97.
Collapse
Affiliation(s)
- Hua Wang
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Zhiyou Peng
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Yiwen Li
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - James J Sahn
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Timothy R Hodges
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Tsung-Han Chou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Qiong Liu
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Xuezhi Zhou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Shuliang Jiao
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Daniel J Liebl
- Department of Neurosurgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Stephen F Martin
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Rong Wen
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
4
|
Xu Z, Lei Y, Qin H, Zhang S, Li P, Yao K. Sigma-1 Receptor in Retina: Neuroprotective Effects and Potential Mechanisms. Int J Mol Sci 2022; 23:ijms23147572. [PMID: 35886921 PMCID: PMC9321618 DOI: 10.3390/ijms23147572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Retinal degenerative diseases are the major factors leading to severe visual impairment and even irreversible blindness worldwide. The therapeutic approach for retinal degenerative diseases is one extremely urgent and hot spot in science research. The sigma-1 receptor is a novel, multifunctional ligand-mediated molecular chaperone residing in endoplasmic reticulum (ER) membranes and the ER-associated mitochondrial membrane (ER-MAM); it is widely distributed in numerous organs and tissues of various species, providing protective effects on a variety of degenerative diseases. Over three decades, considerable research has manifested the neuroprotective function of sigma-1 receptor in the retina and has attempted to explore the molecular mechanism of action. In the present review, we will discuss neuroprotective effects of the sigma-1 receptor in retinal degenerative diseases, mainly in aspects of the following: the localization in different types of retinal neurons, the interactions of sigma-1 receptors with other molecules, the correlated signaling pathways, the influence of sigma-1 receptors to cellular functions, and the potential therapeutic effects on retinal degenerative diseases.
Collapse
|
5
|
Huang L, Xiao H, Xie X, Hu F, Tang F, Smith SB, Gan L. Generation of Sigmar1 conditional knockout mouse using CRISPR-Cas9 gene targeting. Genesis 2022; 60:e23487. [PMID: 35633570 DOI: 10.1002/dvg.23487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/01/2022] [Accepted: 04/25/2022] [Indexed: 11/08/2022]
Abstract
The Sigma 1 receptor (SIGMAR1) is a transmembrane protein located in the mitochondria-associated endoplasmic reticulum membrane, and plays an important role in cell survival as a pluripotent modulator of a variety of signaling pathways related to neurodegeneration. Though SIGMAR1 is a potential target for neurodegenerative diseases, the specific role of SIGMAR1 in different tissue and cell types remains unclear. Here we reported the generation of Sigmar1 conditional knockout (Sigmar1loxP ) mice using CRISPR-Cas9 method to insert loxP sites into the 5'- and 3'-untranslated regions of Sigmar1. We showed that the insertion of loxP sequences did not affect the expression of Sigmar1 and that Sigmar1loxP/loxP mice exhibited no detectable visual defects compared with wild-type mice at the early adult stage. By crossing Sigmar1loxP mice with retina-specific Six3-Cre and ubiquitous CMV-Cre mice, we confirmed the deletion of Sigmar1 coding regions of exons 1-4, and the retina-specific and global loss of SIGMAR1 expression, respectively. Thus, Sigmar1loxP mice provide a valuable tool for unraveling the tissue and cell-type-specific role of Sigmar1.
Collapse
Affiliation(s)
- Liang Huang
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA
| | - Haiyan Xiao
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA.,Department of Cellular Biology and Anatomy, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Xiaoling Xie
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Fang Hu
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Fulei Tang
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Sylvia B Smith
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA.,Department of Cellular Biology and Anatomy, Augusta University Medical College of Georgia, Augusta, Georgia, USA
| | - Lin Gan
- Department of Neuroscience and Regenerative Medicine, Augusta University Medical College of Georgia, Augusta, Georgia, USA.,Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
6
|
Couly S, Goguadze N, Yasui Y, Kimura Y, Wang SM, Sharikadze N, Wu HE, Su TP. Knocking Out Sigma-1 Receptors Reveals Diverse Health Problems. Cell Mol Neurobiol 2022; 42:597-620. [PMID: 33095392 PMCID: PMC8062587 DOI: 10.1007/s10571-020-00983-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Sigma-1 receptor (Sig-1R) is a protein present in several organs such as brain, lung, and heart. In a cell, Sig-1R is mainly located across the membranes of the endoplasmic reticulum and more specifically at the mitochondria-associated membranes. Despite numerous studies showing that Sig-1R could be targeted to rescue several cellular mechanisms in different pathological conditions, less is known about its fundamental relevance. In this review, we report results from various studies and focus on the importance of Sig-1R in physiological conditions by comparing Sig-1R KO mice to wild-type mice in order to investigate the fundamental functions of Sig-1R. We note that the Sig-1R deletion induces cognitive, psychiatric, and motor dysfunctions, but also alters metabolism of heart. Finally, taken together, observations from different experiments demonstrate that those dysfunctions are correlated to poor regulation of ER and mitochondria metabolism altered by stress, which could occur with aging.
Collapse
Affiliation(s)
- Simon Couly
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA.
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Nino Sharikadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| |
Collapse
|
7
|
Zhao J, Gonsalvez GB, Mysona BA, Smith SB, Bollinger KE. Sigma 1 Receptor Contributes to Astrocyte-Mediated Retinal Ganglion Cell Protection. Invest Ophthalmol Vis Sci 2022; 63:1. [PMID: 35103752 PMCID: PMC8819349 DOI: 10.1167/iovs.63.2.1] [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: 10/31/2021] [Accepted: 12/28/2021] [Indexed: 01/22/2023] Open
Abstract
Purpose Sigma 1 receptor (S1R) is expressed in retinal ganglion cells (RGCs) and astrocytes, and its activation is neuroprotective. We evaluated the contribution of S1R within optic nerve head astrocytes (ONHAs) to growth and survival of RGCs in vitro. Methods Wild-type (WT) RGCs and WT or S1R knockout (S1R KO) ONHAs were cocultured for 2, 4, or 7 days. Total and maximal neurite length, neurite root, and extremity counts were measured. Cell death was measured using a TUNEL assay. Signal transducer and activator of transcription 3 phosphorylation levels were evaluated in ONHA-derived lysates by immunoblotting. Results The coculture of WT RGCs with WT or S1R KO ONHAs increased the total and maximal neurite length. Neurite root and extremity counts increased at 4 and 7 days when WT RGCs were cocultured with WT or S1R KO ONHAs. At all timepoints, the total and maximal neurite length decreased for WT RGCs in coculture with S1R KO ONHAs compared with WT ONHAs. Root and extremity counts decreased for WT RGCs in coculture with S1R KO ONHAs compared with WT ONHAs at 2 and 7, but not 4 days. RGC apoptosis increased in S1R KO ONHA coculture and S1R KO-conditioned medium, compared with WT ONHA coculture or WT-conditioned medium. S1R KO ONHA-derived lysates showed decreased phosphorylated signal transducer and activator of transcription 3 levels compared with WT ONHA-derived lysates. Conclusions The absence of S1R within ONHAs has a deleterious effect on RGC neurite growth and RGC survival, reflected in analysis of WT RGC + S1R KO ONHA indirect cocultures. The data suggest that S1R may enhance ganglion cell survival via glia-mediated mechanisms.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| | | | - Barbara A. Mysona
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| | - Sylvia B. Smith
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| | - Kathryn E. Bollinger
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| |
Collapse
|
8
|
Sałaciak K, Pytka K. Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neurosci Biobehav Rev 2022; 132:1114-1136. [PMID: 34736882 PMCID: PMC8559442 DOI: 10.1016/j.neubiorev.2021.10.037] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022]
Abstract
Depression and cognitive disorders are diseases with complex and not-fully understood etiology. Unfortunately, the COVID-19 pandemic dramatically increased the prevalence of both conditions. Since the current treatments are inadequate in many patients, there is a constant need for discovering new compounds, which will be more effective in ameliorating depressive symptoms and treating cognitive decline. Proteins attracting much attention as potential targets for drugs treating these conditions are sigma-1 receptors. Sigma-1 receptors are multi-functional proteins localized in endoplasmic reticulum membranes, which play a crucial role in cellular signal transduction by interacting with receptors, ion channels, lipids, and kinases. Changes in their functions and expression may lead to various diseases, including depression or memory impairments. Thus, sigma-1 receptor modulation might be useful in treating these central nervous system diseases. Importantly, two sigma-1 receptor ligands entered clinical trials, showing that this compound group possesses therapeutic potential. Therefore, based on preclinical studies, this review discusses whether the sigma-1 receptor could be a promising target for drugs treating affective and cognitive disorders.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
9
|
Geva M, Gershoni-Emek N, Naia L, Ly P, Mota S, Rego AC, Hayden MR, Levin LA. Neuroprotection of retinal ganglion cells by the sigma-1 receptor agonist pridopidine in models of experimental glaucoma. Sci Rep 2021; 11:21975. [PMID: 34753986 PMCID: PMC8578336 DOI: 10.1038/s41598-021-01077-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/20/2021] [Indexed: 12/21/2022] Open
Abstract
Optic neuropathies such as glaucoma are characterized by retinal ganglion cell (RGC) degeneration and death. The sigma-1 receptor (S1R) is an attractive target for treating optic neuropathies as it is highly expressed in RGCs, and its absence causes retinal degeneration. Activation of the S1R exerts neuroprotective effects in models of retinal degeneration. Pridopidine is a highly selective and potent S1R agonist in clinical development. We show that pridopidine exerts neuroprotection of retinal ganglion cells in two different rat models of glaucoma. Pridopidine strongly binds melanin, which is highly expressed in the retina. This feature of pridopidine has implications to its ocular distribution, bioavailability, and effective dose. Mitochondria dysfunction is a key contributor to retinal ganglion cell degeneration. Pridopidine rescues mitochondrial function via activation of the S1R, providing support for the potential mechanism driving its neuroprotective effect in retinal ganglion cells.
Collapse
Affiliation(s)
| | | | - Luana Naia
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
| | - Philip Ly
- The Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Sandra Mota
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- FMUC-Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Michael R Hayden
- Prilenia Therapeutics, Herzliya, Israel
- The Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Leonard A Levin
- Department of Ophthalmology and Visual Sciences, McGill University, Montreal, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.
- Montreal Neurological Institute, McGill University, Montreal, Canada.
| |
Collapse
|
10
|
Wu NH, Ye Y, Wan BB, Yu YD, Liu C, Chen QJ. Emerging Benefits: Pathophysiological Functions and Target Drugs of the Sigma-1 Receptor in Neurodegenerative Diseases. Mol Neurobiol 2021; 58:5649-5666. [PMID: 34383254 DOI: 10.1007/s12035-021-02524-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023]
Abstract
The sigma-1 receptor (Sig-1R) is encoded by the SIGMAR1 gene and is a nonopioid transmembrane receptor located in the mitochondrial-associated endoplasmic reticulum membrane (MAM). It helps to locate endoplasmic reticulum calcium channels, regulates calcium homeostasis, and acts as a molecular chaperone to control cell fate and participate in signal transduction. It plays an important role in protecting neurons through a variety of signaling pathways and participates in the regulation of cognition and motor behavior closely related to neurodegenerative diseases. Based on its neuroprotective effects, Sig-1R has now become a breakthrough target for alleviating Alzheimer's disease and other neurodegenerative diseases. This article reviews the most cutting-edge research on the function of Sig-1R under normal or pathologic conditions and target drugs of the sigma-1 receptor in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ning-Hua Wu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
- Basic Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Yu Ye
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Bin-Bin Wan
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Yuan-Dong Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| | - Qing-Jie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| |
Collapse
|
11
|
Li L, He S, Liu Y, Yorio T, Ellis DZ. Sigma-1R Protects Retinal Ganglion Cells in Optic Nerve Crush Model for Glaucoma. Invest Ophthalmol Vis Sci 2021; 62:17. [PMID: 34406331 PMCID: PMC8375012 DOI: 10.1167/iovs.62.10.17] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/28/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to determine the effects of the Sigma-1R (σ-1r) on retinal ganglion cell (RGC) survival following optic nerve crush (ONC) and the signaling mechanism involved in the σ-1r protection. Methods The overall strategy was to induce injury by ONC and mitigate RGC death by increasing σ-1r expression and/or activate σ-1r activity in σ-1r K/O mice and wild type (WT) mice. AAV2-σ-1r vector was used to increase σ-1r expression and σ-1r agonist used to activate the σ-1r and RGCs were counted. Immunohistochemical and Western blot analysis determined phosphorylated (p)-c-Jun, c-Jun, and Caspase-3. Pattern electroretinography (PERG) determined RGC activity. Results RGC counts and function were similar in pentazocine-treated WT mice when compared to untreated mice and in WT mice when compared with σ-1r K/O mice. Pentazocine-induced effects and the effects of σ-1r K/O were only observable after ONC. ONC resulted in decreased RGC counts and activity in both WT and σ-1r K/O mice, with σ-1r K/O mice experiencing significant decreases compared with WT mice. The σ-1r transgenic expression resulted in increased RGC counts and activity following ONC. In WT mice, treatment with σ-1r agonist pentazocine resulted in increased RGC counts and increased activity when compared with untreated WT mice. There were time-dependent increases in c-jun, p-c-jun, and caspase-3 expression in ONC mice that were mitigated with pentazocine-treatment. Conclusions These findings suggest that the apoptotic pathway is involved in RGC losses seen in an ONC model. The σ-1r offers neuroprotection, as activation and/or transgenic expression of σ-1r attenuated the apoptotic pathway and restored RGCs number and function following ONC.
