1
|
Bubis E, Sher I, Ketter-Katz H, Derzane E, Sennlaub F, Rotenstreich Y. Minocycline treatment reduces the activation of mononuclear phagocytes and improves retinal function in a mouse model of Leber congenital amaurosis. Graefes Arch Clin Exp Ophthalmol 2025:10.1007/s00417-025-06768-y. [PMID: 40528096 DOI: 10.1007/s00417-025-06768-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 06/20/2025] Open
Abstract
PURPOSE Leber congenital amaurosis (LCA) is a severe hereditary retinal degeneration characterized by early-onset vision loss. Here, we aimed to characterize the association between retinal mononuclear phagocyte (MP) activation and retinal degeneration in the RPE65/rd12 mouse model of LCA. METHODS Thirty-nine RPE65/rd12 and ten C57BL/6J wild-type mice were used. RPE65/rd12 mice were treated with minocycline by daily intraperitoneal injection (5 mg/kg) for eight weeks starting at age postnatal day 28 (P28). MP cell density in the subretina was determined by choroid-retinal pigment epithelium (RPE) flat mount analysis, and retinal function was determined by electroretinogram (ERG). RESULTS In wild-type C57BL/6J mice, MPs were exclusively located in the inner retinal layers at ages P28-P84. By contrast, in the RPE65/rd12 mice, MPs migrated into the subretina as early as P56 in a central-to-peripheral gradient. By P84, the density of MPs in the subretina increased by nearly 3-fold, reaching 61.3 ± 6.2 cell/mm2 and 33.1 ± 8 cell/mm2 in the central and peripheral retina, respectively. Minocycline treatment significantly reduced MP density in the peripheral subretina (16.2 ± 1.8 MP cell/mm2) compared with mice treated with PBS (27.2 ± 2.4 MP cell/mm2, respectively, p = 0.006). Maximal electroretinogram b-wave responses were significantly higher in minocycline- vs. PBS-treated mice under light-adapted conditions following eight weeks of treatment (mean ± SE: 199µv ± 28µv vs. 129.8µv ± 9.8µv, p = 0.016). CONCLUSIONS Our data indicates that MP migration into the subretina is associated with retinal degeneration in RPE65/rd12 mice. Inhibiting MP migration into the subretina was associated with improved retinal function. These findings may guide the development of therapies targeting MP activation for neuroprotection in LCA and potentially other retinoid cycle-related retinal degeneration blinding diseases.
Collapse
Affiliation(s)
- Ettel Bubis
- Goldschleger Eye Institute, Sheba Medical Center, Tel-Hashomer, 5262100, Israel
- Ophthalmology Department, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv, 6997801, Israel
| | - Ifat Sher
- Goldschleger Eye Institute, Sheba Medical Center, Tel-Hashomer, 5262100, Israel
- Ophthalmology Department, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv, 6997801, Israel
- The TELEM Rubin Excellence in Biomedical Research Program, Sheba Medical Center, Tel Hashomer, 5262100, Israel
| | - Hadas Ketter-Katz
- Goldschleger Eye Institute, Sheba Medical Center, Tel-Hashomer, 5262100, Israel
- Ophthalmology Department, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv, 6997801, Israel
| | - Estela Derzane
- Academic Center for Continuing Medical Education, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Florian Sennlaub
- Institut de la Vision, 17 rue Moreau, Paris, 75012, France
- Sorbonne Universités, UPMC University Paris 06, INSERM, CNRS, Paris, 75012, France
| | - Ygal Rotenstreich
- Goldschleger Eye Institute, Sheba Medical Center, Tel-Hashomer, 5262100, Israel.
- Ophthalmology Department, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv, 6997801, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
2
|
Zhang N, Li Y, Zhang X, Chrenek MA, Wang J, Girardot PE, Sellers JT, Geisert EE, Nickerson JM, Boatright JH. Tauroursodeoxycholic Acid Protects Retinal Ganglion Cells and Reduces Inflammation in Mice Following Optic Nerve Crush. Pharmaceuticals (Basel) 2025; 18:569. [PMID: 40284004 PMCID: PMC12030659 DOI: 10.3390/ph18040569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
Purpose: The aim of this study was to investigate the protective effects of systemically administered tauroursodeoxycholic acid (TUDCA) in an optic nerve crush (ONC) mouse model of retinal ganglion cell (RGC) death. Methods: C57BL/6J mice were injected intraperitoneally (i.p.) three times per week with TUDCA (500 mg/kg) for two weeks, after which unilateral ONC was performed. A control cohort was identically treated with a drug vehicle (phosphate buffered saline; PBS). A separate cohort did not undergo any injections or surgeries (this was termed the "Naïve" group). Pattern electroretinography (PERG) was recorded 3 days after ONC. Retinas were harvested for whole-mount immunofluorescence staining with an antibody against RGC marker Brn3a and imaged by fluorescent confocal microscopy. Apoptotic cells in the ganglion cell layer (GCL) were detected by Terminal Deoxynucleotidyl Transferase-Mediated dUTP Nick End Labeling (TUNEL) performed on fixed retina sections. Glial fibrillary acidic protein (GFAP) immunostaining on fixed retina sections was conducted to detect the activation of Müller cells. Total RNA was extracted from retinas and expression of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and IL-10 was determined by digital droplet PCR (ddPCR). Results: TUDCA treatment preserved visual function as assessed by PERG. P1 and N2 amplitudes from the PBS-treated ONC group were significantly diminished compared to those of the Naïve group (p < 0.001). TUDCA treatment prevented this diminution. The amplitudes of P1 and N2 in the TUDCA-treated ONC group were statistically indistinguishable from those of the Naïve group and were higher than the PBS-treated ONC group (TUDCA+ONC vs. PBS+ONC, P1: 6.99 ± 0.89 µV vs. 3.60 ± 0.69 µV, p < 0.01; N2: -9.30 (IQR: -13.43--6.44) µV vs. -4.47 (IQR: -10.26--2.17) µV). TUDCA treatment preserved RGCs. The ONC-vehicle-only group had 25% fewer RGCs (Brn3a-positive cells) than Naïve eyes (p < 0.0001). TUDCA treatment nearly completely prevented this loss, preserving all but 7.7% of the RGCs, and the number of RGCs in the TUDCA-treated ONC group was significantly higher than in the PBS-treated ONC group (TUDCA+ONC vs. PBS+ONC, 1738.00 ± 14.43 cells per field vs. 1454.00 ± 6.55 cells per field, p < 0.0001). The number of TUNEL-positive cells in the GCL (Naïve vs. PBS+ONC group: 1.00 (IQR: 0.00-2.00) % vs. 37.00 (IQR: 8.50-48.50) %, p < 0.05) and GFAP-positive fibers transversing retina sections (Naïve vs. PBS+ONC group: 33.00 ± 1.15 vs. 185.70 ± 42.37 fibers/retina, p < 0.05), and the expression of IL-6, TNF-α were significantly greater in the PBS-treated ONC group compared to that of the Naïve group (Naïve vs. PBS+ONC group, IL-6: 0.07 (IQR: 0.06-0.31) vs. 0.99 (IQR: 0.56-1.47), p < 0.05, TNF-α: 0.19 ± 0.069 vs. 1.39 ± 0.23; p < 0.01), an increase not observed with TUDCA treatment. Conclusions: Systemic TUDCA treatment significantly preserved RGC function and survival in the mouse ONC model of RGC damage. TUDCA treatment prevented RGC apoptosis, Müller glial cell activation, and retinal expression of several inflammatory cytokines. These data suggest that TUDCA is a promising therapeutic candidate for preserving RGC numbers and function.
Collapse
Affiliation(s)
- Nan Zhang
- Atlanta Veterans Administration Center for Visual and Cognitive Rehabilitation, Decatur, GA 30033, USA; (N.Z.); (Y.L.); (X.Z.); (P.E.G.); (J.T.S.)
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
- Department of Ophthalmology, Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Ying Li
- Atlanta Veterans Administration Center for Visual and Cognitive Rehabilitation, Decatur, GA 30033, USA; (N.Z.); (Y.L.); (X.Z.); (P.E.G.); (J.T.S.)
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
| | - Xian Zhang
- Atlanta Veterans Administration Center for Visual and Cognitive Rehabilitation, Decatur, GA 30033, USA; (N.Z.); (Y.L.); (X.Z.); (P.E.G.); (J.T.S.)
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
- Department of Ophthalmology, Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Micah A. Chrenek
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
| | - Jiaxing Wang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Preston E. Girardot
- Atlanta Veterans Administration Center for Visual and Cognitive Rehabilitation, Decatur, GA 30033, USA; (N.Z.); (Y.L.); (X.Z.); (P.E.G.); (J.T.S.)
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
| | - Jana T. Sellers
- Atlanta Veterans Administration Center for Visual and Cognitive Rehabilitation, Decatur, GA 30033, USA; (N.Z.); (Y.L.); (X.Z.); (P.E.G.); (J.T.S.)
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
| | - Eldon E. Geisert
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
| | - John M. Nickerson
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
| | - Jeffrey H. Boatright
- Atlanta Veterans Administration Center for Visual and Cognitive Rehabilitation, Decatur, GA 30033, USA; (N.Z.); (Y.L.); (X.Z.); (P.E.G.); (J.T.S.)
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.A.C.); (E.E.G.); (J.W.); (J.M.N.)
| |
Collapse
|
3
|
Petrogiannakis G, Guadagnino I, Negueruela S, Di Guida M, Marrocco E, Pizzo M, Torella A, Zanobio M, Karali M, Medina DL, Carrella S, Banfi S. In vitro high-content screening reveals miR-429 as a protective molecule in photoreceptor degeneration. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102434. [PMID: 39877002 PMCID: PMC11773019 DOI: 10.1016/j.omtn.2024.102434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025]
Abstract
Inherited retinal diseases (IRDs) are clinically and genetically heterogeneous disorders characterized by progressive photoreceptor degeneration and irreversible vision loss. MicroRNAs (miRNAs), a class of endogenous non-coding RNAs with post-transcriptional regulatory properties, are known to play a major role in retinal function, both in physiological and pathological conditions. Given their ability to simultaneously modulate multiple molecular pathways, miRNAs represent promising therapeutic tools for disorders with high genetic heterogeneity, such as IRDs. In the present study, we performed high-content imaging (HCI) screening to assess the impact of miRNA overexpression on a photoreceptor cell line undergoing light-induced degeneration. More than 1,200 miRNAs were assayed for putative protective effects in light-stressed 661W photoreceptor-like cells, and the top-performing miRNAs were further validated in independent in vitro assays. miR-429 showed the strongest cell-protective effect in vitro. Adeno-associated viral vector-mediated subretinal delivery of miR-429 in the Rho P23H/+ IRD mouse model preserved electrophysiological responses and was associated with reduced inflammatory processes in the retina. We demonstrate that the HCI in vitro assay we devised is a reliable screening method to select candidate molecules for mutation-independent therapeutic approaches for retinal disorders. Moreover, our data indicate that miR-429 represents a potential therapeutic target against photoreceptor degeneration.