Collapse
Affiliation(s)
- Linya Li
- Department of Pharmacology and Neuroscience University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Shaoqing He
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Yang Liu
- Department of Pharmacology and Neuroscience University of North Texas Health Science Center, Fort Worth, Texas, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Thomas Yorio
- Department of Pharmacology and Neuroscience University of North Texas Health Science Center, Fort Worth, Texas, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Dorette Z Ellis
- Department of Pharmaceutical Sciences, University of North Texas Systems College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
12
|
Aishwarya R, Abdullah CS, Morshed M, Remex NS, Bhuiyan MS. Sigmar1's Molecular, Cellular, and Biological Functions in Regulating Cellular Pathophysiology. Front Physiol 2021; 12:705575. [PMID: 34305655 PMCID: PMC8293995 DOI: 10.3389/fphys.2021.705575] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The Sigma 1 receptor (Sigmar1) is a ubiquitously expressed multifunctional inter-organelle signaling chaperone protein playing a diverse role in cellular survival. Recessive mutation in Sigmar1 have been identified as a causative gene for neuronal and neuromuscular disorder. Since the discovery over 40 years ago, Sigmar1 has been shown to contribute to numerous cellular functions, including ion channel regulation, protein quality control, endoplasmic reticulum-mitochondrial communication, lipid metabolism, mitochondrial function, autophagy activation, and involved in cellular survival. Alterations in Sigmar1’s subcellular localization, expression, and signaling has been implicated in the progression of a wide range of diseases, such as neurodegenerative diseases, ischemic brain injury, cardiovascular diseases, diabetic retinopathy, cancer, and drug addiction. The goal of this review is to summarize the current knowledge of Sigmar1 biology focusing the recent discoveries on Sigmar1’s molecular, cellular, pathophysiological, and biological functions.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
13
|
Barwick SR, Siddiq MS, Wang J, Xiao H, Marshall B, Perry E, Smith SB. Sigma 1 Receptor Co-Localizes with NRF2 in Retinal Photoreceptor Cells. Antioxidants (Basel) 2021; 10:antiox10060981. [PMID: 34205384 PMCID: PMC8234060 DOI: 10.3390/antiox10060981] [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: 05/26/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022] Open
Abstract
Sigma 1 receptor (Sig1R), a modulator of cell survival, has emerged as a novel target for retinal degenerative disease. Studies have shown that activation of Sig1R, using the high affinity ligand (+)-pentazocine ((+)-PTZ), improves cone function in a severe retinopathy model. The rescue is accompanied by normalization of levels of NRF2, a key transcription factor that regulates the antioxidant response. The interaction of Sig1R with a number of proteins has been investigated; whether it interacts with NRF2, however, is not known. We used co-immunoprecipitation (co-IP), proximity ligation assay (PLA), and electron microscopy (EM) immunodetection methods to investigate this question in the 661W cone photoreceptor cell line. For co-IP experiments, immune complexes were precipitated by protein A/G agarose beads and immunodetected using anti-NRF2 antibody. For PLA, cells were incubated with anti-Sig1R polyclonal and anti-NRF2 monoclonal antibodies, then subsequently with (−)-mouse and (+)-rabbit PLA probes. For EM analysis, immuno-EM gold labeling was performed using nanogold-enhanced labeling with anti-NRF2 and anti-Sig1R antibodies, and data were confirmed using colloidal gold labeling. The co-IP experiment suggested that NRF2 was bound in a complex with Sig1R. The PLA assays detected abundant orange fluorescence in cones, indicating that Sig1R and NRF2 were within 40 nm of each other. EM immunodetection confirmed co-localization of Sig1R with NRF2 in cells and in mouse retinal tissue. This study is the first to report co-localization of Sig1R-NRF2 and supports earlier studies implicating modulation of NRF2 as a mechanism by which Sig1R mediates retinal neuroprotection.
Collapse
Affiliation(s)
- Shannon R. Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Mevish S. Siddiq
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Brendan Marshall
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
| | - Elizabeth Perry
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (S.R.B.); (M.S.S.); (J.W.); (H.X.); (B.M.); (E.P.)
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-706-721-7392; Fax: +1-706-721-6120
| |
Collapse
|
14
|
Zhao J, Gonsalvez G, Bartoli M, Mysona BA, Smith SB, Bollinger KE. Sigma 1 Receptor Modulates Optic Nerve Head Astrocyte Reactivity. Invest Ophthalmol Vis Sci 2021; 62:5. [PMID: 34086045 PMCID: PMC8185400 DOI: 10.1167/iovs.62.7.5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/03/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose Stimulation of Sigma 1 Receptor (S1R) is neuroprotective in retina and optic nerve. S1R is expressed in both neurons and glia. The purpose of this work is to evaluate the ability of S1R to modulate reactivity responses of optic nerve head astrocytes (ONHAs) by investigating the extent to which S1R activation alters ONHA reactivity under conditions of ischemic cellular stress. Methods Wild type (WT) and S1R knockout (KO) ONHAs were derived and treated with vehicle or S1R agonist, (+)-pentazocine ((+)-PTZ). Cells were subjected to six hours of oxygen glucose deprivation (OGD) followed by 18 hours of re-oxygenation (OGD/R). Astrocyte reactivity responses were measured. Molecules that regulate ONHA reactivity, signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa B (NF-kB), were evaluated. Results Baseline glial fibrillary acidic protein (GFAP) levels were increased in nonstressed KO ONHAs compared with WT cultures. Baseline cellular migration was also increased in nonstressed KO ONHAs compared with WT. Treatment with (+)-PTZ increased cellular migration in nonstressed WT ONHAs but not in KO ONHAs. Exposure of both WT and KO ONHAs to ischemia (OGD/R), increased GFAP levels and cellular proliferation. However, (+)-PTZ treatment of OGD/R-exposed ONHAs enhanced GFAP levels, cellular proliferation, and cellular migration in WT but not KO cultures. The (+)-PTZ treatment of WT ONHAs also enhanced the OGD/R-induced increase in cellular pSTAT3 levels. However, treatment of WT ONHAs with (+)-PTZ abrogated the OGD/R-induced rise in NF-kB(p65) activation. Conclusions Under ischemic stress conditions, S1R activation enhanced ONHA reactivity characteristics. Future studies should address effects of these responses on RGC survival.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| | - Graydon Gonsalvez
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Manuela Bartoli
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| | - Barbara A. Mysona
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| | - Sylvia B. Smith
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| | - Kathryn E. Bollinger
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta, Georgia, United States
| |
Collapse
|
15
|
Wang J, Xiao H, Barwick SR, Smith SB. Comparison of Sigma 1 Receptor Ligands SA4503 and PRE084 to (+)-Pentazocine in the rd10 Mouse Model of RP. Invest Ophthalmol Vis Sci 2020; 61:3. [PMID: 33137196 PMCID: PMC7645203 DOI: 10.1167/iovs.61.13.3] [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: 06/12/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose Sigma 1 receptor is a novel therapeutic target for retinal disease. Its activation, using a high-affinity, high-specificity ligand (+)-pentazocine ((+)-PTZ), rescues photoreceptor cells in the rd10 mouse model of RP. Here, we asked whether the robust retinal neuroprotective properties of (+)-PTZ are generalizable to SA4503 and PRE084, two other high-affinity sigma 1 receptor ligands. Methods We treated 661W cells with SA4503 or PRE084. Cell viability, oxidative stress, and expression of Nrf2 and NRF2-regulated antioxidant genes (Nqo1, Cat, and Sod1) were assessed. Rd10 mice were administered SA4503 (1 mg/kg), PRE084 (0.5 mg/kg), or (+)-PTZ (0.5 mg/kg). Visual acuity, retinal architecture, and retinal electrophysiologic function were measured in vivo and retinal structure was assessed histologically. Results Similar to (+)-PTZ, SA4503 and PRE084 improved cell viability, attenuated oxidative stress, and increased Nrf2, Nqo1 and Cat expression. Although treatment of rd10 mice with (+)-PTZ improved visual acuity, increased outer retinal thickness, and improved photopic a- and b-wave responses compared with nontreated rd10 mice, treatment with SA4503 or PRE084 did not. The number of photoreceptor nuclei/100 µm retinal length in SA4503- and PRE084-treated rd10 mice (approximately 11/100) did not differ significantly from nontreated rd10 mice, whereas (+)-PTZ-treated mice had significantly more nuclei (approximately 22/100 µm). Conclusions Cell survival and gene regulation experiments yielded similar outcomes when SA4503, PRE084, or (+)-PTZ were conducted in vitro, however neither SA4503 or PRE084 afforded in vivo protection in the severe rd10 retinopathy model comparable to (+)-PTZ. Despite all three compounds demonstrating the potential to activate sigma 1 receptor, the retinal neuroprotective properties of the three ligands differ significantly.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Shannon R. Barwick
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
16
|
Mechanistic Connections between Endoplasmic Reticulum (ER) Redox Control and Mitochondrial Metabolism. Cells 2019; 8:cells8091071. [PMID: 31547228 PMCID: PMC6769559 DOI: 10.3390/cells8091071] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 12/21/2022] Open
Abstract
The past decade has seen the emergence of endoplasmic reticulum (ER) chaperones as key determinants of contact formation between mitochondria and the ER on the mitochondria-associated membrane (MAM). Despite the known roles of ER–mitochondria tethering factors like PACS-2 and mitofusin-2, it is not yet entirely clear how they mechanistically interact with the ER environment to determine mitochondrial metabolism. In this article, we review the mechanisms used to communicate ER redox and folding conditions to the mitochondria, presumably with the goal of controlling mitochondrial metabolism at the Krebs cycle and at the electron transport chain, leading to oxidative phosphorylation (OXPHOS). To achieve this goal, redox nanodomains in the ER and the interorganellar cleft influence the activities of ER chaperones and Ca2+-handling proteins to signal to mitochondria. This mechanism, based on ER chaperones like calnexin and ER oxidoreductases like Ero1α, controls reactive oxygen production within the ER, which can chemically modify the proteins controlling ER–mitochondria tethering, or mitochondrial membrane dynamics. It can also lead to the expression of apoptotic or metabolic transcription factors. The link between mitochondrial metabolism and ER homeostasis is evident from the specific functions of mitochondria–ER contact site (MERC)-localized Ire1 and PERK. These functions allow these two transmembrane proteins to act as mitochondria-preserving guardians, a function that is apparently unrelated to their functions in the unfolded protein response (UPR). In scenarios where ER stress cannot be resolved via the activation of mitochondrial OXPHOS, MAM-localized autophagosome formation acts to remove defective portions of the ER. ER chaperones such as calnexin are again critical regulators of this MERC readout.
Collapse
|
17
|
Ryskamp DA, Korban S, Zhemkov V, Kraskovskaya N, Bezprozvanny I. Neuronal Sigma-1 Receptors: Signaling Functions and Protective Roles in Neurodegenerative Diseases. Front Neurosci 2019; 13:862. [PMID: 31551669 PMCID: PMC6736580 DOI: 10.3389/fnins.2019.00862] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Sigma-1 receptor (S1R) is a multi-functional, ligand-operated protein situated in endoplasmic reticulum (ER) membranes and changes in its function and/or expression have been associated with various neurological disorders including amyotrophic lateral sclerosis/frontotemporal dementia, Alzheimer's (AD) and Huntington's diseases (HD). S1R agonists are broadly neuroprotective and this is achieved through a diversity of S1R-mediated signaling functions that are generally pro-survival and anti-apoptotic; yet, relatively little is known regarding the exact mechanisms of receptor functioning at the molecular level. This review summarizes therapeutically relevant mechanisms by which S1R modulates neurophysiology and implements neuroprotective functions in neurodegenerative diseases. These mechanisms are diverse due to the fact that S1R can bind to and modulate a large range of client proteins, including many ion channels in both ER and plasma membranes. We summarize the effect of S1R on its interaction partners and consider some of the cell type- and disease-specific aspects of these actions. Besides direct protein interactions in the endoplasmic reticulum, S1R is likely to function at the cellular/interorganellar level by altering the activity of several plasmalemmal ion channels through control of trafficking, which may help to reduce excitotoxicity. Moreover, S1R is situated in lipid rafts where it binds cholesterol and regulates lipid and protein trafficking and calcium flux at the mitochondrial-associated membrane (MAM) domain. This may have important implications for MAM stability and function in neurodegenerative diseases as well as cellular bioenergetics. We also summarize the structural and biochemical features of S1R proposed to underlie its activity. In conclusion, S1R is incredibly versatile in its ability to foster neuronal homeostasis in the context of several neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniel A. Ryskamp
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Svetlana Korban
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - Vladimir Zhemkov
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Nina Kraskovskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| |
Collapse
|
18
|
Wang J, Zhao J, Cui X, Mysona BA, Navneet S, Saul A, Ahuja M, Lambert N, Gazaryan IG, Thomas B, Bollinger KE, Smith SB. The molecular chaperone sigma 1 receptor mediates rescue of retinal cone photoreceptor cells via modulation of NRF2. Free Radic Biol Med 2019; 134:604-616. [PMID: 30743048 PMCID: PMC6619428 DOI: 10.1016/j.freeradbiomed.2019.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/01/2019] [Accepted: 02/01/2019] [Indexed: 01/11/2023]
Abstract
Sigma 1 receptor (Sig1R), a putative molecular chaperone, has emerged as a novel therapeutic target for retinal degenerative disease. Earlier studies showed that activation of Sig1R via the high-affinity ligand (+)-pentazocine ((+)-PTZ) induced profound rescue of cone photoreceptor cells in the rd10 mouse model of retinitis pigmentosa; however the mechanism of rescue is unknown. Improved cone function in (+)-PTZ-treated mice was accompanied by reduced oxidative stress and normalization of levels of NRF2, a transcription factor that activates antioxidant response elements (AREs) of hundreds of cytoprotective genes. Here, we tested the hypothesis that modulation of NRF2 is central to Sig1R-mediated cone rescue. Activation of Sig1R in 661W cone cells using (+)-PTZ induced dose-dependent increases in NRF2-ARE binding activity and NRF2 gene/protein expression, whereas silencing Sig1R significantly decreased NRF2 protein levels and increased oxidative stress, although (+)-PTZ did not disrupt NRF2-KEAP1 binding. In vivo studies were conducted to investigate whether, in the absence of NRF2, activation of Sig1R rescues cones. (+)-PTZ was administered systemically for several weeks to rd10/nrf2+/+ and rd10/nrf2-/- mice. Through post-natal day 42, cone function was significant in rd10/nrf2+/+, but minimal in rd10/nrf2-/- mice as indicated by electroretinographic recordings using natural noise stimuli, optical coherence tomography and retinal histological analyses. Immunodetection of cones was limited in (+)-PTZ-treated rd10/nrf2-/-, though considerable in (+)-PTZ-treated rd10/nrf2+/+mice. The data suggest that Sig1R-mediated cone rescue requires NRF2 and provide evidence for a previously-unrecognized relationship between these proteins.
Collapse
Affiliation(s)
- J Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - J Zhao
- 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
| | - X Cui
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - B A Mysona
- 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
| | - S Navneet
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - A Saul
- 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
| | - M Ahuja
- Department of Pharmacology/Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - N Lambert
- Department of Pharmacology/Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - I G Gazaryan
- Department of Anatomy and Cell Biology, New York Medical College, Valhalla, NY, USA
| | - B Thomas
- Department of Pharmacology/Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - K E Bollinger
- 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
| | - S B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; 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
|
Rosen DA, Seki SM, Fernández-Castañeda A, Beiter RM, Eccles JD, Woodfolk JA, Gaultier A. Modulation of the sigma-1 receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Sci Transl Med 2019; 11:eaau5266. [PMID: 30728287 PMCID: PMC6936250 DOI: 10.1126/scitranslmed.aau5266] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/15/2018] [Accepted: 01/15/2019] [Indexed: 12/14/2022]
Abstract
Sepsis is an often deadly complication of infection in which systemic inflammation damages the vasculature, leading to tissue hypoperfusion and multiple organ failure. Currently, the standard of care for sepsis is predominantly supportive, with few therapeutic options available. Because of increased sepsis incidence worldwide, there is an urgent need for discovery of novel therapeutic targets and development of new treatments. The recently discovered function of the endoplasmic reticulum (ER) in regulation of inflammation offers a potential avenue for sepsis control. Here, we identify the ER-resident protein sigma-1 receptor (S1R) as an essential inhibitor of cytokine production in a preclinical model of septic shock. Mice lacking S1R succumb quickly to hypercytokinemia induced by a sublethal challenge in two models of acute inflammation. Mechanistically, we find that S1R restricts the endonuclease activity of the ER stress sensor IRE1 and cytokine expression but does not inhibit the classical inflammatory signaling pathways. These findings could have substantial clinical implications, as we further find that fluvoxamine, an antidepressant therapeutic with high affinity for S1R, protects mice from lethal septic shock and dampens the inflammatory response in human blood leukocytes. Our data reveal the contribution of S1R to the restraint of the inflammatory response and place S1R as a possible therapeutic target to treat bacterial-derived inflammatory pathology.
Collapse
Affiliation(s)
- Dorian A Rosen
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Graduate Program in Pharmacological Sciences, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Scott M Seki
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Anthony Fernández-Castañeda
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Rebecca M Beiter
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jacob D Eccles
- Division of Asthma, Allergy and Immunology, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Judith A Woodfolk
- Division of Asthma, Allergy and Immunology, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
20
|
Wang J, Smith SB. A Novel Mechanism of Sigma 1 Receptor Neuroprotection: Modulation of miR-214-3p. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1185:463-467. [PMID: 31884655 DOI: 10.1007/978-3-030-27378-1_76] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Retinitis pigmentosa (RP) is a blinding disease for which there is no known cure. In a recent study, we reported dramatic rescue of cones in the rd10 mouse model of RP when mice were treated systemically with (+)-pentazocine ((+)-PTZ), a high-affinity ligand for sigma 1 receptor (Sig1R). The molecular mechanisms by which Sig1R provides neuroprotection are unclear. In this report, we used a miRNA PCR array to compare 84 abundantly expressed, well-characterized miRNAs in rd10/Sig1R-/- vs. rd10 and rd10 + PTZ vs. rd10 mice. We found that 13 miRNAs were significantly increased in rd10/Sig1R-/- retinas but were significantly decreased in rd10 + PTZ retinas. The miRNAs were miR-9-5p, miR-27a-3p, miR-126a-5p, miR-146a-5p, miR-10a-5p, miR-34c-5p, miR-503-5p, miR-30c-5p, miR-199-5p, miR-541-5p, miR-214-3p, miR-218-5p, and miR-335-5p. Of these, miR-214-3p is closely related to oxidative stress modulation, which is relevant to degenerative retinopathy. MiR-214-3p expression is ~fivefold higher in rd10/Sig1R-/- vs. rd10. In contrast, miR-214-3p is decreased ~twofold in rd10 + PTZ vs. rd10. Interestingly, miR-214-3p is predicted to bind to Sig1R and Nrf2, a key transcription factor for modulation of oxidative stress. To our knowledge, this is the first evidence that Sig1R may interact with miRNAs in retina. This observation is the underpinning of our hypothesis that a novel mechanism by which Sig1R mediates cone rescue is via interaction with miR-214-3p.
Collapse
Affiliation(s)
- Jing Wang
- 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
|
21
|
Smith SB, Wang J, Cui X, Mysona BA, Zhao J, Bollinger KE. Sigma 1 receptor: A novel therapeutic target in retinal disease. Prog Retin Eye Res 2018; 67:130-149. [PMID: 30075336 PMCID: PMC6557374 DOI: 10.1016/j.preteyeres.2018.07.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023]
Abstract
Retinal degenerative diseases are major causes of untreatable blindness worldwide and efficacious treatments for these diseases are sorely needed. A novel target for treatment of retinal disease is the transmembrane protein Sigma 1 Receptor (Sig1R). This enigmatic protein is an evolutionary isolate with no known homology to any other protein. Sig1R was originally thought to be an opioid receptor. That notion has been dispelled and more recent pharmacological and molecular studies suggest that it is a pluripotent modulator with a number of biological functions, many of which are relevant to retinal disease. This review provides an overview of the discovery of Sig1R and early pharmacologic studies that led to the cloning of the Sig1R gene and eventual elucidation of its crystal structure. Studies of Sig1R in the eye were not reported until the late 1990s, but since that time there has been increasing interest in the potential role of Sig1R as a target for retinal disease. Studies have focused on elucidating the mechanism(s) of Sig1R function in retina including calcium regulation, modulation of oxidative stress, ion channel regulation and molecular chaperone activity. Mechanistic studies have been performed in isolated retinal cells, such as Müller glial cells, microglial cells, optic nerve head astrocytes and retinal ganglion cells as well as in the intact retina. Several compelling studies have provided evidence of powerful in vivo neuroprotective effects against ganglion cell loss as well as photoreceptor cell loss. Also described are studies that have examined retinal structure/function in various models of retinal disease in which Sig1R is absent and reveal that these phenotypes are accelerated compared to retinas of animals that express Sig1R. The collective evidence from analysis of studies over the past 20 years is that Sig1R plays a key role in modulating retinal cellular stress and that it holds great promise as a target in retinal neurodegenerative disease.
Collapse
Affiliation(s)
- 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 30912, Augusta, GA, USA.
| | - Jing Wang
- 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
| | - Xuezhi Cui
- 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
| | - Barbara A Mysona
- 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
| | - Jing Zhao
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Ophthalmology, Medical College of Georgia at Augusta University 30912, Augusta, GA, USA
| | - Kathryn E Bollinger
- 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 30912, Augusta, GA, USA
| |
Collapse
|
22
|
Mysona BA, Zhao J, Smith S, Bollinger KE. Relationship between Sigma-1 receptor and BDNF in the visual system. Exp Eye Res 2018; 167:25-30. [PMID: 29031856 PMCID: PMC5757370 DOI: 10.1016/j.exer.2017.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 12/27/2022]
Abstract
Glaucoma is an incurable optic neuropathy characterized by dysfunction and death of retinal ganglion cells (RGCs). Brain derived neurotrophic factor (BDNF) is an essential neurotrophin that supports RGC function and survival. Despite BDNF's importance, our knowledge of molecular mechanisms that modulate BDNF processing and secretion is incomplete. Sigma-1 receptor (S1R) is associated with increased BDNF in hippocampus and with BDNF secretion by brain-derived astrocytes and neuronal cell lines. Much less is known about the relationship between S1R and BDNF in the visual system. Here, we examine how S1R activation and deletion alter expression of mature BDNF (mBDNF) and proBDNF in retina and cultured optic nerve head (ONH) astrocytes. For S1R activation, the S1R agonist (+)-pentazocine (PTZ, 0.5 mg/kg) was administered by intraperitoneal injection to C57BL/6J mice, 3 times per week, for 5 weeks. Expression of proBDNF and mBDNF was also examined in S1R knockout and age-matched C57BL/6J mice. In vitro, cultured ONH astrocytes were treated with 3 μM PTZ for 24 h followed by collection of media and ONH astrocyte lysates. Results showed that treatment with (+)-PTZ increased mBDNF protein in both retina and hippocampus. In contrast, S1R deletion was associated with retinal mBDNF deficits. In ONH astrocytes S1R agonist (+)-PTZ significantly increased levels of secreted BDNF and proBDNF in cell lysates. These findings support a role for S1R in the modulation of BDNF levels within the retina and optic nerve head. Treatment with S1R agonists might provide benefit in diseases such as glaucoma by increasing BDNF levels from endogenous sources.