Collapse
Affiliation(s)
- Georgios Petrogiannakis
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Irene Guadagnino
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Santiago Negueruela
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Martina Di Guida
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Elena Marrocco
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Mariateresa Pizzo
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Annalaura Torella
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - Mariateresa Zanobio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy
| | - Marianthi Karali
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, Eye Clinic, University of Campania “Luigi Vanvitelli”, Via Pansini 5, 80131 Naples, Italy
| | - Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
- Department of Medical and Translational Science, Federico II University, 80131 Naples, Italy
| | - Sabrina Carrella
- Biology and Evolution of Marine Organisms Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80133 Naples, Italy
| | - Sandro Banfi
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
4
|
Azam M, Jastrzebska B. Mechanisms of Rhodopsin-Related Inherited Retinal Degeneration and Pharmacological Treatment Strategies. Cells 2025; 14:49. [PMID: 39791750 PMCID: PMC11720364 DOI: 10.3390/cells14010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
Retinitis pigmentosa (RP) is a hereditary disease characterized by progressive vision loss ultimately leading to blindness. This condition is initiated by mutations in genes expressed in retinal cells, resulting in the degeneration of rod photoreceptors, which is subsequently followed by the loss of cone photoreceptors. Mutations in various genes expressed in the retina are associated with RP. Among them, mutations in the rhodopsin gene (RHO) are the most common cause of this condition. Due to the involvement of numerous genes and multiple mutations in a single gene, RP is a highly heterogeneous disease making the development of effective treatments particularly challenging. The progression of this disease involves complex cellular responses to restore cellular homeostasis, including the unfolded protein response (UPR) signaling, autophagy, and various cell death pathways. These mechanisms, however, often fail to prevent photoreceptor cell degradation and instead contribute to cell death under certain conditions. Current research focuses on the pharmacological modulation of the components of these pathways and the direct stabilization of mutated receptors as potential treatment strategies. Despite these efforts, the intricate interplay between these mechanisms and the diverse causative mutations involved has hindered the development of effective treatments. Advancing our understanding of the interactions between photoreceptor cell death mechanisms and the specific genetic mutations driving RP is critical to accelerate the discovery and development of therapeutic strategies for this currently incurable disease.
Collapse
Affiliation(s)
- Maria Azam
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | - Beata Jastrzebska
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
- Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
5
|
Meseguer D, Furtado J, Zapadka T, Rodriguez A, Na D, Grajales-Reyes JG, Demb J, Eichmann A, Schneeberger M. Early exposure to Western Diet exacerbates visual outcomes in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625688. [PMID: 39677769 PMCID: PMC11642743 DOI: 10.1101/2024.11.27.625688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Obesity, a growing pandemic in Western societies, significantly impacts metabolic health and contributes to visual disorders. While the systemic consequences of obesity, such as chronic inflammation and insulin resistance, are well-studied in adults, its early-life effects on retinal health remain underexplored. Using a maternal Western Diet (WD) exposure model, we investigated the developmental impact of early-life metabolic disturbances on retinal and cognitive function. Our findings reveal that WD exposure from gestation to early adulthood accelerates the onset of features resembling diabetic retinopathy, including increased retinal vascularization, inflammation, and compromised blood-retina barrier integrity, observed within just four months. Females exhibited heightened vulnerability, showing pronounced ocular defects such as anophthalmia, microphthalmia, and congenital cataracts. These results underscore a critical developmental window during which metabolic disruptions predispose to sex-specific retinal and neurovascular pathologies. This work bridges the link between pediatric and adult obesity, highlighting the urgent need for early interventions to mitigate long-term visual impairments that could further impair recognition memory.
Collapse
Affiliation(s)
- David Meseguer
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Thomas Zapadka
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA. Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Amanda Rodriguez
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Daxiang Na
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Jose G. Grajales-Reyes
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Department of Anesthesiology, Yale University, New Haven, CT, USA
| | - Jonathan Demb
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA. Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Université de Paris, INSERM, PARCC, F-75015, Paris, France. Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA
| |
Collapse
|
6
|
Liao C, Chen S, Chen X, Yi W, Fan Y, Chen Y, Ye T, Chen Y. Inhibition of JNK ameliorates rod photoreceptor degeneration in a mouse model of retinitis pigmentosa. FEBS Lett 2024; 598:2683-2701. [PMID: 39010325 DOI: 10.1002/1873-3468.14978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 07/17/2024]
Abstract
Retinitis pigmentosa (RP) is an inherited eye disease that causes progressive vision loss. Microglial activation and inflammation play essential roles in photoreceptor degeneration in RP, although the underlying mechanisms remain unclear. Here, we examined the progressive degeneration of photoreceptors in rd1 mice, a mouse model of RP. We investigated the molecular changes in various retinal cells in rd1 mice using single-cell RNA sequencing and found that potentiation of JNK signaling is associated with photoreceptor degeneration in RP. Moreover, inflammation-related molecules, which function downstream of JNK, are elevated in RP. Furthermore, inhibiting JNK alleviates microglial activation and rescues photoreceptor degeneration in rd1 mice. Thus, our findings suggest that targeting JNK is a promising approach for slowing RP progression.
Collapse
Affiliation(s)
- Chunyan Liao
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Shuai Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Xuxu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Wanying Yi
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, China
| | - Yingying Fan
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, China
| | - Tao Ye
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, China
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, China
| |
Collapse
|
7
|
Yang M, Yao J, Jia L, Kocab AJ, Zacks DN. Preservation of retinal structure and function in two mouse models of inherited retinal degeneration by ONL1204, an inhibitor of the Fas receptor. Cell Death Dis 2024; 15:576. [PMID: 39117629 PMCID: PMC11310419 DOI: 10.1038/s41419-024-06970-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Due to the large number of genes and mutations that result in inherited retinal degenerations (IRD), there has been a paucity of therapeutic options for these patients. There is a large unmet need for therapeutic approaches targeting shared pathophysiologic pathways in a mutation-independent manner. The Fas receptor is a major activator and regulator of retinal cell death and inflammation in a variety of ocular diseases. We previously reported the activation of Fas-mediated photoreceptor (PR) cell death in two different IRD mouse models, rd10 and P23H, and demonstrated the protective effect of genetic Fas inhibition. The purpose of this study was to examine the effects of pharmacologic inhibition of Fas in these two models by intravitreal injection with a small peptide inhibitor of the Fas receptor, ONL1204. A single intravitreal injection of ONL1204 was given to one eye of rd10 mice at P14. Two intravitreal injections of ONL1204 were given to the P23H mice, once at P14 and again at 2-months of age. The fellow eyes were injected with vehicle alone. Fas activation, rate of PR cell death, retinal function, and the activation of immune cells in the retina were evaluated. In both rd10 and P23H mice, ONL1204 treatment resulted in decreased number of TUNEL (+) PRs, decreased caspase 8 activity, enhanced photoreceptor cell counts, and improved visual function compared with vehicle treated fellow eyes. Treatment with ONL1204 also reduced immune cell activation in the retinas of both rd10 and P23H mice. The protective effect of pharmacologic inhibition of Fas by ONL1204 in two distinct mouse models of retinal degeneration suggests that targeting this common pathophysiologic mechanism of cell death and inflammation represents a potential therapeutic approach to preserve the retina in patients with IRD, regardless of the genetic underpinning.
Collapse
Affiliation(s)
- Mengling Yang
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
- Eye Center of Xiangya Hospital, Xiangya School of medicine, Central South University, Changsha, Hunan, China
| | - Jingyu Yao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
| | - Lin Jia
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA
| | | | - David N Zacks
- Department of Ophthalmology and Visual Sciences, University of Michigan, Kellogg Eye Center, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Nguyen Y, Rudd Zhong Manis J, Ronczkowski NM, Bui T, Oxenrider A, Jadeja RN, Thounaojam MC. Unveiling the gut-eye axis: how microbial metabolites influence ocular health and disease. Front Med (Lausanne) 2024; 11:1377186. [PMID: 38799150 PMCID: PMC11122920 DOI: 10.3389/fmed.2024.1377186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024] Open
Abstract
The intricate interplay between the gut microbiota and ocular health has surpassed conventional medical beliefs, fundamentally reshaping our understanding of organ interconnectivity. This review investigates into the intricate relationship between gut microbiota-derived metabolites and their consequential impact on ocular health and disease pathogenesis. By examining the role of specific metabolites, such as short-chain fatty acids (SCFAs) like butyrate and bile acids (BAs), herein we elucidate their significant contributions to ocular pathologies, thought-provoking the traditional belief of organ sterility, particularly in the field of ophthalmology. Highlighting the dynamic nature of the gut microbiota and its profound influence on ocular health, this review underlines the necessity of comprehending the complex workings of the gut-eye axis, an emerging field of science ready for further exploration and scrutiny. While acknowledging the therapeutic promise in manipulating the gut microbiome and its metabolites, the available literature advocates for a targeted, precise approach. Instead of broad interventions, it emphasizes the potential of exploiting specific microbiome-related metabolites as a focused strategy. This targeted approach compared to a precision tool rather than a broad-spectrum solution, aims to explore the therapeutic applications of microbiome-related metabolites in the context of various retinal diseases. By proposing a nuanced strategy targeted at specific microbial metabolites, this review suggests that addressing specific deficiencies or imbalances through microbiome-related metabolites might yield expedited and pronounced outcomes in systemic health, extending to the eye. This focused strategy holds the potential in bypassing the irregularity associated with manipulating microbes themselves, paving a more efficient pathway toward desired outcomes in optimizing gut health and its implications for retinal diseases.
Collapse
Affiliation(s)
- Yvonne Nguyen
- Mercer University School of Medicine, Macon, GA, United States
| | | | | | - Tommy Bui
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Allston Oxenrider
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Ravirajsinh N. Jadeja
- Biochemistry and Molecular Biology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Menaka C. Thounaojam
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| |
Collapse
|
9
|
Etebar F, Harkin DG, White AR, Dando SJ. Non-invasive in vivo imaging of brain and retinal microglia in neurodegenerative diseases. Front Cell Neurosci 2024; 18:1355557. [PMID: 38348116 PMCID: PMC10859418 DOI: 10.3389/fncel.2024.1355557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Microglia play crucial roles in immune responses and contribute to fundamental biological processes within the central nervous system (CNS). In neurodegenerative diseases, microglia undergo functional changes and can have both protective and pathogenic roles. Microglia in the retina, as an extension of the CNS, have also been shown to be affected in many neurological diseases. While our understanding of how microglia contribute to pathological conditions is incomplete, non-invasive in vivo imaging of brain and retinal microglia in living subjects could provide valuable insights into their role in the neurodegenerative diseases and open new avenues for diagnostic biomarkers. This mini-review provides an overview of the current brain and retinal imaging tools for studying microglia in vivo. We focus on microglia targets, the advantages and limitations of in vivo microglia imaging approaches, and applications for evaluating the pathogenesis of neurological conditions, such as Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Fazeleh Etebar
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Damien G. Harkin
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Anthony R. White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Samantha J. Dando
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| |
Collapse
|
10
|
Yang P, Mustafi D, Pepple KL. Immunology of Retinitis Pigmentosa and Gene Therapy-Associated Uveitis. Cold Spring Harb Perspect Med 2024; 14:a041305. [PMID: 37037600 PMCID: PMC10562523 DOI: 10.1101/cshperspect.a041305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
The underlying immune state of inherited retinal degenerations (IRDs) and retinitis pigmentosa (RP) has been an emerging area of interest, wherein the consequences have never been greater given the widespread recognition of gene therapy-associated uveitis (GTU) in gene therapy clinical trials. Whereas some evidence suggests that the adaptive immune system may play a role, the majority of studies indicate that the innate immune system is likely the primary driver of neuroinflammation in RP. During retinal degeneration, discrete mechanisms activate resident microglia and promote infiltrating macrophages that can either be protective or detrimental to photoreceptor cell death. This persistent stimulation of innate immunity, overlaid by the introduction of viral antigens as part of gene therapy, has the potential to trigger a complex microglia/macrophage-driven proinflammatory state. A better understanding of the immune pathophysiology in IRD and GTU will be necessary to improve the success of developing novel treatments for IRDs.