Collapse
Affiliation(s)
- Barbara A Mysona
- James and Jean Culver Vision Discovery Institute, Augusta, GA, 30912, United States; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Jing Zhao
- James and Jean Culver Vision Discovery Institute, Augusta, GA, 30912, United States; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Sylvia Smith
- James and Jean Culver Vision Discovery Institute, Augusta, GA, 30912, United States; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Kathryn E Bollinger
- James and Jean Culver Vision Discovery Institute, Augusta, GA, 30912, United States; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States.
| |
Collapse
|
23
|
Mohamed R, Sharma I, Ibrahim AS, Saleh H, Elsherbiny NM, Fulzele S, Elmasry K, Smith SB, Al-Shabrawey M, Tawfik A. Hyperhomocysteinemia Alters Retinal Endothelial Cells Barrier Function and Angiogenic Potential via Activation of Oxidative Stress. Sci Rep 2017; 7:11952. [PMID: 28931831 PMCID: PMC5607263 DOI: 10.1038/s41598-017-09731-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 07/28/2017] [Indexed: 01/16/2023] Open
Abstract
Hyperhomocysteinemia (HHcy) is associated with several human visual disorders, such as diabetic retinopathy (DR) and age-related macular degeneration (AMD). Breakdown of the blood-retinal barrier (BRB) is linked to vision loss in DR and AMD. Our previous work revealed that HHcy altered BRB in retinal endothelial cells in vivo. Here we hypothesize that homocysteine (Hcy) alters retinal endothelial cell barrier function and angiogenic potential via activation of oxidative stress. Human retinal endothelial cells (HRECs) treated with and without different concentrations of Hcy showed a reduction of tight junction protein expression, increased FITC dextran leakage, decreased transcellular electrical resistance and increased angiogenic potential. In addition, HRECs treated with Hcy showed increased production of reactive oxygen species (ROS). The anti-oxidant N-acetyl-cysteine (NAC) reduced ROS formation and decreased FITC-dextran leakage in Hcy treated HRECs. A mouse model of HHcy, in which cystathionine-β-synthase is deficient (cbs−/−), was evaluated for oxidative stress by dichlolorofluorescein (DCF), dihydroethidium (DHE) staining. There was a marked increase in ROS production and augmented GSH reductase and antioxidant regulator NRF2 activity, but decreased antioxidant gene expression in retinas of hyperhomocysteinemic mice. Our results suggest activation of oxidative stress as a possible mechanism of HHcy induced retinal endothelial cell dysfunction.
Collapse
Affiliation(s)
- Riyaz Mohamed
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA
| | - Isha Sharma
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA
| | - Ahmed S Ibrahim
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA.,Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Heba Saleh
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA
| | - Nehal M Elsherbiny
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA.,Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Sadanand Fulzele
- Department: Orthopedic Surgery, MCG, Augusta University, Augusta, GA, USA
| | - Khaled Elmasry
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA.,Department of Cellular Biology and Anatomy, MCG, Augusta University, Augusta, GA, USA
| | - Sylvia B Smith
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA.,Department of Cellular Biology and Anatomy, MCG, Augusta University, Augusta, GA, USA.,Department of Ophthalmology, MCG, Augusta University, Augusta, GA, USA
| | - Mohamed Al-Shabrawey
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA.,Department of Cellular Biology and Anatomy, MCG, Augusta University, Augusta, GA, USA.,Department of Ophthalmology, MCG, Augusta University, Augusta, GA, USA
| | - Amany Tawfik
- Department of Oral Biology and Anatomy, Dental College of Georgia, Augusta University, Augusta, GA, USA. .,James and Jean Culver Vision Discovery Institute, Medical College of Georgia (MCG), Augusta University, Augusta, USA. .,Department of Cellular Biology and Anatomy, MCG, Augusta University, Augusta, GA, USA. .,Department of Ophthalmology, MCG, Augusta University, Augusta, GA, USA.
| |
Collapse
|
24
|
Yang H, Fu Y, Liu X, Shahi PK, Mavlyutov TA, Li J, Yao A, Guo SZW, Pattnaik BR, Guo LW. Role of the sigma-1 receptor chaperone in rod and cone photoreceptor degenerations in a mouse model of retinitis pigmentosa. Mol Neurodegener 2017; 12:68. [PMID: 28927431 PMCID: PMC5606113 DOI: 10.1186/s13024-017-0202-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/09/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Retinitis pigmentosa (RP) is the most common inherited retinal degenerative disease yet with no effective treatment available. The sigma-1 receptor (S1R), a ligand-regulated chaperone, emerges as a potential retina-protective therapeutic target. In particular, pharmacological activation of S1R was recently shown to rescue cones in the rd10 mouse, a rod Pde6b mutant that recapitulates the RP pathology of autonomous rod degeneration followed by secondary death of cones. The mechanisms underlying the S1R protection for cones are not understood in detail. METHODS By rearing rd10/S1R-/- and rd10/S1R+/+ mice in dim light to decelerate rapid rod/cone degeneration, we were able to compare their retinal biochemistry, histology and functions throughout postnatal 3-6 weeks (3 W-6 W). RESULTS The receptor-interacting protein kinases (RIP1/RIP3) and their interaction (proximity ligation) dramatically up-regulated after 5 W in rd10/S1R-/- (versus rd10/S1R+/+) retinas, indicative of intensified necroptosis activation, which was accompanied by exacerbated loss of cones. Greater rod loss in rd10/S1R-/- versus rd10/S1R+/+ retinas was evidenced by more cleaved Caspase3 (4 W) and lower rod electro-retinographic a-waves (4 W-6 W), concomitant with reduced LC3-II and CHOP (4 W-6 W), markers of autophagy and endoplasmic reticulum stress response, respectively. However, the opposite occurred at 3 W. CONCLUSION This study reveals previously uncharacterized S1R-associated mechanisms during rd10 photoreceptor degeneration, including S1R's influences on necroptosis and autophagy as well as its biphasic role in rod degeneration upstream of cone death.
Collapse
Affiliation(s)
- Huan Yang
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Yingmei Fu
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Nan Road, Shanghai, 200030 People’s Republic of China
| | - Xinying Liu
- Department of Pediatrics, Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, SMI 112, Madison, WI 53706 USA
| | - Pawan K. Shahi
- Department of Pediatrics, Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, SMI 112, Madison, WI 53706 USA
| | - Timur A. Mavlyutov
- Department of Anesthesiology, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Jun Li
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- Department of Ophthalmology, the First Hospital of China Medical University, Shenyang, 110001 People’s Republic of China
- Department of Ophthalmology, the 3rd People’s Hospital of Dalian, Dalian, 116033 People’s Republic of China
| | - Annie Yao
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Steven Z.-W. Guo
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Bikash R. Pattnaik
- Department of Pediatrics, Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, SMI 112, Madison, WI 53706 USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI 53705 USA
| | - Lian-Wang Guo
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI 53705 USA
- Department of Surgery and Department of Physiology &Cell Biology, the Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
25
|
Zhao J, Mysona BA, Wang J, Gonsalvez GB, Smith SB, Bollinger KE. Sigma 1 receptor regulates ERK activation and promotes survival of optic nerve head astrocytes. PLoS One 2017; 12:e0184421. [PMID: 28898265 PMCID: PMC5595338 DOI: 10.1371/journal.pone.0184421] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 08/23/2017] [Indexed: 11/18/2022] Open
Abstract
The sigma 1 receptor (S1R) is a unique transmembrane protein that has been shown to regulate neuronal differentiation and cellular survival. It is expressed within several cell types throughout the nervous system and visceral organs, including neurons and glia within the eye. S1R ligands are therapeutic targets for diseases ranging from neurodegenerative conditions to neoplastic disorders. However, effects of S1R activation and inhibition within glia cells are not well characterized. Within the eye, the astrocytes at the optic nerve head are crucial to the health and survival of the neurons that send visual information to the brain. In this study, we used the S1R-specific agonist, (+)-pentazocine, to evaluate S1R activation within optic nerve head-derived astrocytes (ONHAs). Treatment of ONHAs with (+)-pentazocine attenuated the level and duration of stress-induced ERK phosphorylation following oxidative stress exposure and promoted survival of ONHAs. These effects were specific to S1R activation because they were not observed in ONHAs that were depleted of S1R using siRNA-mediated knockdown. Collectively, our results suggest that S1R activation suppresses ERK1/2 phosphorylation and protects ONHAs from oxidative stress-induced death.
Collapse
Affiliation(s)
- Jing Zhao
- James and Jean Culver Vision Discovery Institute, Augusta, Georgia, United States of America
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Barbara A. Mysona
- James and Jean Culver Vision Discovery Institute, Augusta, Georgia, United States of America
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Jing Wang
- James and Jean Culver Vision Discovery Institute, Augusta, Georgia, United States of America
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Graydon B. Gonsalvez
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Sylvia B. Smith
- James and Jean Culver Vision Discovery Institute, Augusta, Georgia, United States of America
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Kathryn E. Bollinger
- James and Jean Culver Vision Discovery Institute, Augusta, Georgia, United States of America
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| |
Collapse
|
26
|
Wang J, Saul A, Cui X, Roon P, Smith SB. Absence of Sigma 1 Receptor Accelerates Photoreceptor Cell Death in a Murine Model of Retinitis Pigmentosa. Invest Ophthalmol Vis Sci 2017; 58:4545-4558. [PMID: 28877319 PMCID: PMC5586962 DOI: 10.1167/iovs.17-21947] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023] Open
Abstract
Purpose Sigma 1 Receptor (Sig1R) is a novel therapeutic target in neurodegenerative diseases, including retinal disease. Sig1R-/- mice have late-onset retinal degeneration with ganglion cell loss that worsens under stress. Whether Sig1R plays a role in maintaining other retinal neurons is unknown, but was investigated here using rd10 mice, a model of severe photoreceptor degeneration. Methods Wild-type, rd10, and rd10/Sig1R-/- mice were subjected to ERG and spectral-domain optical coherence tomography (SD-OCT) to assess visual function/structure in situ. Retinas imaged microscopically were subjected to morphometric analysis, immunodetection of cones, and analysis of gliosis. Oxidative and endoplasmic reticulum (ER) stress was evaluated at mRNA/protein levels. Results Photopic ERG responses were reduced significantly in rd10/Sig1R-/- versus rd10 mice at P28 (31 ± 6 vs. 56 ± 7 μV), indicating accelerated cone loss when Sig1R was absent. At P28, SD-OCT revealed reduced retinal thickness in rd10/Sig1R-/- mice (60% of WT) versus rd10 (80% of WT). Morphometric analysis disclosed profound photoreceptor nuclei loss in rd10/Sig1R-/- versus rd10 mice. rd10/Sig1R-/- mice had 35% and 60% fewer photoreceptors, respectively, at P28 and P35, than rd10. Peanut agglutinin cone labeling decreased significantly; gliosis increased significantly in rd10/Sig1R-/- versus rd10 mice. At P21, NRF2 levels increased in rd10/Sig1R-/- mice versus rd10 and downstream antioxidants increased indicating oxidative stress. At P28, ER stress genes/proteins, especially XBP1, a potent transcriptional activator of the unfolded protein response and CHOP, a proapoptotic transcription factor, increased significantly in rd10/Sig1R-/- mice versus rd10. Conclusions Photoreceptor cell degeneration accelerates and cone function diminishes much earlier in rd10/Sig1R-/- than rd10 mice emphasizing the importance of Sig1R as a modulator of retinal cell survival.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Alan Saul
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| | - Xuezhi Cui
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Penny Roon
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
27
|
Potential independent action of sigma receptor ligands through inhibition of the Kv2.1 channel. Oncotarget 2017; 8:59345-59358. [PMID: 28938641 PMCID: PMC5601737 DOI: 10.18632/oncotarget.19581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/16/2017] [Indexed: 12/14/2022] Open
Abstract
The sigma-1 receptor (σ1-R) and sigma-2 receptor (σ2-R) are potential drug targets for treatment of cancer, pain, depression, retinal degeneration and other neuronal diseases. Previous reports show that sigma-1 receptor modulates the activities of multiple channels. We are interested in possible sigma receptor modulation of Kv2.1, a K+ channel abundant in retinal photoreceptors. We tested the effect of established sigma receptor ligands on Kv2.1 channels which were stably expressed in HEK293 cells. Surprisingly, σ1-R antagonists inhibited Kv2.1 currents in both wild type and σ1-R knockout HEK293 cells that we engineered using the CRISPR/Cas9 technology. Moreover, PB28, a σ1-R antagonist and also σ2-R agonist, inhibited Kv2.1 in σ1-R knockout cells, but this action was not blocked by the σ2-R antagonists that did not have an effect on Kv2.1. We also observed inhibition of electroretinogram by PB28 in wild type as well as σ1-R knockout mice. Thus, the results in this study indicate that the Kv2.1-inhibiting function of the sigma ligands is not sigma receptor dependent, suggesting a direct effect of these ligands on the Kv2.1 channel.