Collapse
Affiliation(s)
- Paul Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregan 97239, USA
| | - Debarshi Mustafi
- Department of Ophthalmology, Roger and Karalis Johnson Retina Center, University of Washington, Seattle, Washington 98109, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington 98109, USA
- Department of Ophthalmology, Seattle Children's Hospital, Seattle, Washington 98109, USA
| | - Kathryn L Pepple
- Department of Ophthalmology, Roger and Karalis Johnson Retina Center, University of Washington, Seattle, Washington 98109, USA
| |
Collapse
|
11
|
Li J, Huang Z, Jin Y, Liang L, Li Y, Xu K, Zhou W, Li X. Neuroprotective Effect of Tauroursodeoxycholic Acid (TUDCA) on In Vitro and In Vivo Models of Retinal Disorders: A Systematic Review. Curr Neuropharmacol 2024; 22:1374-1390. [PMID: 37691227 PMCID: PMC11092919 DOI: 10.2174/1570159x21666230907152207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 02/15/2023] [Accepted: 03/07/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Tauroursodeoxycholic acid (TUDCA) is a naturally produced hydrophilic bile acid that has been used for centuries in Chinese medicine. Numerous recent in vitro and in vivo studies have shown that TUDCA has neuroprotective action in various models of retinal disorders. OBJECTIVE To systematically review the scientific literature and provide a comprehensive summary on the neuroprotective action and the mechanisms involved in the cytoprotective effects of TUDCA. METHODS A systematic review was conducted in accordance with the PRISMA (The Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. Systematic literature search of United States National Library of Medicine (PubMed), Web of Science, Embase, Scopus and Cochrane Library was performed, which covered all original articles published up to July 2022. The terms, "TUDCA" in combination with "retina", "retinal protection", "neuroprotection" were searched. Possible biases were identified with the adopted SYRCLE's tool. RESULTS Of the 423 initially gathered studies, 24 articles met inclusion/exclusion criteria for full-text review. Six of them were in vitro experiments, 17 studies reported in vivo data and one study described both in vitro and in vivo data. The results revealed the effect of TUDCA on different retinal diseases, such as retinitis pigmentosa (RP), diabetic retinopathy (DR), retinal degeneration (RD), retinal ganglion cell (RGC) injury, Leber's hereditary optic neuropathy (LHON), choroidal neovascularization (CNV), and retinal detachment (RDT). The quality scores of the in vivo studies were ranged from 5 to 7 points (total 10 points), according to SYRCLE's risk of bias tool. Both in vitro and in vivo data suggested that TUDCA could effectively delay degeneration and apoptosis of retinal neurons, preserve retinal structure and function, and its mechanism of actions might be related with inhibiting apoptosis, decreasing inflammation, attenuating oxidative stress, suppressing endoplasmic reticulum (ER) stress, and reducing angiogenesis. CONCLUSION This systematic review demonstrated that TUDCA has neuroprotective effect on in vivo and in vitro models of retinal disorders, reinforcing the currently available evidence that TUDCA could be a promising therapeutic agent in retinal diseases treatment. However, well designed clinical trials are necessary to appraise the efficacy of TUDCA in clinical setting.
Collapse
Affiliation(s)
- Jiaxian Li
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Ziyang Huang
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Yu Jin
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Lina Liang
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Yamin Li
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Kai Xu
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Wei Zhou
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Xiaoyu Li
- Department of Eye Function Laboratory, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| |
Collapse
|
12
|
Pérez-Fernández V, Thananjeyan AL, Ullah F, Münch G, Cameron M, Gyengesi E. The effects of a highly bioavailable curcumin Phytosome TM preparation on the retinal architecture and glial reactivity in the GFAP-IL6 mice. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1205542. [PMID: 38983084 PMCID: PMC11182199 DOI: 10.3389/fopht.2023.1205542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/08/2023] [Indexed: 07/11/2024]
Abstract
Uncontrolled, chronic inflammation in the retina can disturb retinal structure and function leading to impaired visual function. For the first time, in a mouse model of chronic neuroinflammation (GFAP-IL6), we investigated the impact of chronic glial activation on the retinal microglia population and structure. In addition, we tested a curcumin PhytosomeTM preparation with enhanced bioavailability to investigate the effects of a cytokine-suppressing anti-inflammatory drug on retinal architecture. Curcumin PhytosomeTM was fed to 3-month old GFAP-IL6 mice for 4 weeks and compared to their untreated GFAP-IL6 counterparts as well as wild type mice on control diet. Microglial numbers and morphology together with neuronal numbers were characterized using immunohistochemistry and cell reconstruction in the retina, using retinal wholemount and slices. GFAP-IL6 mice showed a significant increase in Iba1-labelled mononuclear phagocytes, including microglia, and displayed altered glial morphology. This resulted in a reduction in cone density and a thinning of the retinal layers compared to wild type mice. Curcumin PhytosomeTM treatment contributed to decreased microglial density, significantly decreasing both soma and cell size compared to control diet, as well as preventing the thinning of the retinal layers. This study is the first to characterize the impact of chronic retinal inflammation in the GFAP-IL6 mouse and the therapeutic benefit of enhanced bioavailable curcumin PhytosomeTM to significantly reduce microglia density and prevent neuronal loss. These data suggest that curcumin could be used as a complementary therapy alongside traditional treatments to reduce associated retinal inflammation in a variety of retinal diseases.
Collapse
Affiliation(s)
- Víctor Pérez-Fernández
- Department of Anatomy and Cell Biology, Western Sydney University, Campbelltown, NSW, Australia
| | | | - Faheem Ullah
- Department of Pharmacology, Western Sydney University, Campbelltown, NSW, Australia
- Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| | - Gerald Münch
- Department of Pharmacology, Western Sydney University, Campbelltown, NSW, Australia
| | - Morven Cameron
- Department of Anatomy and Cell Biology, Western Sydney University, Campbelltown, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
13
|
Díaz-Lezama N, Kajtna J, Wu J, Ayten M, Koch SF. Microglial and macroglial dynamics in a model of retinitis pigmentosa. Vision Res 2023; 210:108268. [PMID: 37295269 DOI: 10.1016/j.visres.2023.108268] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
In retinal degenerative diseases, such as retinitis pigmentosa (RP), the characteristic photoreceptor cell death is associated with changes of microglia and macroglia cells. Gene therapy, a promising treatment option for RP, is based on the premise that glial cell remodeling does not impact vision rescue. However, the dynamics of glial cells after treatment at late disease stages are not well understood. Here, we tested the reversibility of specific RP glia phenotypes in a Pde6b-deficient RP gene therapy mouse model. We demonstrated an increased number of activated microglia, retraction of microglial processes, reactive gliosis of Müller cells, astrocyte remodelling and an upregulation of glial fibrillary acidic protein (GFAP) in response to photoreceptor degeneration. Importantly, these changes returned to normal following rod rescue at late disease stages. These results suggest that therapeutic approaches restore the homeostasis between photoreceptors and glial cells.
Collapse
Affiliation(s)
- Nundehui Díaz-Lezama
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Jacqueline Kajtna
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Jiou Wu
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Monika Ayten
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Susanne F Koch
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, 81377 Munich, Germany.
| |
Collapse
|
14
|
Xing C, Huang X, Wang D, Yu D, Hou S, Cui H, Song L. Roles of bile acids signaling in neuromodulation under physiological and pathological conditions. Cell Biosci 2023; 13:106. [PMID: 37308953 PMCID: PMC10258966 DOI: 10.1186/s13578-023-01053-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/13/2023] [Indexed: 06/14/2023] Open
Abstract
Bile acids (BA) are important physiological molecules not only mediating nutrients absorption and metabolism in peripheral tissues, but exerting neuromodulation effect in the central nerve system (CNS). The catabolism of cholesterol to BA occurs predominantly in the liver by the classical and alternative pathways, or in the brain initiated by the neuronal-specific enzyme CYP46A1 mediated pathway. Circulating BA could cross the blood brain barrier (BBB) and reach the CNS through passive diffusion or BA transporters. Brain BA might trigger direct signal through activating membrane and nucleus receptors or affecting activation of neurotransmitter receptors. Peripheral BA may also provide the indirect signal to the CNS via farnesoid X receptor (FXR) dependent fibroblast growth factor 15/19 (FGF15/19) pathway or takeda G protein coupled receptor 5 (TGR5) dependent glucagon-like peptide-1 (GLP-1) pathway. Under pathological conditions, alterations in BA metabolites have been discovered as potential pathogenic contributors in multiple neurological disorders. Attractively, hydrophilic ursodeoxycholic acid (UDCA), especially tauroursodeoxycholic acid (TUDCA) can exert neuroprotective roles by attenuating neuroinflammation, apoptosis, oxidative or endoplasmic reticulum stress, which provides promising therapeutic effects for treatment of neurological diseases. This review summarizes recent findings highlighting the metabolism, crosstalk between brain and periphery, and neurological functions of BA to elucidate the important role of BA signaling in the brain under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Chen Xing
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China.
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
| | - Dongxue Wang
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - Dengjun Yu
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - Shaojun Hou
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
- Anhui Medical University, Heifei, 230032, China
| | - Haoran Cui
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China
| | - Lung Song
- Beijing Institute of Basic Medical Sciences, Taiping Road #27, Beijing, 100850, China.
- Anhui Medical University, Heifei, 230032, China.
| |
Collapse
|
15
|
Zhen F, Zou T, Wang T, Zhou Y, Dong S, Zhang H. Rhodopsin-associated retinal dystrophy: Disease mechanisms and therapeutic strategies. Front Neurosci 2023; 17:1132179. [PMID: 37077319 PMCID: PMC10106759 DOI: 10.3389/fnins.2023.1132179] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
Rhodopsin is a light-sensitive G protein-coupled receptor that initiates the phototransduction cascade in rod photoreceptors. Mutations in the rhodopsin-encoding gene RHO are the leading cause of autosomal dominant retinitis pigmentosa (ADRP). To date, more than 200 mutations have been identified in RHO. The high allelic heterogeneity of RHO mutations suggests complicated pathogenic mechanisms. Here, we discuss representative RHO mutations as examples to briefly summarize the mechanisms underlying rhodopsin-related retinal dystrophy, which include but are not limited to endoplasmic reticulum stress and calcium ion dysregulation resulting from protein misfolding, mistrafficking, and malfunction. Based on recent advances in our understanding of disease mechanisms, various treatment methods, including adaptation, whole-eye electrical stimulation, and small molecular compounds, have been developed. Additionally, innovative therapeutic treatment strategies, such as antisense oligonucleotide therapy, gene therapy, optogenetic therapy, and stem cell therapy, have achieved promising outcomes in preclinical disease models of rhodopsin mutations. Successful translation of these treatment strategies may effectively ameliorate, prevent or rescue vision loss related to rhodopsin mutations.
Collapse
Affiliation(s)
- Fangyuan Zhen
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Provincial Ophthalmic Hospital, Zhengzhou, China
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tongdan Zou
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ting Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yongwei Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Provincial Ophthalmic Hospital, Zhengzhou, China
| | - Shuqian Dong
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Henan Provincial Ophthalmic Hospital, Zhengzhou, China
- *Correspondence: Shuqian Dong, ; Houbin Zhang,
| | - Houbin Zhang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
- *Correspondence: Shuqian Dong, ; Houbin Zhang,
| |
Collapse
|
16
|
Karamali F, Behtaj S, Babaei-Abraki S, Hadady H, Atefi A, Savoj S, Soroushzadeh S, Najafian S, Nasr Esfahani MH, Klassen H. Potential therapeutic strategies for photoreceptor degeneration: the path to restore vision. J Transl Med 2022; 20:572. [PMID: 36476500 PMCID: PMC9727916 DOI: 10.1186/s12967-022-03738-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/29/2022] [Indexed: 12/12/2022] Open
Abstract
Photoreceptors (PRs), as the most abundant and light-sensing cells of the neuroretina, are responsible for converting light into electrical signals that can be interpreted by the brain. PR degeneration, including morphological and functional impairment of these cells, causes significant diminution of the retina's ability to detect light, with consequent loss of vision. Recent findings in ocular regenerative medicine have opened promising avenues to apply neuroprotective therapy, gene therapy, cell replacement therapy, and visual prostheses to the challenge of restoring vision. However, successful visual restoration in the clinical setting requires application of these therapeutic approaches at the appropriate stage of the retinal degeneration. In this review, firstly, we discuss the mechanisms of PR degeneration by focusing on the molecular mechanisms underlying cell death. Subsequently, innovations, recent developments, and promising treatments based on the stage of disorder progression are further explored. Then, the challenges to be addressed before implementation of these therapies in clinical practice are considered. Finally, potential solutions to overcome the current limitations of this growing research area are suggested. Overall, the majority of current treatment modalities are still at an early stage of development and require extensive additional studies, both pre-clinical and clinical, before full restoration of visual function in PR degeneration diseases can be realized.