Collapse
|
28
|
Cancer nanotheranostics: A review of the role of conjugated ligands for overexpressed receptors. Eur J Pharm Sci 2017; 104:273-292. [DOI: 10.1016/j.ejps.2017.04.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/13/2022]
|
29
|
Zhao L, Chen G, Li J, Fu Y, Mavlyutov TA, Yao A, Nickells RW, Gong S, Guo LW. An intraocular drug delivery system using targeted nanocarriers attenuates retinal ganglion cell degeneration. J Control Release 2017; 247:153-166. [PMID: 28063892 PMCID: PMC5323250 DOI: 10.1016/j.jconrel.2016.12.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/08/2016] [Accepted: 12/29/2016] [Indexed: 02/03/2023]
Abstract
Glaucoma is a common blinding disease characterized by loss of retinal ganglion cells (RGCs). To date, there is no clinically available treatment directly targeting RGCs. We aim to develop an RGC-targeted intraocular drug delivery system using unimolecular micelle nanoparticles (unimNPs) to prevent RGC loss. The unimNPs were formed by single/individual multi-arm star amphiphilic block copolymer poly(amidoamine)-polyvalerolactone-poly(ethylene glycol) (PAMAM-PVL-PEG). While the hydrophobic PAMAM-PVL core can encapsulate hydrophobic drugs, the hydrophilic PEG shell provides excellent water dispersity. We conjugated unimNPs with the cholera toxin B domain (CTB) for RGC-targeting and with Cy5.5 for unimNP-tracing. To exploit RGC-protective sigma-1 receptor (S1R), we loaded unimNPs with an endogenous S1R agonist dehydroepiandrosterone (DHEA) as an FDA-approved model drug. These unimNPs produced a steady DHEA release in vitro for over two months at pH7.4. We then co-injected (mice, intraocular) unimNPs with the glutamate analog N-methyl-d-aspartate (NMDA), which is excito-toxic and induces RGC death. The CTB-conjugated unimNPs (i.e., targeted NPs) accumulated at the RGC layer and effectively preserved RGCs at least for 14days, whereas the unimNPs without CTB (i.e., non-targeted NPs) showed neither accumulation at nor protection of NMDA-treated RGCs. Consistent with S1R functions, targeted NPs relative to non-targeted NPs showed markedly better inhibitory effects on apoptosis and oxidative/inflammatory stresses in the RGC layer. Hence, the DHEA-loaded, CTB-conjugated unimNPs represent an RGC/S1R dual-targeted nanoplatform that generates an efficacious template for further development of a sustainable intraocular drug delivery system to protect RGCs, which may be applicable to treatments directed at glaucomatous pathology.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Guojun Chen
- Department of Materials Science and Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jun Li
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; Department of Ophthalmology, The First Hospital of China Medical University, Shenyang 110001, China.; Department of Ophthalmology, The 3rd People's Hospital of Dalian, Dalian 116033, China
| | - Yingmei Fu
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Nan Road, Shanghai 200030, China
| | - Timur A Mavlyutov
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Annie Yao
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Robert W Nickells
- Department of Ophthalmology and Vision Sciences, University of Wisconsin-Madison, 1300 University Ave, Madison, WI 53706, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Shaoqin Gong
- Department of Materials Science and Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Lian-Wang Guo
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
30
|
Mysona B, Kansara N, Zhao J, Bollinger K. The Role of Sigma 1 Receptor as a Neuroprotective Target in Glaucoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:299-307. [PMID: 28315279 DOI: 10.1007/978-3-319-50174-1_20] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The role of sigma 1 receptor (S1R) in glaucoma is emerging as a promising field of study. Glaucoma is an optic neuropathy that shares common pathogenic mechanisms with other neurodegenerative diseases such as Alzheimer's and Parkinson's disease . S1R modulates multiple cellular functions associated with neurodegeneration . These include Ca2+ ion homeostasis, endoplasmic reticulum (ER) and oxidative stress , survival signaling pathways, neurotrophin secretion, and glial activation. S1R may also have neurorestorative properties including enhancement of neuronal plasticity and neurite outgrowth. Recent studies using agonists for S1R within the eye provide hope that it could be a therapeutic target for glaucoma. Understanding the role of S1R in glaucoma may help us to stop the progression of this sight threatening disease.
Collapse
Affiliation(s)
- Barbara Mysona
- Departments of Cellular Biology and Anatomy and Ophthalmology and the James and Jean Culver Vision Discovery Institute, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Neil Kansara
- Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
- The James and Jean Culver Vision Discovery Institute, Augusta, GA, 30912, USA
| | - Jing Zhao
- The James and Jean Culver Vision Discovery Institute, Augusta, GA, 30912, USA
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Kathryn Bollinger
- Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
- The James and Jean Culver Vision Discovery Institute, Augusta, GA, 30912, USA.
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
31
|
Wang J, Cui X, Roon P, Saul A, Smith SB. The Role of Sigma1R in Mammalian Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:267-284. [PMID: 28315277 DOI: 10.1007/978-3-319-50174-1_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This review article focuses on studies of Sigma 1 Receptor (Sigma1R) and retina . It provides a brief overview of the earliest pharmacological studies performed in the late 1990s that provided evidence of the presence of Sigma1R in various ocular tissues. It then describes work from a number of labs concerning the location of Sigma1R in several retinal cell types including ganglion, Müller glia , and photoreceptors . The role of Sigma1R ligands in retinal neuroprotection is emphasized. Early studies performed in vitro clearly showed that targeting Sigma1R could attenuate stress-induced retinal cell loss. These studies were followed by in vivo experiments. Data about the usefulness of targeting Sigma1R to prevent ganglion cell loss associated with diabetic retinopathy are reviewed. Mechanisms of Sigma1R-mediated retinal neuroprotection involving Müller cells , especially in modulating oxidative stress are described along with information about the retinal phenotype of mice lacking Sigma1R (Sigma1R -/- mice). The retina develops normally in Sigma1R -/- mice, but after many months there is evidence of apoptosis in the optic nerve head, decreased ganglion cell function and eventual loss of these cells. Additional studies using the Sigma1R -/- mice provide strong evidence that in the retina, Sigma1R plays a key role in modulating cellular stress. Recent work has shown that targeting Sigma1R may extend beyond protection of ganglion cells to include photoreceptor cell degeneration as well.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15th Street, CB 1114, Augusta, GA, 30912-2000, USA
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Xuezhi Cui
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15th Street, CB 1114, Augusta, GA, 30912-2000, USA
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Penny Roon
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15th Street, CB 1114, Augusta, GA, 30912-2000, USA
| | - Alan Saul
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1120 15th Street, CB 1114, Augusta, GA, 30912-2000, USA
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Sylvia B Smith
- Departments of Cellular Biology and Anatomy and Ophthalmology and the James and Jean Culver Vision Discovery Institute, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
32
|
Mancuso R, Navarro X. Sigma-1 Receptor in Motoneuron Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:235-254. [PMID: 28315275 DOI: 10.1007/978-3-319-50174-1_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS ) is a neurodegenerative disease affecting spinal cord and brain motoneurons , leading to paralysis and early death. Multiple etiopathogenic mechanisms appear to contribute in the development of ALS , including glutamate excitotoxicity, oxidative stress , protein misfolding, mitochondrial defects, impaired axonal transport, inflammation and glial cell alterations. The Sigma-1 receptor is highly expressed in motoneurons of the spinal cord, particularly enriched in the endoplasmic reticulum (ER) at postsynaptic cisternae of cholinergic C-terminals. Several evidences point to participation of Sigma-1R alterations in motoneuron degeneration. Thus, mutations of the transmembrane domain of the Sigma-1R have been described in familial ALS cases. Interestingly, Sigma-1R KO mice display muscle weakness and motoneuron loss. On the other hand, Sigma-1R agonists promote neuroprotection and neurite elongation through activation of protein kinase C on motoneurons in vitro and in vivo after ventral root avulsion. Remarkably, treatment of SOD1 mice, the most usual animal model of ALS , with Sigma-1R agonists resulted in significantly enhanced motoneuron function and preservation, and increased animal survival. Sigma-1R activation also reduced microglial reactivity and increased the glial expression of neurotrophic factors. Two main interconnected mechanisms seem to underlie the effects of Sigma-1R manipulation on motoneurons: modulation of neuronal excitability and regulation of calcium homeostasis. In addition, Sigma-1R also contributes to regulating protein degradation, and reducing oxidative stress. Therefore, the multi-functional nature of the Sigma-1R represents an attractive target for treating aspects of ALS and other motoneuron diseases .
Collapse
Affiliation(s)
- Renzo Mancuso
- Center for Biological Sciences, University of Southampton, Southampton General Hospital, SO16 6YD, Southampton, UK
| | - Xavier Navarro
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
33
|
Mavlyutov TA, Guo LW. Peeking into Sigma-1 Receptor Functions Through the Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:285-297. [PMID: 28315278 PMCID: PMC6283661 DOI: 10.1007/978-3-319-50174-1_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review discusses recent advances towards understanding the sigma-1 receptor (S1R) as an endogenous neuro-protective mechanism in the retina , a favorable experimental model system. The exquisite architecture of the mammalian retina features layered and intricately wired neurons supported by non-neuronal cells. Ganglion neurons, photoreceptors , as well as the retinal pigment epithelium, are susceptible to degeneration that leads to major retinal diseases such as glaucoma , diabetic retinopathy , and age-related macular degeneration (AMD), and ultimately, blindness. The S1R protein is found essentially in every retinal cell type, with high abundance in the ganglion cell layer. Ultrastructural studies of photoreceptors, bipolar cells, and ganglion cells show a predominant localization of S1R in the nuclear envelope. A protective role of S1R for ganglion and photoreceptor cells is supported by in vitro and in vivo experiments. Most recently, studies suggest that S1R may also protect retinal neurons via its activities in Müller glia and microglia. The S1R functions in the retina may be attributed to a reduction of excitotoxicity, oxidative stress , ER stress response, or inflammation. S1R knockout mice are being used to delineate the S1R-specific effects. In summary, while significant progress has been made towards the objective of establishing a S1R-targeted paradigm for retinal neuro-protection , critical questions remain. In particular, context-dependent effects and potential side effects of interventions targeting S1R need to be studied in more diverse and more clinically relevant animal models.
Collapse
Affiliation(s)
- Timur A Mavlyutov
- Department of Surgery and McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, 53705, Madison, WI, USA
| | - Lian-Wang Guo
- Department of Surgery and McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, 53705, Madison, WI, USA
| |
Collapse
|
34
|
Ryskamp D, Wu J, Geva M, Kusko R, Grossman I, Hayden M, Bezprozvanny I. The sigma-1 receptor mediates the beneficial effects of pridopidine in a mouse model of Huntington disease. Neurobiol Dis 2017; 97:46-59. [PMID: 27818324 PMCID: PMC5214572 DOI: 10.1016/j.nbd.2016.10.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 09/13/2016] [Accepted: 10/30/2016] [Indexed: 11/25/2022] Open
Abstract
The tri-nucleotide repeat expansion underlying Huntington disease (HD) results in corticostriatal synaptic dysfunction and subsequent neurodegeneration of striatal medium spiny neurons (MSNs). HD is a devastating autosomal dominant disease with no disease-modifying treatments. Pridopidine, a postulated "dopamine stabilizer", has been shown to improve motor symptoms in clinical trials of HD. However, the target(s) and mechanism of action of pridopidine remain to be fully elucidated. As binding studies identified sigma-1 receptor (S1R) as a high-affinity receptor for pridopidine, we evaluated the relevance of S1R as a therapeutic target of pridopidine in HD. S1R is an endoplasmic reticulum - (ER) resident transmembrane protein and is regulated by ER calcium homeostasis, which is perturbed in HD. Consistent with ER calcium dysregulation, we observed striatal upregulation of S1R in aged YAC128 transgenic HD mice and HD patients. We previously demonstrated that dendritic MSN spines are lost in aged corticostriatal co-cultures from YAC128 mice. We report here that pridopidine and the chemically similar S1R agonist 3-PPP prevent MSN spine loss in aging YAC128 co-cultures. Spine protection was blocked by neuronal deletion of S1R. Pridopidine treatment suppressed supranormal ER Ca2+ release, restored ER calcium levels and reduced excessive store-operated calcium (SOC) entry in spines, which may account for its synaptoprotective effects. Normalization of ER Ca2+ levels by pridopidine was prevented by S1R deletion. To evaluate long-term effects of pridopidine, we analyzed expression profiles of calcium signaling genes. Pridopidine elevated striatal expression of calbindin and homer1a, whereas their striatal expression was reduced in aged Q175KI and YAC128 HD mouse models compared to WT. Pridopidine and 3-PPP are proposed to prevent calcium dysregulation and synaptic loss in a YAC128 corticostriatal co-culture model of HD. The actions of pridopidine were mediated by S1R and led to normalization of ER Ca2+ release, ER Ca2+ levels and spine SOC entry in YAC128 MSNs. This is a new potential mechanism of action for pridopidine, highlighting S1R as a potential target for HD therapy. Upregulation of striatal proteins that regulate calcium, including calbindin and homer1a, upon chronic therapy with pridopidine, may further contribute to long-term beneficial effects of pridopidine in HD.
Collapse
Affiliation(s)
- Daniel Ryskamp
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jun Wu
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Michal Geva
- Teva Pharmaceutical Industries, 5 Basel St., Petach Tikva 49131, Israel.
| | | | - Iris Grossman
- Teva Pharmaceutical Industries, 5 Basel St., Petach Tikva 49131, Israel.
| | - Michael Hayden
- Teva Pharmaceutical Industries, 5 Basel St., Petach Tikva 49131, Israel.
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
35
|
Ha Y, Liu H, Zhu S, Yi P, Liu W, Nathanson J, Kayed R, Loucas B, Sun J, Frishman LJ, Motamedi M, Zhang W. Critical Role of the CXCL10/C-X-C Chemokine Receptor 3 Axis in Promoting Leukocyte Recruitment and Neuronal Injury during Traumatic Optic Neuropathy Induced by Optic Nerve Crush. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:352-365. [PMID: 27960090 DOI: 10.1016/j.ajpath.2016.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/14/2016] [Accepted: 10/13/2016] [Indexed: 12/11/2022]
Abstract
Traumatic optic neuropathy (TON) is an acute injury of the optic nerve secondary to trauma. Loss of retinal ganglion cells (RGCs) is a key pathological process in TON, yet mechanisms responsible for RGC death remain unclear. In a mouse model of TON, real-time noninvasive imaging revealed a dramatic increase in leukocyte rolling and adhesion in veins near the optic nerve (ON) head at 9 hours after ON injury. Although RGC dysfunction and loss were not detected at 24 hours after injury, massive leukocyte infiltration was observed in the superficial retina. These cells were identified as T cells, microglia/monocytes, and neutrophils but not B cells. CXCL10 is a chemokine that recruits leukocytes after binding to its receptor C-X-C chemokine receptor (CXCR) 3. The levels of CXCL10 and CXCR3 were markedly elevated in TON, and up-regulation of CXCL10 was mediated by STAT1/3. Deleting CXCR3 in leukocytes significantly reduced leukocyte recruitment, and prevented RGC death at 7 days after ON injury. Treatment with CXCR3 antagonist attenuated TON-induced RGC dysfunction and cell loss. In vitro co-culture of primary RGCs with leukocytes resulted in increased RGC apoptosis, which was exaggerated in the presence of CXCL10. These results indicate that leukocyte recruitment in retinal vessels near the ON head is an early event in TON and the CXCL10/CXCR3 axis has a critical role in recruiting leukocytes and inducing RGC death.
Collapse
Affiliation(s)
- Yonju Ha
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Hua Liu
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, Texas
| | - Shuang Zhu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Panpan Yi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Wei Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Jared Nathanson
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas
| | - Bradford Loucas
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | | | - Massoud Motamedi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas; Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, Texas
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas; Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
36
|
Promsote W, Powell FL, Veean S, Thounaojam M, Markand S, Saul A, Gutsaeva D, Bartoli M, Smith SB, Ganapathy V, Martin PM. Oral Monomethyl Fumarate Therapy Ameliorates Retinopathy in a Humanized Mouse Model of Sickle Cell Disease. Antioxid Redox Signal 2016; 25:921-935. [PMID: 27393735 PMCID: PMC5144884 DOI: 10.1089/ars.2016.6638] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS Sickle retinopathy (SR) is a major cause of blindness in sickle cell disease (SCD). The genetic mutation responsible for SCD is known, however; oxidative stress and inflammation also figure prominently in the development and progression of pathology. Development of therapies for SR is hampered by the lack of (a) animal models that accurately recapitulate human SR and (b) strategies for noninvasive yet effective retinal drug delivery. This study addressed both issues by validating the Townes humanized SCD mouse as a model of SR and demonstrating the efficacy of oral administration of the antioxidant fumaric acid ester monomethyl fumarate (MMF) in the disease. RESULTS In vivo ophthalmic imaging, electroretinography, and postmortem histological RNA and protein analyses were used to monitor retinal health and function in normal (HbAA) and sickle (HbSS) hemoglobin-producing mice over a one-year period and in additional HbAA and HbSS mice treated with MMF (15 mg/ml) for 5 months. Functional and morphological abnormalities and molecular hallmarks of oxidative stress/inflammation were evident early in HbSS retinas and increased in number and severity with age. Treatment with MMF, a known inducer of Nrf2, induced γ-globin expression and fetal hemoglobin production, improved hematological profiles, and ameliorated SR-related pathology. Innovation and Conclusion: United States Food and Drug Administration-approved formulations in which MMF is the primary bioactive ingredient are currently available to treat multiple sclerosis; such drugs may be effective for treatment of ocular and systemic complications of SCD, and given the pleiotropic effects, other nonsickle-related diseases in which oxidative stress, inflammation, and retinal vascular pathology figure prominently. Antioxid. Redox Signal. 25, 921-935.
Collapse
Affiliation(s)
- Wanwisa Promsote
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Folami Lamoke Powell
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Satyam Veean
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Menaka Thounaojam
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Shanu Markand
- 3 Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Alan Saul
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Diana Gutsaeva
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Manuela Bartoli
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Sylvia B Smith
- 2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,3 Department of Cellular Biology and Anatomy, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| | - Vadivel Ganapathy
- 5 Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Pamela M Martin
- 1 Department of Biochemistry and Molecular Biology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,2 Department of Ophthalmology, The Medical College of Georgia at Augusta University , Augusta, Georgia .,4 The Culver Vision Discovery Institute, The Medical College of Georgia at Augusta University , Augusta, Georgia
| |
Collapse
|
37
|
Taylor CP, Traynelis SF, Siffert J, Pope LE, Matsumoto RR. Pharmacology of dextromethorphan: Relevance to dextromethorphan/quinidine (Nuedexta®) clinical use. Pharmacol Ther 2016; 164:170-82. [PMID: 27139517 DOI: 10.1016/j.pharmthera.2016.04.010] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dextromethorphan (DM) has been used for more than 50years as an over-the-counter antitussive. Studies have revealed a complex pharmacology of DM with mechanisms beyond blockade of N-methyl-d-aspartate (NMDA) receptors and inhibition of glutamate excitotoxicity, likely contributing to its pharmacological activity and clinical potential. DM is rapidly metabolized to dextrorphan, which has hampered the exploration of DM therapy separate from its metabolites. Coadministration of DM with a low dose of quinidine inhibits DM metabolism, yields greater bioavailability and enables more specific testing of the therapeutic properties of DM apart from its metabolites. The development of the drug combination DM hydrobromide and quinidine sulfate (DM/Q), with subsequent approval by the US Food and Drug Administration for pseudobulbar affect, led to renewed interest in understanding DM pharmacology. This review summarizes the interactions of DM with brain receptors and transporters and also considers its metabolic and pharmacokinetic properties. To assess the potential clinical relevance of these interactions, we provide an analysis comparing DM activity from in vitro functional assays with the estimated free drug DM concentrations in the brain following oral DM/Q administration. The findings suggest that DM/Q likely inhibits serotonin and norepinephrine reuptake and also blocks NMDA receptors with rapid kinetics. Use of DM/Q may also antagonize nicotinic acetylcholine receptors, particularly those composed of α3β4 subunits, and cause agonist activity at sigma-1 receptors.
Collapse
Affiliation(s)
| | - Stephen F Traynelis
- Dept. of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Joao Siffert
- Avanir Pharmaceuticals, Inc., Aliso Viejo, CA, USA
| | - Laura E Pope
- Avanir Pharmaceuticals, Inc., Aliso Viejo, CA, USA
| | - Rae R Matsumoto
- College of Pharmacy, Touro University California, Vallejo, CA, USA
| |
Collapse
|
38
|
Wang J, Saul A, Roon P, Smith SB. Activation of the molecular chaperone, sigma 1 receptor, preserves cone function in a murine model of inherited retinal degeneration. Proc Natl Acad Sci U S A 2016; 113:E3764-72. [PMID: 27298364 PMCID: PMC4932934 DOI: 10.1073/pnas.1521749113] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Retinal degenerative diseases are major causes of untreatable blindness, and novel approaches to treatment are being sought actively. Here we explored the activation of a unique protein, sigma 1 receptor (Sig1R), in the treatment of PRC loss because of its multifaceted role in cellular survival. We used Pde6β(rd10) (rd10) mice, which harbor a mutation in the rod-specific phosphodiesterase gene Pde6β and lose rod and cone photoreceptor cells (PRC) within the first 6 wk of life, as a model for severe retinal degeneration. Systemic administration of the high-affinity Sig1R ligand (+)-pentazocine [(+)-PTZ] to rd10 mice over several weeks led to the rescue of cone function as indicated by electroretinographic recordings using natural noise stimuli and preservation of cone cells upon spectral domain optical coherence tomography and retinal histological examination. The protective effect appears to result from the activation of Sig1R, because rd10/Sig1R(-/-) mice administered (+)-PTZ exhibited no cone preservation. (+)-PTZ treatment was associated with several beneficial cellular phenomena including attenuated reactive gliosis, reduced microglial activation, and decreased oxidative stress in mutant retinas. To our knowledge, this is the first report that activation of Sig1R attenuates inherited PRC loss. The findings may have far-reaching therapeutic implications for retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology/Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912
| | - Alan Saul
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Penny Roon
- Department of Cellular Biology/Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Sylvia B Smith
- Department of Cellular Biology/Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912; James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912; Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| |
Collapse
|
39
|
Wang J, Cui X, Roon P, Smith SB. Role of Sigma 1 Receptor in Retinal Degeneration of the Ins2Akita/+ Murine Model of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2016; 57:2770-81. [PMID: 27206247 PMCID: PMC4884059 DOI: 10.1167/iovs.15-18995] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/17/2016] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Sigma receptor 1 (Sigma1R), a nonopioid putative molecular chaperone, has neuroprotective properties in retina. This study sought to determine whether delaying administration of (+)-pentazocine, a high-affinity Sigma1R ligand after onset of diabetes in Ins2Akita/+ diabetic mice would afford retinal neuroprotection and to determine consequences on retinal phenotype in Ins2Akita/+ diabetic mice in the absence of Sigma1R. METHODS Ins2Akita/+ diabetic and WT mice received intraperitoneal injections of (+)-pentazocine beginning 4 or 8 weeks after onset of diabetes; eyes were harvested at 25 weeks. Retinal histologic sections were analyzed to determine thicknesses of retinal layers, number of ganglion cells, and evidence of gliosis (increased glial fibrillary acidic protein levels). Ins2Akita/+/Sig1R-/-mice were generated and subjected to in vivo assessment of retinal architecture (optical coherence tomography [OCT]) and retinal vasculature using fluorescein angiography (FA) at 12 and 16 weeks compared with age-matched Ins2Akita/+ mice. Eyes were then harvested for retinal morphometric assessment and gliosis assessment. RESULTS Wild-type mice had 13 ± 0.06 cells/100 μm retinal length; cell bodies in Ins2Akita/+ mice injected 4 and 8 weeks after onset of diabetes with (+)-pentazocine retained significantly more ganglion cells compared with Ins2Akita/+ mice (9 ± 0.04) and demonstrated significant attenuation of gliosis. Ins2Akita/+/Sig1R-/-mouse retinas, analyzed to determine whether the Ins2Akita/+ phenotype was accelerated when lacking Sigma1R, revealed increased nerve fiber layer thickness (OCT), evidence of vitreal opacities, and vessel beading (FA) compared with Ins2Akita/+ mice. Morphometric analysis revealed significantly fewer ganglion cells in Ins2Akita/+/Sig1R-/-mice compared with Ins2Akita/+ mice. CONCLUSIONS Sigma1R may be a novel retinal stress modulator, and targeting it even after disease onset may afford retinal neuroprotection.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Xuezhi Cui
- Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
| | - Penny Roon
- Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, United States
- The James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
40
|
Shanmugam AK, Mysona BA, Wang J, Zhao J, Tawfik A, Sanders A, Markand S, Zorrilla E, Ganapathy V, Bollinger KE, Smith SB. Progesterone Receptor Membrane Component 1 (PGRMC1) Expression in Murine Retina. Curr Eye Res 2015; 41:1105-1112. [PMID: 26642738 DOI: 10.3109/02713683.2015.1085579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Sigma receptors 1 (σR1) and 2 (σR2) are thought to be two distinct proteins which share the ability to bind multiple ligands, several of which are common to both receptors. Whether σR1 and σR2 share overlapping biological functions is unknown. Recently, progesterone receptor membrane component 1 (PGRMC1) was shown to contain the putative σR2 binding site. PGRMC1 has not been studied in retina. We hypothesize that biological interactions between σR1 and PGRMC1 will be evidenced by compensatory upregulation of PGRMC1 in σR1-/- mice. METHODS Immunofluorescence, RT-PCR, and immunoblotting methods were used to analyze expression of PGRMC1 in wild-type mouse retina. Tissues from σR1-/- mice were used to investigate whether a biological interaction exists between σR1 and PGRMC1. RESULTS In the eye, PGRMC1 is expressed in corneal epithelium, lens, ciliary body epithelium, and retina. In retina, PGRMC1 is present in Müller cells and retinal pigment epithelium. This expression pattern is similar, but not identical to σR1. PGRMC1 protein levels in neural retina and eye cup from σR1-/- mice did not differ from wild-type mice. Nonocular tissues, lung, heart, and kidney showed similar Pgrmc1 gene expression in wild-type and σR1-/- mice. In contrast, liver, brain, and intestine showed increased Pgrmc1 gene expression in σR1-/- mice. CONCLUSION Despite potential biological overlap, deletion of σR1 did not result in a compensatory change in PGRMC1 protein levels in σR1-/- mouse retina. Increased Pgrmc1 gene expression in organs with high lipid content such as liver, brain, and intestine indicates a possible tissue-specific interaction between σR1 and PGRMC1. The current studies establish the presence of PGRMC1 in retina and lay the foundation for analysis of its biological function.