Collapse
Affiliation(s)
- Fereshteh Karamali
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sanaz Behtaj
- grid.1022.10000 0004 0437 5432Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia ,grid.1022.10000 0004 0437 5432Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222 Australia
| | - Shahnaz Babaei-Abraki
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hanieh Hadady
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Soraya Savoj
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sareh Soroushzadeh
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Najafian
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Henry Klassen
- grid.266093.80000 0001 0668 7243Gavin Herbert Eye Institute, Irvine, CA USA
| |
Collapse
|
17
|
Martínez-Gil N, Kutsyr O, Noailles A, Fernández-Sánchez L, Vidal L, Sánchez-Sáez X, Sánchez-Castillo C, Lax P, Cuenca N, García AG, Maneu V. Purinergic Receptors P2X7 and P2X4 as Markers of Disease Progression in the rd10 Mouse Model of Inherited Retinal Dystrophy. Int J Mol Sci 2022; 23:ijms232314758. [PMID: 36499084 PMCID: PMC9739106 DOI: 10.3390/ijms232314758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
The purinergic receptor P2X7 (P2X7R) is implicated in all neurodegenerative diseases of the central nervous system. It is also involved in the retinal degeneration associated with glaucoma, age-related macular degeneration, and diabetic retinopathy, and its overexpression in the retina is evident in these disorders. Retinitis pigmentosa is a progressive degenerative disease that ultimately leads to blindness. Here, we investigated the expression of P2X7R during disease progression in the rd10 mouse model of RP. As the purinergic receptor P2X4 is widely co-expressed with P2X7R, we also studied its expression in the retina of rd10 mice. The expression of P2X7R and P2X4R was examined by immunohistochemistry, flow cytometry, and western blotting. In addition, we analyzed retinal functionality by electroretinographic recordings of visual responses and optomotor tests and retinal morphology. We found that the expression of P2X7R and P2X4R increased in rd10 mice concomitant with disease progression, but with different cellular localization. Our findings suggest that P2X7R and P2X4R might play an important role in RP progression, which should be further analyzed for the pharmacological treatment of inherited retinal dystrophies.
Collapse
Affiliation(s)
- Natalia Martínez-Gil
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 Alicante, Spain
| | - Oksana Kutsyr
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, 03690 Alicante, Spain
| | - Agustina Noailles
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 Alicante, Spain
| | - Laura Fernández-Sánchez
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, 03690 Alicante, Spain
| | - Lorena Vidal
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 Alicante, Spain
| | - Xavier Sánchez-Sáez
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 Alicante, Spain
| | - Carla Sánchez-Castillo
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 Alicante, Spain
| | - Pedro Lax
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 Alicante, Spain
| | - Nicolás Cuenca
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 Alicante, Spain
| | - Antonio G. García
- Departamento de Farmacología y Terapéutica, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Victoria Maneu
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, 03690 Alicante, Spain
- Correspondence:
| |
Collapse
|
18
|
Palevski D, Ben-David G, Weinberger Y, Haj Daood R, Fernández JA, Budnik I, Levy-Mendelovich S, Kenet G, Nisgav Y, Weinberger D, Griffin JH, Livnat T. 3K3A-Activated Protein C Prevents Microglia Activation, Inhibits NLRP3 Inflammasome and Limits Ocular Inflammation. Int J Mol Sci 2022; 23:ijms232214196. [PMID: 36430674 PMCID: PMC9694680 DOI: 10.3390/ijms232214196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with pleiotropic cytoprotective properties albeit without the bleeding risks. The anti-inflammatory activities of 3K3A-APC were demonstrated in multiple preclinical injury models, including various neurological disorders. We determined the ability of 3K3A-APC to inhibit ocular inflammation in a murine model of lipopolysaccharide (LPS)-induced uveitis. Leukocyte recruitment, microglia activation, NLRP3 inflammasome and IL-1β levels were assessed using flow cytometry, retinal cryosection histology, retinal flatmount immunohistochemistry and vascular imaging, with and without 3K3A-APC treatment. LPS triggered robust inflammatory cell recruitment in the posterior chamber. The 3K3A-APC treatment significantly decreased leukocyte numbers and inhibited leukocyte extravasation from blood vessels into the retinal parenchyma to a level similar to controls. Resident microglia, which underwent an inflammatory transition following LPS injection, remained quiescent in eyes treated with 3K3A-APC. An inflammation-associated increase in retinal thickness, observed in LPS-injected eyes, was diminished by 3K3A-APC treatment, suggesting its clinical relevancy. Finally, 3K3A-APC treatment inhibited inflammasome activation, determined by lower levels of NLRP3 and its downstream effector IL-1β. Our results highlight the anti-inflammatory properties of 3K3A-APC in ocular inflammation and suggest its potential use as a novel treatment for retinal diseases associated with inflammation.
Collapse
Affiliation(s)
- Dahlia Palevski
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Gil Ben-David
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Yehonatan Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Rabeei Haj Daood
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - José A. Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ivan Budnik
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Sarina Levy-Mendelovich
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Gili Kenet
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Yael Nisgav
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
| | - Dov Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tami Livnat
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
- Correspondence:
| |
Collapse
|
19
|
Effect of Tauroursodeoxycholic Acid on Inflammation after Ocular Alkali Burn. Int J Mol Sci 2022; 23:ijms231911717. [PMID: 36233018 PMCID: PMC9570278 DOI: 10.3390/ijms231911717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022] Open
Abstract
Inflammation is the main cause of corneal and retinal damage in an ocular alkali burn (OAB). The aim of this study was to investigate the effect of tauroursodeoxycholic acid (TUDCA) on ocular inflammation in a mouse model of an OAB. An OAB was induced in C57BL/6j mouse corneas by using 1 M NaOH. TUDCA (400 mg/kg) or PBS was injected intraperitoneally (IP) once a day for 3 days prior to establishing the OAB model. A single injection of Infliximab (6.25 mg/kg) was administered IP immediately after the OAB. The TUDCA suppressed the infiltration of the CD45-positive cells and decreased the mRNA and protein levels of the upregulated TNF-α and IL-1β in the cornea and retina of the OAB. Furthermore, the TUDCA treatment inhibited the retinal glial activation after an OAB. The TUDCA treatment not only ameliorated CNV and promoted corneal re-epithelization but also attenuated the RGC apoptosis and preserved the retinal structure after the OAB. Finally, the TUDCA reduced the expression of the endoplasmic reticulum (ER) stress molecules, IRE1, GRP78 and CHOP, in the retinal tissues of the OAB mice. The present study demonstrated that the TUDCA inhibits ocular inflammation and protects the cornea and retina from injury in an OAB mouse model. These results provide a potential therapeutic intervention for the treatment of an OAB.
Collapse
|
20
|
Martínez-Gil N, Maneu V, Kutsyr O, Fernández-Sánchez L, Sánchez-Sáez X, Sánchez-Castillo C, Campello L, Lax P, Pinilla I, Cuenca N. Cellular and molecular alterations in neurons and glial cells in inherited retinal degeneration. Front Neuroanat 2022; 16:984052. [PMID: 36225228 PMCID: PMC9548552 DOI: 10.3389/fnana.2022.984052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple gene mutations have been associated with inherited retinal dystrophies (IRDs). Despite the spectrum of phenotypes caused by the distinct mutations, IRDs display common physiopathology features. Cell death is accompanied by inflammation and oxidative stress. The vertebrate retina has several attributes that make this tissue vulnerable to oxidative and nitrosative imbalance. The high energy demands and active metabolism in retinal cells, as well as their continuous exposure to high oxygen levels and light-induced stress, reveal the importance of tightly regulated homeostatic processes to maintain retinal function, which are compromised in pathological conditions. In addition, the subsequent microglial activation and gliosis, which triggers the secretion of pro-inflammatory cytokines, chemokines, trophic factors, and other molecules, further worsen the degenerative process. As the disease evolves, retinal cells change their morphology and function. In disease stages where photoreceptors are lost, the remaining neurons of the retina to preserve their function seek out for new synaptic partners, which leads to a cascade of morphological alterations in retinal cells that results in a complete remodeling of the tissue. In this review, we describe important molecular and morphological changes in retinal cells that occur in response to oxidative stress and the inflammatory processes underlying IRDs.
Collapse
Affiliation(s)
- Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Isabel Pinilla
- Aragón Institute for Health Research (IIS Aragón), Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa University Hospital, Zaragoza, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, Spain
- Isabel Pinilla,
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
- *Correspondence: Nicolás Cuenca,
| |
Collapse
|
21
|
Kumari A, Ayala-Ramirez R, Zenteno JC, Huffman K, Sasik R, Ayyagari R, Borooah S. Single cell RNA sequencing confirms retinal microglia activation associated with early onset retinal degeneration. Sci Rep 2022; 12:15273. [PMID: 36088481 PMCID: PMC9464204 DOI: 10.1038/s41598-022-19351-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Mutations in the Membrane-type frizzled related protein (Mfrp) gene results in an early-onset retinal degeneration associated with retinitis pigmentosa, microphthalmia, optic disc drusen and foveal schisis. In the current study, a previously characterized mouse model of human retinal degeneration carrying homozygous c.498_499insC mutations in Mfrp (MfrpKI/KI) was used. Patients carrying this mutation have retinal degeneration at an early age. The model demonstrates subretinal deposits and develops early-onset photoreceptor degeneration. We observed large subretinal deposits in MfrpKI/KI mice which were strongly CD68 positive and co-localized with autofluorescent spots. Single cell RNA sequencing of MfrpKI/KI mice retinal microglia showed a significantly higher number of pan-macrophage marker Iba-1 and F4/80 positive cells with increased expression of activation marker (CD68) and lowered microglial homeostatic markers (TMEM119, P2ry13, P2ry13, Siglech) compared with wild type mice confirming microglial activation as observed in retinal immunostaining showing microglia activation in subretinal region. Trajectory analysis identified a small cluster of microglial cells with activation transcriptomic signatures that could represent a subretinal microglia population in MfrpKI/KI mice expressing higher levels of APOE. We validated these findings using immunofluorescence staining of retinal cryosections and found a significantly higher number of subretinal Iba-1/ApoE positive microglia in MfrpKI/KI mice with some subretinal microglia also expressing lowered levels of microglial homeostatic marker TMEM119, confirming microglial origin. In summary, we confirm that MfrpKI/KI mice carrying the c.498_499insC mutation had a significantly higher population of activated microglia in their retina with distinct subsets of subretinal microglia. Further, studies are required to confirm whether the association of increased subretinal microglia in MfrpKI/KI mice are causal in degeneration.