Collapse
Affiliation(s)
- Arul K Shanmugam
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA.,b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA
| | - Barbara A Mysona
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA.,b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA
| | - Jing Wang
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA.,b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA
| | - Jing Zhao
- b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA.,c Department of Ophthalmology , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA
| | - Amany Tawfik
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA.,b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA
| | - A Sanders
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA
| | - Shanu Markand
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA.,b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA
| | - Eric Zorrilla
- d Harold L. Dorris Neurological Research Institute , The Scripps Research Institute , La Jolla , CA , USA
| | - Vadivel Ganapathy
- b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA.,e Department of Cell Biology and Biochemistry , Texas Tech University Health Sciences Center , Lubbock , TX , USA
| | - Kathryn E Bollinger
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA.,b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA.,c Department of Ophthalmology , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA
| | - Sylvia B Smith
- a Department of Cellular Biology and Anatomy , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA.,b James & Jean Culver Vision Discovery Institute , Georgia Regents University , Augusta , GA , USA.,c Department of Ophthalmology , Medical College of Georgia, Georgia Regents University , Augusta , GA , USA
| |
Collapse
|
41
|
van Waarde A, Rybczynska AA, Ramakrishnan NK, Ishiwata K, Elsinga PH, Dierckx RAJO. Potential applications for sigma receptor ligands in cancer diagnosis and therapy. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1848:2703-14. [PMID: 25173780 DOI: 10.1016/j.bbamem.2014.08.022] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/04/2014] [Accepted: 08/19/2014] [Indexed: 01/03/2023]
Abstract
Sigma receptors (sigma-1 and sigma-2) represent two independent classes of proteins. Their endogenous ligands may include the hallucinogen N,N-dimethyltryptamine (DMT) and sphingolipid-derived amines which interact with sigma-1 receptors, besides steroid hormones (e.g., progesterone) which bind to both sigma receptor subpopulations. The sigma-1 receptor is a ligand-regulated molecular chaperone with various ion channels and G-protein-coupled membrane receptors as clients. The sigma-2 receptor was identified as the progesterone receptor membrane component 1 (PGRMC1). Although sigma receptors are over-expressed in tumors and up-regulated in rapidly dividing normal tissue, their ligands induce significant cell death only in tumor tissue. Sigma ligands may therefore be used to selectively eradicate tumors. Multiple mechanisms appear to underlie cell killing after administration of sigma ligands, and the signaling pathways are dependent both on the type of ligand and the type of tumor cell. Recent evidence suggests that the sigma-2 receptor is a potential tumor and serum biomarker for human lung cancer and an important target for inhibiting tumor invasion and cancer progression. Current radiochemical efforts are focused on the development of subtype-selective radioligands for positron emission tomography (PET) imaging. Right now, the mostpromising tracers are [18F]fluspidine and [18F]FTC-146 for sigma-1 receptors and [11C]RHM-1 and [18F]ISO-1 for the sigma-2 subtype. Nanoparticles coupled to sigma ligands have shown considerable potential for targeted delivery of antitumor drugs in animal models of cancer, but clinical studies exploring this strategy in cancer patients have not yet been reported. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Anna A Rybczynska
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Nisha K Ramakrishnan
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Kiichi Ishiwata
- Tokyo Metropolitan Institute of Gerontology, Research Team for Neuroimaging, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo 173-0015, Japan
| | - Philip H Elsinga
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; University of Ghent, University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
42
|
Wang J, Shanmugam A, Markand S, Zorrilla E, Ganapathy V, Smith SB. Sigma 1 receptor regulates the oxidative stress response in primary retinal Müller glial cells via NRF2 signaling and system xc(-), the Na(+)-independent glutamate-cystine exchanger. Free Radic Biol Med 2015; 86:25-36. [PMID: 25920363 PMCID: PMC4554890 DOI: 10.1016/j.freeradbiomed.2015.04.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 04/02/2015] [Accepted: 04/07/2015] [Indexed: 01/18/2023]
Abstract
Oxidative stress figures prominently in retinal diseases, including diabetic retinopathy, and glaucoma. Ligands for σ1R, a unique transmembrane protein localized to the endoplasmic reticulum, mitochondria, and nuclear and plasma membranes, have profound retinal neuroprotective properties in vitro and in vivo. Studies to determine the mechanism of σ1R-mediated retinal neuroprotection have focused mainly on neurons. Little is known about the effects of σ1R on Müller cell function, yet these radial glial cells are essential for homeostatic support of the retina. Here we investigated whether σ1R mediates the oxidative stress response of Müller cells using wild-type (WT) and σ1R-knockout (σ1RKO) mice. We observed increased endogenous reactive oxygen species (ROS) levels in σ1RKO Müller cells compared to WT, which was accompanied by decreased expression of Sod1, catalase, Nqo1, Hmox1, Gstm6, and Gpx1. The protein levels of SOD1, CAT, NQO1, and GPX1 were also significantly decreased. The genes encoding these antioxidants contain an antioxidant response element (ARE), which under stress is activated by NRF2, a transcription factor that typically resides in the cytoplasm bound by KEAP1. In the σ1RKO Müller cells Nrf2 expression was decreased significantly at the gene (and protein) level, whereas Keap1 gene (and protein) levels were markedly increased. NRF2-ARE binding affinity was decreased markedly in σ1RKO Müller cells. We investigated system xc(-), the cystine-glutamate exchanger important for synthesis of glutathione (GSH), and observed decreased function in σ1RKO Müller cells compared to WT as well as decreased GSH and GSH/GSSG ratios. This was accompanied by decreased gene and protein levels of xCT, the unique component of system xc(-). We conclude that Müller glial cells lacking σ1R manifest elevated ROS, perturbation of antioxidant balance, suppression of NRF2 signaling, and impaired function of system xc(-). The data suggest that the oxidative stress-mediating function of retinal Müller glial cells may be compromised in the absence of σ1R. The neuroprotective role of σ1R may be linked directly to the oxidative stress-mediating properties of supportive glial cells.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA 30912-2000, USA; James and Jean Culver Vision Discovery Institute, Augusta, GA 30912-2000, USA
| | - Arul Shanmugam
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA 30912-2000, USA; James and Jean Culver Vision Discovery Institute, Augusta, GA 30912-2000, USA
| | - Shanu Markand
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA 30912-2000, USA; James and Jean Culver Vision Discovery Institute, Augusta, GA 30912-2000, USA
| | - Eric Zorrilla
- Harold L. Dorris Neurological Research Institute, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vadivel Ganapathy
- James and Jean Culver Vision Discovery Institute, Augusta, GA 30912-2000, USA; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912-2000, USA
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA 30912-2000, USA; James and Jean Culver Vision Discovery Institute, Augusta, GA 30912-2000, USA; Department of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912-2000, USA.
| |
Collapse
|
43
|
Abstract
This review compares the biological and physiological function of Sigma receptors [σRs] and their potential therapeutic roles. Sigma receptors are widespread in the central nervous system and across multiple peripheral tissues. σRs consist of sigma receptor one (σ1R) and sigma receptor two (σ2R) and are expressed in numerous regions of the brain. The sigma receptor was originally proposed as a subtype of opioid receptors and was suggested to contribute to the delusions and psychoses induced by benzomorphans such as SKF-10047 and pentazocine. Later studies confirmed that σRs are non-opioid receptors (not an µ opioid receptor) and play a more diverse role in intracellular signaling, apoptosis and metabolic regulation. σ1Rs are intracellular receptors acting as chaperone proteins that modulate Ca2+ signaling through the IP3 receptor. They dynamically translocate inside cells, hence are transmembrane proteins. The σ1R receptor, at the mitochondrial-associated endoplasmic reticulum membrane, is responsible for mitochondrial metabolic regulation and promotes mitochondrial energy depletion and apoptosis. Studies have demonstrated that they play a role as a modulator of ion channels (K+ channels; N-methyl-d-aspartate receptors [NMDAR]; inositol 1,3,5 triphosphate receptors) and regulate lipid transport and metabolism, neuritogenesis, cellular differentiation and myelination in the brain. σ1R modulation of Ca2+ release, modulation of cardiac myocyte contractility and may have links to G-proteins. It has been proposed that σ1Rs are intracellular signal transduction amplifiers. This review of the literature examines the mechanism of action of the σRs, their interaction with neurotransmitters, pharmacology, location and adverse effects mediated through them.
Collapse
Affiliation(s)
- Colin G Rousseaux
- a Department of Pathology and Laboratory Medicine , University of Ottawa , Ottawa , ON , Canada and
| | | |
Collapse
|
44
|
Mavlyutov TA, Epstein M, Guo LW. Subcellular localization of the sigma-1 receptor in retinal neurons - an electron microscopy study. Sci Rep 2015; 5:10689. [PMID: 26033680 PMCID: PMC4649997 DOI: 10.1038/srep10689] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/27/2015] [Indexed: 11/09/2022] Open
Abstract
The Sigma-1 receptor (S1R) is known to play a protective role in the central nervous system including the retina. A major barrier for understanding the underlying mechanism is an ambiguity of S1R subcellular localizations. We thus conducted the first electron microscopy (EM) study of S1R subcellular distribution in the mouse retina. Immuno-EM imaging showed previously under-appreciated S1R presence in photoreceptor cells. Unlike in other cell types in previous reports, in photoreceptor cells S1R was found in the nuclear envelope but not localized in the endoplasmic reticulum (ER), raising a possibility of S1R-mediated modulatory mechanisms different than conventionally thought. While in bipolar cells S1R was detected only in the nuclear envelope, in ganglion cells S1R was identified predominantly in the nuclear envelope and found in the ER as well. A predominant localization of S1R in the nuclear envelope in all three retinal neurons implicates a potential role of S1R in modulating nuclear activities. Moreover, its absence in the plasma membrane and presence in the subsurface ER cisternae that are juxtaposed to the plasma membrane in ganglion cells may lend mechanistic insights generally important for frequently reported S1R modulations of ion channels in neurons.
Collapse
Affiliation(s)
- Timur A. Mavlyutov
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
| | - Miles Epstein
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, 41 Bardeen Medical Laboratory, 470 N Charter Street, Madison, WI 53706, USA
| | - Lian-Wang Guo
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
| |
Collapse
|
45
|
Shanmugam A, Wang J, Markand S, Perry RL, Tawfik A, Zorrilla E, Ganapathy V, Smith SB. Sigma receptor 1 activation attenuates release of inflammatory cytokines MIP1γ, MIP2, MIP3α, and IL12 (p40/p70) by retinal Müller glial cells. J Neurochem 2015; 132:546-58. [PMID: 25439327 PMCID: PMC4451448 DOI: 10.1111/jnc.13002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 11/25/2014] [Indexed: 12/27/2022]
Abstract
The high-affinity sigma receptor 1 (σR1) ligand (+)-pentazocine ((+)-PTZ) affords profound retinal neuroprotection in vitro and in vivo by a yet-unknown mechanism. A common feature of retinal disease is Müller cell reactive gliosis, which includes cytokine release. Here, we investigated whether lipopolysaccharide (LPS) stimulates cytokine release by primary mouse Müller cells and whether (+)-PTZ alters release. Using a highly sensitive inflammatory antibody array we observed significant release of macrophage inflammatory proteins (MIP1γ, MIP2, MIP3α) and interleukin-12 (IL12 (p40/p70)) in LPS-treated cells compared to controls, and a significant decrease in secretion upon (+)-PTZ treatment. Müller cells from σR1 knockout mice demonstrated increased MIP1γ, MIP2, MIP3α and IL12 (p40/p70) secretion when exposed to LPS compared to LPS-stimulated WT cells. We investigated whether cytokine secretion was accompanied by cytosolic-to-nuclear NFκB translocation and whether endothelial cell adhesion/migration was altered by released cytokines. Cells exposed to LPS demonstrated increased NFκB nuclear location, which was reduced significantly in (+)-PTZ-treated cells. Media conditioned by LPS-stimulated-Müller cells induced leukocyte-endothelial cell adhesion and endothelial cell migration, which was attenuated by (+)-PTZ treatment. The findings suggest that release of certain inflammatory cytokines by Müller cells can be attenuated by σR1 ligands providing insights into the retinal neuroprotective role of this receptor.