Collapse
Affiliation(s)
- Asha Kumari
- Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA
| | - Raul Ayala-Ramirez
- Department of Biochemistry, Faculty of Medicine, UNAM, Mexico City, Mexico
- Department of Genetics, Conde de Valenciana, Institute of Ophthalmology, Mexico City, Mexico
| | - Juan Carlos Zenteno
- Department of Biochemistry, Faculty of Medicine, UNAM, Mexico City, Mexico
- Department of Genetics, Conde de Valenciana, Institute of Ophthalmology, Mexico City, Mexico
| | - Kristyn Huffman
- Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA
| | - Roman Sasik
- School of Medicine, Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA, USA
| | - Radha Ayyagari
- Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA.
| | - Shyamanga Borooah
- Shiley Eye Institute, University of California, San Diego, 9415 Campus Point Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
22
|
Yao J, Wang T, Jia L, Qiu Y, Zacks DN. Loss of Fas Receptor Function Preserves Photoreceptor Structure and Function in Two Mouse Models of Inherited Retinal Degeneration. Invest Ophthalmol Vis Sci 2022; 63:5. [PMID: 36083588 PMCID: PMC9469031 DOI: 10.1167/iovs.63.10.5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The genetic heterogeneity of inherited retinal degeneration (IRD) has limited the development of mutation-specific therapies, necessitating the development of therapeutic approaches targeting broadly shared pathophysiologic pathways. The Fas receptor has been reported as a contributor to retinal cell death and inflammation in a wide variety of ocular diseases. The purpose of this study was to assess targeting the Fas pathway as a novel mutation-independent approach to improve photoreceptor survival in IRD. Methods We examined the effects of genetic inactivation of the Fas receptor on retinal degeneration in two distinct IRD mouse models, P23H and rd10. The Fas-lpr mouse, which contains a functionally inactive Fas receptor, was crossed with the P23H and rd10 mice to generate P23H/Fas-lpr and rd10/Fas-lpr mice. Fas activation, photoreceptor survival and retinal function were assessed. Results We detected elevated levels of Fas receptor and microglial activation in the retinas of both P23H and rd10 mice. Inactivation of Fas in these two IRD models (P23H/Fas-lpr and rd10/Fas-lpr mice) resulted in reduced cell death, increased photoreceptor survival, improved retinal function, and reduced microglial activation and inflammatory cytokine production. Conclusions The protective effect of a nonfunctional Fas receptor in two different mouse models of retinal degeneration suggests that whereas the individual IRD mutation may be specific, the retina's response to the different stressors appears to be shared and driven by Fas. Reducing Fas activity might represent a potential mutation-independent therapeutic approach to preserve retinal structure and function in patients with IRD.
Collapse
Affiliation(s)
- Jingyu Yao
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Tiantian Wang
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States.,Department of Ophthalmology, Xiangya School of Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Jia
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Yaoyan Qiu
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States.,Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - David N Zacks
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| |
Collapse
|
23
|
Fehér J, Élő Á, István L, Nagy ZZ, Radák Z, Scuderi G, Artico M, Kovács I. Microbiota mitochondria disorders as hubs for early age-related macular degeneration. GeroScience 2022; 44:2623-2653. [PMID: 35978068 PMCID: PMC9385247 DOI: 10.1007/s11357-022-00620-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/01/2022] [Indexed: 01/07/2023] Open
Abstract
Age-related macular degeneration (AMD) is a progressive neurodegenerative disease affecting the central area (macula lutea) of the retina. Research on the pathogenic mechanism of AMD showed complex cellular contribution governed by such risk factors as aging, genetic predisposition, diet, and lifestyle. Recent studies suggested that microbiota is a transducer and a modifier of risk factors for neurodegenerative diseases, and mitochondria may be one of the intracellular targets of microbial signaling molecules. This review explores studies supporting a new concept on the contribution of microbiota-mitochondria disorders to AMD. We discuss metabolic, vascular, immune, and neuronal mechanism in AMD as well as key alterations of photoreceptor cells, retinal pigment epithelium (RPE), Bruch's membrane, choriocapillaris endothelial, immune, and neuronal cells. Special attention was paid to alterations of mitochondria contact sites (MCSs), an organelle network of mitochondria, endoplasmic reticulum, lipid droplets (LDs), and peroxisomes being documented based on our own electron microscopic findings from surgically removed human eyes. Morphometry of Bruch's membrane lipids and proteoglycans has also been performed in early AMD and aged controls. Microbial metabolites (short-chain fatty acids, polyphenols, and secondary bile acids) and microbial compounds (lipopolysaccharide, peptidoglycan, and bacterial DNA)-now called postbiotics-in addition to local effects on resident microbiota and mucous membrane, regulate systemic metabolic, vascular, immune, and neuronal mechanisms in normal conditions and in various common diseases. We also discuss their antioxidant, anti-inflammatory, and metabolic effects as well as experimental and clinical observations on regulating the main processes of photoreceptor renewal, mitophagy, and autophagy in early AMD. These findings support an emerging concept that microbiota-mitochondria disorders may be a crucial pathogenic mechanism of early AMD; and similarly, to other age-related neurodegenerative diseases, new treatment approaches should be targeted at these disorders.
Collapse
Affiliation(s)
- János Fehér
- PRIMAVERA Program, Nutripharma Hungaria Ltd., Budapest, Hungary
| | - Ágnes Élő
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Lilla István
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Zoltán Zsolt Nagy
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Zsolt Radák
- grid.472475.70000 0000 9243 1481Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Gianluca Scuderi
- grid.7841.aOphthalmology Unit, NESMOS Department, Sant’Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Marco Artico
- grid.417007.5Department of Sensory Organs, “Sapienza” University of Rome, Roma, Italy
| | - Illés Kovács
- grid.11804.3c0000 0001 0942 9821Department of Ophthalmology, Semmelweis University, Budapest, Hungary ,grid.5386.8000000041936877XDepartment of Ophthalmology, Weill Cornell Medical College, New York City, NY USA
| |
Collapse
|
24
|
Fernández-Sánchez L, Albertos-Arranz H, Ortuño-Lizarán I, Lax P, Cuenca N. Neuroprotective Effects of Tauroursodeoxicholic Acid Involves Vascular and Glial Changes in Retinitis Pigmentosa Model. Front Neuroanat 2022; 16:858073. [PMID: 35493706 PMCID: PMC9039202 DOI: 10.3389/fnana.2022.858073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/10/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose Retinitis pigmentosa is primarily characterized by a massive photoreceptor loss. But a global retinal remodeling occurs in later stages of the disease. At that phase, glial cells and retinal vasculature are also strongly affected. The main aim of the present work is to assess if the bile acid Tauroursodeoxicholic acid (TUDCA), which has a demonstrated neuroprotective effect in numerous neurodegenerative diseases, is able to prevent glial and vascular degeneration in the P23H rat retina. Methods Homozygous P23H (line 3) animals were injected weekly with a TUDCA (500 mg/kg, i.p.) or vehicle solution, from the postnatal day (P) 21 to P120. Sprague-Dawley rats (SD) were used as control. Retinal cross-sections and wholemounts were immunostained using different glial and vascular markers and visualized with confocal microscopy. Retinal blood vessels were stained with nicotinamide adenine dinucleotide phosphate (NADPH) diaphorase histochemistry and retinal vascular networks were drawn by hand using a camera lucida. Results At P120, the photoreceptor degeneration observed in P23H rats was accompanied by a reduction in the vascular network density and complexity at the deep capillary plexus. In addition, astrocytes showed gliotic features and the outer processes of Müller cells displayed an aberrant distribution in ring-shaped structures. When treated with TUDCA, P23H rats displayed better-preserved vessels and capillary loops in the deep capillary plexus which are associated with the partial preservation of photoreceptors. TUDCA treatment also increased the number of astrocytes and reduced the presence of Müller cell process clusters in the outer retina. Conclusion This work suggests that, besides its neuroprotective effect on photoreceptor cells, TUDCA treatment also protects from vascular and glial degeneration, a fact that encourages the use of TUDCA as a powerful therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Henar Albertos-Arranz
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Isabel Ortuño-Lizarán
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante, Spain
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL-FISABIO Foundation), Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
- *Correspondence: Nicolás Cuenca,
| |
Collapse
|
25
|
Bianchi M, De Salvo A, Asplund M, Carli S, Di Lauro M, Schulze‐Bonhage A, Stieglitz T, Fadiga L, Biscarini F. Poly(3,4-ethylenedioxythiophene)-Based Neural Interfaces for Recording and Stimulation: Fundamental Aspects and In Vivo Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104701. [PMID: 35191224 PMCID: PMC9036021 DOI: 10.1002/advs.202104701] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/04/2022] [Indexed: 05/29/2023]
Abstract
Next-generation neural interfaces for bidirectional communication with the central nervous system aim to achieve the intimate integration with the neural tissue with minimal neuroinflammatory response, high spatio-temporal resolution, very high sensitivity, and readout stability. The design and manufacturing of devices for low power/low noise neural recording and safe and energy-efficient stimulation that are, at the same time, conformable to the brain, with matched mechanical properties and biocompatibility, is a convergence area of research where neuroscientists, materials scientists, and nanotechnologists operate synergically. The biotic-abiotic neural interface, however, remains a formidable challenge that prompts for new materials platforms and innovation in device layouts. Conductive polymers (CP) are attractive materials to be interfaced with the neural tissue and to be used as sensing/stimulating electrodes because of their mixed ionic-electronic conductivity, their low contact impedance, high charge storage capacitance, chemical versatility, and biocompatibility. This manuscript reviews the state-of-the-art of poly(3,4-ethylenedioxythiophene)-based neural interfaces for extracellular recording and stimulation, focusing on those technological approaches that are successfully demonstrated in vivo. The aim is to highlight the most reliable and ready-for-clinical-use solutions, in terms of materials technology and recording performance, other than spot major limitations and identify future trends in this field.
Collapse
Affiliation(s)
- Michele Bianchi
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
| | - Anna De Salvo
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Sezione di FisiologiaUniversità di Ferraravia Fossato di Mortara 17Ferrara44121Italy
| | - Maria Asplund
- Division of Nursing and Medical TechnologyLuleå University of TechnologyLuleå971 87Sweden
- Department of Microsystems Engineering‐IMTEKUniversity of FreiburgFreiburg79110Germany
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburg79110Germany
| | - Stefano Carli
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Present address:
Department of Environmental and Prevention SciencesUniversità di FerraraFerrara44121Italy
| | - Michele Di Lauro
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
| | - Andreas Schulze‐Bonhage
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburg79110Germany
- Epilepsy CenterFaculty of MedicineUniversity of FreiburgFreiburg79110Germany
| | - Thomas Stieglitz
- Department of Microsystems Engineering‐IMTEKUniversity of FreiburgFreiburg79110Germany
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburg79110Germany
| | - Luciano Fadiga
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Sezione di FisiologiaUniversità di Ferraravia Fossato di Mortara 17Ferrara44121Italy
| | - Fabio Biscarini
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Life Science DepartmentUniversità di Modena e Reggio EmiliaVia Campi 103Modena41125Italy
| |
Collapse
|
26
|
Guo L, Choi S, Bikkannavar P, Cordeiro MF. Microglia: Key Players in Retinal Ageing and Neurodegeneration. Front Cell Neurosci 2022; 16:804782. [PMID: 35370560 PMCID: PMC8968040 DOI: 10.3389/fncel.2022.804782] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/11/2022] [Indexed: 12/20/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and play a key role in maintaining the normal function of the retina and brain. During early development, microglia migrate into the retina, transform into a highly ramified phenotype, and scan their environment constantly. Microglia can be activated by any homeostatic disturbance that may endanger neurons and threaten tissue integrity. Once activated, the young microglia exhibit a high diversity in their phenotypes as well as their functions, which relate to either beneficial or harmful consequences. Microglial activation is associated with the release of cytokines, chemokines, and growth factors that can determine pathological outcomes. As the professional phagocytes in the retina, microglia are responsible for the clearance of pathogens, dead cells, and protein aggregates. However, their phenotypic diversity and phagocytic capacity is compromised with ageing. This may result in the accumulation of protein aggregates and myelin debris leading to retinal neuroinflammation and neurodegeneration. In this review, we describe microglial phenotypes and functions in the context of the young and ageing retina, and the mechanisms underlying changes in ageing. Additionally, we review microglia-mediated retinal neuroinflammation and discuss the mechanisms of microglial involvement in retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Guo
- Institute of Ophthalmology, University College London, London, United Kingdom
- *Correspondence: Li Guo,
| | - Soyoung Choi
- Institute of Ophthalmology, University College London, London, United Kingdom
| | | | - M. Francesca Cordeiro
- Institute of Ophthalmology, University College London, London, United Kingdom
- Imperial College Ophthalmology Research Group, Imperial College London, London, United Kingdom
- M. Francesca Cordeiro,
| |
Collapse
|
27
|
Wang SK, Cepko CL. Targeting Microglia to Treat Degenerative Eye Diseases. Front Immunol 2022; 13:843558. [PMID: 35251042 PMCID: PMC8891158 DOI: 10.3389/fimmu.2022.843558] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
Microglia have been implicated in many degenerative eye disorders, including retinitis pigmentosa, age-related macular degeneration, glaucoma, diabetic retinopathy, uveitis, and retinal detachment. While the exact roles of microglia in these conditions are still being discovered, evidence from animal models suggests that they can modulate the course of disease. In this review, we highlight current strategies to target microglia in the eye and their potential as treatments for both rare and common ocular disorders. These approaches include depleting microglia with chemicals or radiation, reprogramming microglia using homeostatic signals or other small molecules, and inhibiting the downstream effects of microglia such as by blocking cytokine activity or phagocytosis. Finally, we describe areas of future research needed to fully exploit the therapeutic value of microglia in eye diseases.
Collapse
Affiliation(s)
- Sean K. Wang
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Constance L. Cepko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
- Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
- *Correspondence: Constance L. Cepko,
| |
Collapse
|
28
|
Kaur G, Singh NK. The Role of Inflammation in Retinal Neurodegeneration and Degenerative Diseases. Int J Mol Sci 2021; 23:ijms23010386. [PMID: 35008812 PMCID: PMC8745623 DOI: 10.3390/ijms23010386] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Retinal neurodegeneration is predominantly reported as the apoptosis or impaired function of the photoreceptors. Retinal degeneration is a major causative factor of irreversible vision loss leading to blindness. In recent years, retinal degenerative diseases have been investigated and many genes and genetic defects have been elucidated by many of the causative factors. An enormous amount of research has been performed to determine the pathogenesis of retinal degenerative conditions and to formulate the treatment modalities that are the critical requirements in this current scenario. Encouraging results have been obtained using gene therapy. We provide a narrative review of the various studies performed to date on the role of inflammation in human retinal degenerative diseases such as age-related macular degeneration, inherited retinal dystrophies, retinitis pigmentosa, Stargardt macular dystrophy, and Leber congenital amaurosis. In addition, we have highlighted the pivotal role of various inflammatory mechanisms in the progress of retinal degeneration. This review also offers an assessment of various therapeutic approaches, including gene-therapies and stem-cell-based therapies, for degenerative retinal diseases.
Collapse
Affiliation(s)
- Geetika Kaur
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Correspondence:
| |
Collapse
|
29
|
Loss of αA or αB-Crystallin Accelerates Photoreceptor Cell Death in a Mouse Model of P23H Autosomal Dominant Retinitis Pigmentosa. Int J Mol Sci 2021; 23:ijms23010070. [PMID: 35008496 PMCID: PMC8744961 DOI: 10.3390/ijms23010070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/08/2023] Open
Abstract
Inherited retinal degenerations (IRD) are a leading cause of visual impairment and can result from mutations in any one of a multitude of genes. Mutations in the light-sensing protein rhodopsin (RHO) is a leading cause of IRD with the most common of those being a missense mutation that results in substitution of proline-23 with histidine. This variant, also known as P23H-RHO, results in rhodopsin misfolding, initiation of endoplasmic reticulum stress, the unfolded protein response, and activation of cell death pathways. In this study, we investigate the effect of α-crystallins on photoreceptor survival in a mouse model of IRD secondary to P23H-RHO. We find that knockout of either αA- or αB-crystallin results in increased intraretinal inflammation, activation of apoptosis and necroptosis, and photoreceptor death. Our data suggest an important role for the ⍺-crystallins in regulating photoreceptor survival in the P23H-RHO mouse model of IRD.
Collapse
|
30
|
Bringer MA, Gabrielle PH, Bron AM, Creuzot-Garcher C, Acar N. The gut microbiota in retinal diseases. Exp Eye Res 2021; 214:108867. [PMID: 34856206 DOI: 10.1016/j.exer.2021.108867] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/25/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023]
Abstract
The gut microbiota is a complex ecosystem that inhabits the gastrointestinal tract and consists of archaea, fungi, viruses, and bacteria, with bacteria being dominant. From birth onwards, it coevolves dynamically together with the host. The composition of the gut microbiota is under the influence of a complex interplay between both host and environmental factors. Scientific advances in the past few decades have shown that it is essential in maintaining homeostasis and tipping the balance between health and disease. In addition to its role in food digestion, the gut microbiota is implicated in regulating multiple physiological processes in the host gut mucosa and in distant organs such as the brain. Persistent imbalance between gut microbial communities, termed "dysbiosis," has been associated with several inflammatory and metabolic diseases as well as with central nervous system disorders. In this review, we present the state of the art of current knowledge on an emerging concept, the microbiota-retina axis, and the potential role of its disturbance in the development of retinopathies. We also describe several microbiota-targeting strategies that could constitute preventive and therapeutic tools for retinopathies.
Collapse
Affiliation(s)
- Marie-Agnès Bringer
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, F-21000, Dijon, France.
| | - Pierre-Henry Gabrielle
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, F-21000, Dijon, France; Ophthalmology Department, University Hospital, F-21000, Dijon, France
| | - Alain M Bron
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, F-21000, Dijon, France; Ophthalmology Department, University Hospital, F-21000, Dijon, France
| | - Catherine Creuzot-Garcher
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, F-21000, Dijon, France; Ophthalmology Department, University Hospital, F-21000, Dijon, France
| | - Niyazi Acar
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, F-21000, Dijon, France
| |
Collapse
|
31
|
Rodrigo MJ, Garcia-Herranz D, Aragón-Navas A, Subias M, Martinez-Rincón T, Mendez-Martínez S, Cardiel MJ, García-Feijoo J, Ruberte J, Herrero-Vanrell R, Pablo L, Garcia-Martin E, Bravo-Osuna I. Long-term corticosteroid-induced chronic glaucoma model produced by intracameral injection of dexamethasone-loaded PLGA microspheres. Drug Deliv 2021; 28:2427-2446. [PMID: 34763590 PMCID: PMC8592597 DOI: 10.1080/10717544.2021.1998245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
PURPOSE To evaluate a new chronic glaucoma model produced by intracameral injection of dexamethasone-loaded poly lactic-co-glycolic acid microspheres (Dex-PLGA-Ms) over six months. METHODS Healthy rats received two injections (at baseline and Week 4) of Dex-PLGA-Ms into the anterior chamber of the right eye. Clinical signs and intraocular pressure (IOP) were weekly recorded. The structure of the retina and optic nerve was in vivo evaluated using optical coherence tomography (OCT) every two weeks and functionally using dark- and light-adapted electroretinography at 0-12-24 weeks. Histological studies were also performed. RESULTS IOP progressively increased up to hypertension (23.22 ± 3.63 mmHg) in both eyes but did so later in left eyes. OCT quantified a decrease in full-thickness retina posterior pole (R), retinal-nerve-fiber layer (RNFL), and ganglion-cell layer (GCL) thickness up to 24 weeks. Right eyes showed higher neuroretinal thickness loss up to week 8. RNFL experienced the highest percentage thickness loss at the inferior-superior axis, while in GCL the inner sectors of the horizontal axis (Nasal-Temporal) suffered the greatest decrease in thickness. Retinal ganglion cell, photoreceptor, and intermediate cell functionality decreased over time. Increased deposition of collagen IV was also found in zonular fibers and the ciliary body. CONCLUSIONS This work shows the usefulness of drug delivery systems, not to treat pathology but to induce it. Only two injections of Dex-PLGA-Ms in the anterior chamber of rat eyes were enough to progressively create ocular hypertension and subsequent functional and structural neuroretinal degeneration, at least over 6 months.
Collapse
Affiliation(s)
- M J Rodrigo
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain.,Miguel Servet Ophthalmology Research Group (GIMSO), Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain.,National Ocular Pathology Network (OFTARED), Carlos III Health Institute, Madrid, Spain
| | - D Garcia-Herranz
- Complutense University of Madrid. Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Spain.,Health Research Institute, San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - A Aragón-Navas
- Complutense University of Madrid. Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Spain.,Health Research Institute, San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - M Subias
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain.,Miguel Servet Ophthalmology Research Group (GIMSO), Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain
| | - T Martinez-Rincón
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain.,Miguel Servet Ophthalmology Research Group (GIMSO), Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain
| | - S Mendez-Martínez
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain.,Miguel Servet Ophthalmology Research Group (GIMSO), University of Zaragoza, Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain
| | - M J Cardiel
- Miguel Servet Ophthalmology Research Group (GIMSO), University of Zaragoza, Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain.,Department of Pathology, Lozano Blesa University Hospital, Zaragoza, Spain
| | - J García-Feijoo
- Complutense University of Madrid. Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, UCM 920415. National Ocular Pathology Network (OFTARED), Carlos III Health Institute, Spain.,Servicio de Oftalmología, Hospital Clínico San Carlos, Madrid, Spain.,Departamento de Inmunología, Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid (UCM), IdISSC, Madrid, Spain
| | - J Ruberte
- Animal Biotechnology and Gene Therapy Centre (CBATEG), Universitat Autònoma de Barcelona, Bellaterra, Spain.,Networked Biomedical Research Centre for Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain.,Department of Animal Health and Anatomy, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - R Herrero-Vanrell
- National Ocular Pathology Network (OFTARED), Carlos III Health Institute, Madrid, Spain.,Complutense University of Madrid. Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Spain.,Health Research Institute, San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - L Pablo
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain.,National Ocular Pathology Network (OFTARED), Carlos III Health Institute, Madrid, Spain.,Miguel Servet Ophthalmology Research Group (GIMSO), University of Zaragoza, Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain
| | - E Garcia-Martin
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain.,National Ocular Pathology Network (OFTARED), Carlos III Health Institute, Madrid, Spain.,Miguel Servet Ophthalmology Research Group (GIMSO), University of Zaragoza, Aragon Health Research Institute (IIS Aragon), Zaragoza, Spain
| | - I Bravo-Osuna
- National Ocular Pathology Network (OFTARED), Carlos III Health Institute, Madrid, Spain.,Complutense University of Madrid. Innovation, Therapy and Pharmaceutical Development in Ophthalmology (InnOftal) Research Group, UCM 920415, Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Spain.,Health Research Institute, San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| |
Collapse
|
32
|
Sun LF, Ma Y, Ji YY, Wu Z, Wang YH, Mou H, Jin ZB. Circular Rims2 Deficiency Causes Retinal Degeneration. Adv Biol (Weinh) 2021; 5:e2100906. [PMID: 34738746 DOI: 10.1002/adbi.202100906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/19/2021] [Indexed: 11/08/2022]
Abstract
Circular RNAs (circRNAs) refer to a newly recognized family of non-coding RNA with single-stranded RNAs. Despite emerging evidence indicating that circRNAs are abundantly expressed in various tissues, especially in the brain and retina, the role of circRNAs in retinal function and diseases is still largely unknown. Circular Rims2 (circRims2) is highly expressed and conserved in both the human and mouse brains. However, little is known about the expression and function of circRims2 in the retina. In the current study, the high-throughput RNA-seq analysis reveals a high expression of circRims2 in the retina. In addition, it is found that circRims2 is mainly located in plexiform layers that contain synapses between retinal neurons. Knocking down circRims2 with short hairpin RNA through subretinal adeno-associated viral (AAV) delivery in the mice leads to the decrease of the thickness of the outer and inner segment (OS/IS) layers and outer nuclear layer (ONL), and cessation of scotopic and photopic electroretinogram responses. Furthermore, the current study finds that circRims2 deficiency evokes retinal inflammation and activates the tumor necrosis factor (TNF) signaling pathway. Therefore, circRims2 may play an important role in the maintenance of retinal structure and function, and circRims2 deficiency may lead to pathogenic changes in the retina.