Collapse
Affiliation(s)
- Arul Shanmugam
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
| | - Shanu Markand
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
| | - Richard L Perry
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
| | - Amany Tawfik
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
| | - Eric Zorrilla
- Harold L. Dorris Neurological Research Institute, The Scripps Research Institute, La Jolla, California, USA
| | - Vadivel Ganapathy
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, USA
- Department of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
46
|
Zhao J, Ha Y, Liou GI, Gonsalvez GB, Smith SB, Bollinger KE. Sigma receptor ligand, (+)-pentazocine, suppresses inflammatory responses of retinal microglia. Invest Ophthalmol Vis Sci 2014; 55:3375-84. [PMID: 24812552 PMCID: PMC4042630 DOI: 10.1167/iovs.13-12823] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 04/29/2014] [Indexed: 02/05/2023] Open
Abstract
PURPOSE To evaluate the effects of the σ 1 receptor (σR1) agonist, (+)-pentazocine, on lipopolysaccharide (LPS)-induced inflammatory changes in retinal microglia cells. METHODS Retinal microglia cells were isolated from Sprague-Dawley rat pups. Cells were treated with LPS with or without (+)-pentazocine and with or without the σR1 antagonist BD1063. Morphologic changes were assayed. Cell viability was assessed by using MTT assay. Supernatant levels of tumor necrosis factor α (TNF-α), interleukin 10, (IL-10), monocyte chemoattractant protein-1 (MCP-1), and nitric oxide (NO) were determined. Reactive oxygen species (ROS) formation was assayed, and levels of mitogen-activated protein kinases (MAPKs) were analyzed by using Western blot. RESULTS The σR1 protein was expressed in retinal microglia. Incubation with LPS and/or (+)-pentazocine did not alter cell viability or σR1 protein levels. Incubation with LPS for 24 hours induced a marked change in microglial morphology and a significant increase in secreted levels of TNF-α, IL-10, MCP-1, and NO. Pretreatment with (+)-pentazocine inhibited the LPS-induced morphologic changes. Release of TNF-α, IL-10, MCP-1, and NO was reduced with (+)-pentazocine. Intracellular ROS formation was suppressed with (+)-pentazocine. Phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was reduced in the presence of (+)-pentazocine. The σR1 antagonist BD1063 blocked the (+)-pentazocine-mediated inhibition of LPS-induced morphologic changes. In addition, BD1063 treatment blocked (+)-pentazocine-mediated suppression of LPS-induced TNF-α, IL-10, MCP-1, NO, and intracellular ROS release. CONCLUSIONS Treatment with (+)-pentazocine suppressed inflammatory responses of retinal microglia and inhibited LPS-induced activation of ERK/JNK MAPK. In neurodegenerative disease, (+)-pentazocine may exert neuroprotective effects through manipulation of microglia.
Collapse
Affiliation(s)
- Jing Zhao
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
| | - Yonju Ha
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Gregory I. Liou
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
- Department of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Graydon B. Gonsalvez
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Sylvia B. Smith
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
| | - Kathryn E. Bollinger
- James and Jean Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States
| |
Collapse
|
47
|
Ha Y, Shanmugam AK, Markand S, Zorrilla E, Ganapathy V, Smith SB. Sigma receptor 1 modulates ER stress and Bcl2 in murine retina. Cell Tissue Res 2014; 356:15-27. [PMID: 24469320 PMCID: PMC3976706 DOI: 10.1007/s00441-013-1774-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 11/18/2013] [Indexed: 01/08/2023]
Abstract
Sigma receptor 1 (σR1), a non-opiate transmembrane protein located on endoplasmic reticulum (ER) and mitochondrial membranes, is considered to be a molecular chaperone. Marked protection against cell death has been observed when ligands for σR1 have been used in in vitro and in vivo models of retinal cell death. Mice lacking σR1 (σR1(-/-)) manifest late-onset loss of retinal ganglion cells and retinal electrophysiological changes (after many months). The role of σR1 in the retina and the mechanisms by which its ligands afford neuroprotection are unclear. We therefore used σR1(-/-) mice to investigate the expression of ER stress genes (BiP/GRP78, Atf6, Atf4, Ire1α) and proteins involved in apoptosis (BCL2, BAX) and to examine the retinal transcriptome at young ages. Whereas no significant changes occurred in the expression of major ER stress genes (over a period of a year) in neural retina, marked changes were observed in these genes, especially Atf6, in isolated retinal Müller glial cells. BCL2 levels decreased in σR1(-/-) retina concomitantly with decreases in NFkB and pERK1/2. We postulate that σR1 regulates ER stress in retinal Müller cells and that the role of σR1 in retinal neuroprotection probably involves BCL2 and some of the proteins that modify its expression (such as ERK, NFκB). Data from the analysis of the retinal transcriptome of σR1 null mice provide new insights into the role of σR1 in retinal neuroprotection.
Collapse
Affiliation(s)
- Yonju Ha
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Arul K. Shanmugam
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Shanu Markand
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Eric Zorrilla
- Harold L. Dorris Neurological Research Institute, The Scripps Research Institute, La Jolla, CA
| | - Vadivel Ganapathy
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Georgia Regents University, Augusta, GA
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Georgia Regents University, Augusta, GA
- Department of Ophthalmology, Medical College of Georgia, Georgia Regents University, Augusta, GA
| |
Collapse
|
48
|
Contribution of retinal ganglion cells to the mouse electroretinogram. Doc Ophthalmol 2014; 128:155-68. [DOI: 10.1007/s10633-014-9433-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/06/2014] [Indexed: 01/21/2023]
|
49
|
Tawfik A, Al-Shabrawey M, Roon P, Sonne S, Covar JA, Matragoon S, Ganapathy PS, Atherton SS, El-Remessy A, Ganapathy V, Smith SB. Alterations of retinal vasculature in cystathionine-Beta-synthase mutant mice, a model of hyperhomocysteinemia. Invest Ophthalmol Vis Sci 2013; 54:939-49. [PMID: 23307965 DOI: 10.1167/iovs.12-10536] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Mice with moderate/severe hyperhomocysteinemia due to deficiency or absence of the cbs gene encoding cystathionine-beta-synthase (CBS) have marked retinal disruption, ganglion cell loss, optic nerve mitochondrial dysfunction, and ERG defects; those with mild hyperhomocysteinemia have delayed retinal morphological/functional phenotype. Excess homocysteine is a risk factor for cardiovascular diseases; however, it is not known whether excess homocysteine alters retinal vasculature. METHODS Cbs(+/+), cbs(+/-), and cbs(-/-) mice (age ∼3 weeks) were subjected to angiography; retinas were harvested for cryosections, flat-mount preparations, or trypsin digestion and subjected to immunofluorescence microscopy to visualize vessels using isolectin-B4, to detect angiogenesis using anti-VEGF and anti-endoglin (anti-CD105) and activated glial cells (anti-glial fibrillary acidic protein [anti-GFAP]) and to investigate the blood-retinal barrier using the tight junction markers zonula occludens-1 (ZO-1) and occludin. Expression of vegf was determined by quantitative RT-PCR (qRT-PCR) and immunoblotting. Human retinal endothelial cells (HRECs) were treated with excess homocysteine to analyze permeability. RESULTS Angiography revealed vascular leakage in cbs(-/-) mice; immunohistochemical analysis demonstrated vascular patterns consistent with ischemia; isolectin-B4 labeling revealed a capillary-free zone centrally and new vessels with capillary tufts midperipherally. This was associated with increased vegf mRNA and protein, CD105, and GFAP in cbs(-/-) retinas concomitant with a marked decrease in ZO-1 and occludin. Homocysteine-treated HRECs showed increased permeability. CONCLUSIONS Severe elevation of homocysteine in cbs(-/-) mutant mice is accompanied by alterations in retinal vasculature (ischemia, neovascularization, and incompetent blood-retinal barrier). The marked disruption of retinal structure and decreased visual function reported in cbs(-/-) mice may reflect vasculopathy as well as neuropathy.
Collapse
Affiliation(s)
- Amany Tawfik
- Departments of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ha Y, Saul A, Tawfik A, Zorrilla EP, Ganapathy V, Smith SB. Diabetes accelerates retinal ganglion cell dysfunction in mice lacking sigma receptor 1. Mol Vis 2012; 18:2860-70. [PMID: 23233788 PMCID: PMC3519370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 11/28/2012] [Indexed: 12/02/2022] Open
Abstract
PURPOSE Sigma receptor 1 (σR1) is a non-opioid transmembrane protein that may act as a molecular chaperone at the endoplasmic reticulum-mitochondrial membrane. Ligands for σR1, such as (+)-pentazocine [(+)-PTZ], confer marked retinal neuroprotection in vivo and in vitro. Recently we analyzed the retinal phenotype of mice lacking σR1 (σR1 KO) and observed normal retinal morphology and function in young mice (5-30 weeks) but diminished negative scotopic threshold responses (nSTRs), retinal ganglion cell (RGC) loss, and disruption of optic nerve axons consistent with inner retinal dysfunction by 1 year. These data led us to test the hypothesis that σR1 may be critical in forestalling chronic retinal stress; diabetes was used as the model of chronic stress. METHODS To determine whether σR1 is required for (+)-PTZ neuroprotective effects, primary RGCs isolated from wild-type (WT) and σR1 KO mice were exposed to xanthine-xanthine oxidase (10 µM:2 mU/ml) to induce oxidative stress in the presence or absence of (+)-PTZ. Cell death was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) analysis. To assess effects of chronic stress on RGC function, diabetes was induced in 3-week C57BL/6 (WT) and σR1 KO mice, using streptozotocin to yield four groups: WT nondiabetic (WT non-DB), WT diabetic (WT-DB), σR1 KO non-DB, and σR1 KO-DB. After 12 weeks of diabetes, when mice were 15-weeks old, intraocular pressure (IOP) was recorded, electrophysiologic testing was performed (including detection of nSTRs), and the number of RGCs was counted in retinal histological sections. RESULTS In vitro studies showed that (+)-PTZ could not prevent oxidative stress-induced death of RGCs harvested from σR1 KO mice but afforded robust protection against death of RGCs harvested from WT mice. In the studies of chronic stress induced by diabetes, the IOP measured in the four mouse groups was within the normal range; however, there was a significant increase in the IOP of σR1 KO-DB mice (16 ± 0.5 mmHg) compared to the other groups tested (σR1 KO non-DB, WT non-DB, WT-DB: ~12 ± 0.6 mmHg). Regarding electrophysiologic testing, the nSTRs of σR1 KO non-DB mice were similar to WT non-DB mice at 15 weeks; however, they were significantly lower in σR1 KO-DB mice (5 ± 1 µV) compared to the other groups, including, notably, σR1 KO-nonDB (12±2 µV). As expected, the number of RGCs in σR1 KO non-DB mice was similar to WT non-DB mice at 15 weeks, but under chronic stress of diabetes there were fewer RGCs in retinas of σR1 KO-DB mice. CONCLUSIONS This is the first report showing unequivocally that the neuroprotective effects of (+)-PTZ require σR1. σR1 KO mice show normal retinal structure and function at young ages; however, when subjected to the chronic stress of diabetes, there is an acceleration of retinal functional deficits in σR1 KO mice such that ganglion cell dysfunction is observed at a much earlier age than nondiabetic σR1 KO mice. The data support the hypothesis that σR1 plays a key role in modulating retinal stress and may be an important target for retinal disease.
Collapse
Affiliation(s)
- Yonju Ha
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
- Vision Discovery Institute, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
| | - Alan Saul
- Vision Discovery Institute, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
- Department of Ophthalmology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
| | - Amany Tawfik
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
- Vision Discovery Institute, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
| | | | - Vadivel Ganapathy
- Vision Discovery Institute, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
- Vision Discovery Institute, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
- Department of Ophthalmology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA
| |
Collapse
|