Collapse
Affiliation(s)
- Lan-Fang Sun
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yue Ma
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren HospitalCapital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Yang-Yang Ji
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhen Wu
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ya-Han Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren HospitalCapital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Hao Mou
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren HospitalCapital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren HospitalCapital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| |
Collapse
|
33
|
Van Bergen T, Hu TT, Little K, De Groef L, Moons L, Stitt AW, Vermassen E, Feyen JHM. Targeting Plasma Kallikrein With a Novel Bicyclic Peptide Inhibitor (THR-149) Reduces Retinal Thickening in a Diabetic Rat Model. Invest Ophthalmol Vis Sci 2021; 62:18. [PMID: 34677569 PMCID: PMC8556562 DOI: 10.1167/iovs.62.13.18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 08/27/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the effect of plasma kallikrein (PKal)-inhibition by THR-149 on preventing key pathologies associated with diabetic macular edema (DME) in a rat model. Methods Following streptozotocin-induced diabetes, THR-149 or its vehicle was administered in the rat via either a single intravitreal injection or three consecutive intravitreal injections (with a 1-week interval; both, 12.5 µg/eye). At 4 weeks post-diabetes, the effect of all groups was compared by histological analysis of Iba1-positive retinal inflammatory cells, inflammatory cytokines, vimentin-positive Müller cells, inwardly rectifying potassium and water homeostasis-related channels (Kir4.1 and AQP4, respectively), vascular leakage (fluorescein isothiocyanate-labeled bovine serum albumin), and retinal thickness. Results Single or repeated THR-149 injections resulted in reduced inflammation, as depicted by decreasing numbers and activation state of immune cells and IL-6 cytokine levels in the diabetic retina. The processes of reactive gliosis, vessel leakage, and retinal thickening were only significantly reduced after multiple THR-149 administrations. Individual retinal layer analysis showed that repeated THR-149 injections significantly decreased diabetes-induced thickening of the inner plexiform, inner nuclear, outer nuclear, and photoreceptor layers. At the glial-vascular interface, reduced Kir4.1-channel levels in the diabetic retina were restored to control non-diabetic levels in the presence of THR-149. In contrast, little or no effect of THR-149 was observed on the AQP4-channel levels. Conclusions These data demonstrate that repeated THR-149 administration reduces several DME-related key pathologies such as retinal thickening and neuropil disruption in the diabetic rat. These observations indicate that modulation of the PKal pathway using THR-149 has clinical potential to treat patients with DME.
Collapse
Affiliation(s)
| | | | - Karis Little
- Queen's University Belfast, Belfast, United Kingdom
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Alan W. Stitt
- Oxurion NV, Heverlee, Belgium
- Queen's University Belfast, Belfast, United Kingdom
| | | | | |
Collapse
|
34
|
Vernazza S, Oddone F, Tirendi S, Bassi AM. Risk Factors for Retinal Ganglion Cell Distress in Glaucoma and Neuroprotective Potential Intervention. Int J Mol Sci 2021; 22:7994. [PMID: 34360760 PMCID: PMC8346985 DOI: 10.3390/ijms22157994] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a population of neurons of the central nervous system (CNS) extending with their soma to the inner retina and with their axons to the optic nerve. Glaucoma represents a group of neurodegenerative diseases where the slow progressive death of RGCs results in a permanent loss of vision. To date, although Intra Ocular Pressure (IOP) is considered the main therapeutic target, the precise mechanisms by which RGCs die in glaucoma have not yet been clarified. In fact, Primary Open Angle Glaucoma (POAG), which is the most common glaucoma form, also occurs without elevated IOP. This present review provides a summary of some pathological conditions, i.e., axonal transport blockade, glutamate excitotoxicity and changes in pro-inflammatory cytokines along the RGC projection, all involved in the glaucoma cascade. Moreover, neuro-protective therapeutic approaches, which aim to improve RGC degeneration, have also been taken into consideration.
Collapse
Affiliation(s)
- Stefania Vernazza
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
| | | | - Sara Tirendi
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES), University of Genoa, 16126 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| |
Collapse
|
35
|
Fu J, Aung MH, Prunty MC, Hanif AM, Hutson LM, Boatright JH, Pardue MT. Tauroursodeoxycholic Acid Protects Retinal and Visual Function in a Mouse Model of Type 1 Diabetes. Pharmaceutics 2021; 13:1154. [PMID: 34452115 PMCID: PMC8400977 DOI: 10.3390/pharmaceutics13081154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Previous studies demonstrated that systemic treatment with tauroursodeoxycholic acid (TUDCA) is protective in in vivo mouse models of retinal degeneration and in culture models of hyperglycemia. This study tested the hypothesis that TUDCA will preserve visual and retinal function in a mouse model of early diabetic retinopathy (DR). METHODS Adult C57BL/6J mice were treated with streptozotocin (STZ) and made diabetic at 8-10 weeks of age. Control and diabetic mice were treated with vehicle or TUDCA starting 1 or 3 weeks after induction of diabetes, and were assessed bimonthly for visual function via an optomotor response and monthly for retinal function via scotopic electroretinograms. RESULTS Diabetic mice showed significantly reduced spatial frequency and contrast sensitivity thresholds compared to control mice, while diabetic mice treated early with TUDCA showed preservation at all timepoints. A-wave, b-wave, and oscillatory potential 2 (OP2) amplitudes decreased in diabetic mice. Diabetic mice also exhibited delays in a-wave and OP2-implicit times. Early TUDCA treatment ameliorated a-wave, b-wave, and OP2 deficits. Late TUDCA treatment showed reduced preservation effects compared to early treatment. CONCLUSIONS Early TUDCA treatment preserved visual function in an STZ-mouse model of Type I diabetes. These data add to a growing body of preclinical research that may support testing whether TUDCA may be an effective early clinical intervention against declining visual function caused by diabetic retinopathy.
Collapse
Affiliation(s)
- Jieming Fu
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA; (J.F.); (M.H.A.); (M.C.P.); (A.M.H.); (L.M.H.)
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22904, USA
| | - Moe H. Aung
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA; (J.F.); (M.H.A.); (M.C.P.); (A.M.H.); (L.M.H.)
- Neuroscience, Emory University, Atlanta, GA 30322, USA
- Department of Ophthalmology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Megan C. Prunty
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA; (J.F.); (M.H.A.); (M.C.P.); (A.M.H.); (L.M.H.)
- Case Western Reserve University School of Medicine, Urology Institute of University Hospitals, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Adam M. Hanif
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA; (J.F.); (M.H.A.); (M.C.P.); (A.M.H.); (L.M.H.)
- Casey Eye Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Lauren M. Hutson
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA; (J.F.); (M.H.A.); (M.C.P.); (A.M.H.); (L.M.H.)
- Department of Psychology and Neuroscience, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Jeffrey H. Boatright
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA; (J.F.); (M.H.A.); (M.C.P.); (A.M.H.); (L.M.H.)
- Neuroscience, Emory University, Atlanta, GA 30322, USA
- Ophthalmology, Emory University, Atlanta, GA 30322, USA
| | - Machelle T. Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA 30033, USA; (J.F.); (M.H.A.); (M.C.P.); (A.M.H.); (L.M.H.)
- Neuroscience, Emory University, Atlanta, GA 30322, USA
- Ophthalmology, Emory University, Atlanta, GA 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| |
Collapse
|
36
|
Riccitelli S, Di Paolo M, Ashley J, Bisti S, Di Marco S. The Timecourses of Functional, Morphological, and Molecular Changes Triggered by Light Exposure in Sprague-Dawley Rat Retinas. Cells 2021; 10:1561. [PMID: 34205615 PMCID: PMC8234029 DOI: 10.3390/cells10061561] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/01/2021] [Accepted: 06/16/2021] [Indexed: 01/11/2023] Open
Abstract
Retinal neurodegeneration can impair visual perception at different levels, involving not only photoreceptors, which are the most metabolically active cells, but also the inner retina. Compensatory mechanisms may hide the first signs of these impairments and reduce the likelihood of receiving timely treatments. Therefore, it is essential to characterize the early critical steps in the neurodegenerative progression to design adequate therapies. This paper describes and correlates early morphological and biochemical changes in the degenerating retina with in vivo functional analysis of retinal activity and investigates the progression of neurodegenerative stages for up to 7 months. For these purposes, Sprague-Dawley rats were exposed to 1000 lux light either for different durations (12 h to 24 h) and examined seven days afterward (7d) or for a fixed duration (24 h) and monitored at various time points following the exposure (up to 210d). Flash electroretinogram (fERG) recordings were correlated with morphological and histological analyses to evaluate outer and inner retinal disruptions, gliosis, trophic factor release, and microglial activation. Twelve hours or fifteen hours of exposure to constant light led to a severe retinal dysfunction with only minor morphological changes. Therefore, early pathological signs might be hidden by compensatory mechanisms that silence retinal dysfunction, accounting for the discrepancy between photoreceptor loss and retinal functional output. The long-term analysis showed a transient functional recovery, maximum at 45 days, despite a progressive loss of photoreceptors and coincident increases in glial fibrillary acidic protein (GFAP) and basic fibroblast growth factor-2 (bFGF-2) expression. Interestingly, the progression of the disease presented different patterns in the dorsal and ventral retina. The information acquired gives us the potential to develop a specific diagnostic tool to monitor the disease's progression and treatment efficacy.
Collapse
Affiliation(s)
- Serena Riccitelli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.R.); (M.D.P.); (S.B.)
| | - Mattia Di Paolo
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.R.); (M.D.P.); (S.B.)
| | - James Ashley
- School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK;
| | - Silvia Bisti
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.R.); (M.D.P.); (S.B.)
- Istituto Nazionale di Biostrutture e Biosistemi (INBB), 00136 Roma, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
| | - Stefano Di Marco
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (S.R.); (M.D.P.); (S.B.)
- Istituto Nazionale di Biostrutture e Biosistemi (INBB), 00136 Roma, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS, Ospedale Policlinico San Martino, 16132 Genova, Italy
| |
Collapse
|
37
|
Ruiz-Pastor MJ, Kutsyr O, Lax P, Cuenca N. Decrease in DHA and other fatty acids correlates with photoreceptor degeneration in retinitis pigmentosa. Exp Eye Res 2021; 209:108667. [PMID: 34119484 DOI: 10.1016/j.exer.2021.108667] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/21/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022]
Abstract
Fatty acids, and especially docosahexaenoic acid (DHA), are essential for photoreceptor cell integrity and are involved in the phototransduction cascade. In this study, we analyzed the changes in the fatty acid profile in the retina of the rd10 mouse, model of retinitis pigmentosa, in order to identify potential risk factors for retinal degeneration and possible therapeutic approaches. Fatty acids from C57BL/6J and rd10 mouse retinas were extracted with Folch's method and analyzed by gas chromatography/mass spectrometry. Changes in retinal morphology were evaluated by immunohistochemistry. The rd10 mouse retina showed a decreased number of photoreceptor rows and alterations in photoreceptor morphology compared to C57BL/6J mice. The total amount of fatty acids dropped by 29.4% in the dystrophic retinas compared to C57BL/6J retinas. A positive correlation was found between the retinal content of specific fatty acids and the number of photoreceptor rows. We found that the amount of several short-chain and long-chain saturated fatty acids, as well as monounsaturated fatty acids, decreased in the retina of rd10 mice. Moreover, the content of the n-6 polyunsaturated fatty acid arachidonic acid and the n-3 polyunsaturated DHA decreased markedly in the dystrophic retina. The fall of DHA was more pronounced, hence the n-6/n-3 ratio was significantly increased in the diseased retina. The content of specific fatty acids in the retina decreased with photoreceptor degeneration in retinitis pigmentosa mice, with a remarkable reduction in DHA and other saturated and unsaturated fatty acids. These fatty acids could be essential for photoreceptor cell viability, and they should be evaluated for the design of therapeutical strategies and nutritional supplements.
Collapse
Affiliation(s)
- María José Ruiz-Pastor
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain.
| | - Nicolás Cuenca
- Department of Physiology, Genetics, and Microbiology, University of Alicante, Alicante, Spain.
| |
Collapse
|
38
|
Kutsyr O, Maestre-Carballa L, Lluesma-Gomez M, Martinez-Garcia M, Cuenca N, Lax P. Retinitis pigmentosa is associated with shifts in the gut microbiome. Sci Rep 2021; 11:6692. [PMID: 33758301 PMCID: PMC7988170 DOI: 10.1038/s41598-021-86052-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome is known to influence the pathogenesis and progression of neurodegenerative diseases. However, there has been relatively little focus upon the implications of the gut microbiome in retinal diseases such as retinitis pigmentosa (RP). Here, we investigated changes in gut microbiome composition linked to RP, by assessing both retinal degeneration and gut microbiome in the rd10 mouse model of RP as compared to control C57BL/6J mice. In rd10 mice, retinal responsiveness to flashlight stimuli and visual acuity were deteriorated with respect to observed in age-matched control mice. This functional decline in dystrophic animals was accompanied by photoreceptor loss, morphologic anomalies in photoreceptor cells and retinal reactive gliosis. Furthermore, 16S rRNA gene amplicon sequencing data showed a microbial gut dysbiosis with differences in alpha and beta diversity at the genera, species and amplicon sequence variants (ASV) levels between dystrophic and control mice. Remarkably, four fairly common ASV in healthy gut microbiome belonging to Rikenella spp., Muribaculaceace spp., Prevotellaceae UCG-001 spp., and Bacilli spp. were absent in the gut microbiome of retinal disease mice, while Bacteroides caecimuris was significantly enriched in mice with RP. The results indicate that retinal degenerative changes in RP are linked to relevant gut microbiome changes. The findings suggest that microbiome shifting could be considered as potential biomarker and therapeutic target for retinal degenerative diseases.
Collapse
Affiliation(s)
- Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Lucía Maestre-Carballa
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Mónica Lluesma-Gomez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Manuel Martinez-Garcia
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain.
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain.
| |
Collapse
|
39
|
Retinal Inflammation, Cell Death and Inherited Retinal Dystrophies. Int J Mol Sci 2021; 22:ijms22042096. [PMID: 33672611 PMCID: PMC7924201 DOI: 10.3390/ijms22042096] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a group of retinal disorders that cause progressive and severe loss of vision because of retinal cell death, mainly photoreceptor cells. IRDs include retinitis pigmentosa (RP), the most common IRD. IRDs present a genetic and clinical heterogeneity that makes it difficult to achieve proper treatment. The progression of IRDs is influenced, among other factors, by the activation of the immune cells (microglia, macrophages, etc.) and the release of inflammatory molecules such as chemokines and cytokines. Upregulation of tumor necrosis factor alpha (TNFα), a pro-inflammatory cytokine, is found in IRDs. This cytokine may influence photoreceptor cell death. Different cell death mechanisms are proposed, including apoptosis, necroptosis, pyroptosis, autophagy, excessive activation of calpains, or parthanatos for photoreceptor cell death. Some of these cell death mechanisms are linked to TNFα upregulation and inflammation. Therapeutic approaches that reduce retinal inflammation have emerged as useful therapies for slowing down the progression of IRDs. We focused this review on the relationship between retinal inflammation and the different cell death mechanisms involved in RP. We also reviewed the main anti-inflammatory therapies for the treatment of IRDs.
Collapse
|
40
|
Aggio-Bruce R, Chu-Tan JA, Wooff Y, Cioanca AV, Schumann U, Natoli R. Inhibition of microRNA-155 Protects Retinal Function Through Attenuation of Inflammation in Retinal Degeneration. Mol Neurobiol 2021; 58:835-854. [PMID: 33037565 PMCID: PMC7843561 DOI: 10.1007/s12035-020-02158-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/01/2020] [Indexed: 01/14/2023]
Abstract
Although extensively investigated in inflammatory conditions, the role of pro-inflammatory microRNAs (miRNAs), miR-155 and miR-146a, has not been well-studied in retinal degenerative diseases. We therefore aimed to explore the role and regulation of these miRNA in the degenerating retina, with a focus on miR-155. C57BL/6J mice were subjected to photo-oxidative damage for up to 5 days to induce focal retinal degeneration. MiR-155 expression was quantified by qRT-PCR in whole retina, serum, and small-medium extracellular vesicles (s-mEVs), and a PrimeFlow™ assay was used to identify localisation of miR-155 in retinal cells. Constitutive miR-155 knockout (KO) mice and miR-155 and miR-146a inhibitors were utilised to determine the role of these miRNA in the degenerating retina. Electroretinography was employed as a measure of retinal function, while histological quantification of TUNEL+ and IBA1+ positive cells was used to quantify photoreceptor cell death and infiltrating immune cells, respectively. Upregulation of miR-155 was detected in retinal tissue, serum and s-mEVs in response to photo-oxidative damage, localising to the nucleus of a subset of retinal ganglion cells and glial cells and in the cytoplasm of photoreceptors. Inhibition of miR-155 showed increased function from negative controls and a less pathological pattern of IBA1+ cell localisation and morphology at 5 days photo-oxidative damage. While neither dim-reared nor damaged miR-155 KO animals showed retinal histological difference from controls, following photo-oxidative damage, miR-155 KO mice showed increased a-wave relative to controls. We therefore consider miR-155 to be associated with the inflammatory response of the retina in response to photoreceptor-specific degeneration.
Collapse
Affiliation(s)
- Riemke Aggio-Bruce
- The John Curtin School of Medical Research, The Australian National University, Garran Road, Acton, Australian Capital Territory, 2601, Australia
- The Australian National University Medical School, Mills Road, Australian Capital Territory, Acton, 2601, Australia
| | - Joshua A Chu-Tan
- The John Curtin School of Medical Research, The Australian National University, Garran Road, Acton, Australian Capital Territory, 2601, Australia
- The Australian National University Medical School, Mills Road, Australian Capital Territory, Acton, 2601, Australia
| | - Yvette Wooff
- The John Curtin School of Medical Research, The Australian National University, Garran Road, Acton, Australian Capital Territory, 2601, Australia
- The Australian National University Medical School, Mills Road, Australian Capital Territory, Acton, 2601, Australia
| | - Adrian V Cioanca
- The John Curtin School of Medical Research, The Australian National University, Garran Road, Acton, Australian Capital Territory, 2601, Australia
| | - Ulrike Schumann
- The John Curtin School of Medical Research, The Australian National University, Garran Road, Acton, Australian Capital Territory, 2601, Australia
| | - Riccardo Natoli
- The John Curtin School of Medical Research, The Australian National University, Garran Road, Acton, Australian Capital Territory, 2601, Australia.
- The Australian National University Medical School, Mills Road, Australian Capital Territory, Acton, 2601, Australia.
| |
Collapse
|
41
|
Kutsyr O, Sánchez-Sáez X, Martínez-Gil N, de Juan E, Lax P, Maneu V, Cuenca N. Gradual Increase in Environmental Light Intensity Induces Oxidative Stress and Inflammation and Accelerates Retinal Neurodegeneration. Invest Ophthalmol Vis Sci 2021; 61:1. [PMID: 32744596 PMCID: PMC7441298 DOI: 10.1167/iovs.61.10.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) is a blinding neurodegenerative disease of the retina that can be affected by many factors. The present study aimed to analyze the effect of different environmental light intensities in rd10 mice retina. Methods C57BL/6J and rd10 mice were bred and housed under three different environmental light intensities: scotopic (5 lux), mesopic (50 lux), and photopic (300 lux). Visual function was studied using electroretinography and optomotor testing. The structural and morphological integrity of the retinas was evaluated by optical coherence tomography imaging and immunohistochemistry. Additionally, inflammatory processes and oxidative stress markers were analyzed by flow cytometry and western blotting. Results When the environmental light intensity was higher, retinal function decreased in rd10 mice and was accompanied by light-dependent photoreceptor loss, followed by morphological alterations, and synaptic connectivity loss. Moreover, light-dependent retinal degeneration was accompanied by an increased number of inflammatory cells, which became more activated and phagocytic, and by an exacerbated reactive gliosis. Furthermore, light-dependent increment in oxidative stress markers in rd10 mice retina pointed to a possible mechanism for light-induced photoreceptor degeneration. Conclusions An increase in rd10 mice housing light intensity accelerates retinal degeneration, activating cell death, oxidative stress pathways, and inflammatory cells. Lighting intensity is a key factor in the progression of retinal degeneration, and standardized lighting conditions are advisable for proper analysis and interpretation of experimental results from RP animal models, and specifically from rd10 mice. Also, it can be hypothesized that light protection could be an option to slow down retinal degeneration in some cases of RP.
Collapse
|
42
|
Sectoral activation of glia in an inducible mouse model of autosomal dominant retinitis pigmentosa. Sci Rep 2020; 10:16967. [PMID: 33046772 PMCID: PMC7552392 DOI: 10.1038/s41598-020-73749-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022] Open
Abstract
Retinitis pigmentosa (RP) is a group of blinding disorders caused by diverse mutations, including in rhodopsin (RHO). Effective therapies have yet to be discovered. The I307N Rho mouse is a light-inducible model of autosomal dominant RP. Our purpose was to describe the glial response in this mouse model to educate future experimentation. I307N Rho mice were exposed to 20,000 lx of light for thirty minutes to induce retinal degeneration. Immunofluorescence staining of cross-sections and flat-mounts was performed to visualize the response of microglia and Müller glia. Histology was correlated with spectral-domain optical coherence tomography imaging (SD-OCT). Microglia dendrites extended between photoreceptors within two hours of induction, withdrew their dendrites between twelve hours and one day, appeared ameboid by three days, and assumed a ramified morphology by one month. Glial activation was more robust in the inferior retina and modulated across the boundary of light damage. SD-OCT hyper-reflectivity overlapped with activated microglia. Finally, microglia transiently adhered to the RPE before which RPE cells appeared dysmorphic. Our data demonstrate the spatial and temporal pattern of glial activation in the I307N Rho mouse, and correlate these patterns with SD-OCT images, assisting in interpretation of SD-OCT images in preclinical models and in human RP.
Collapse
|
43
|
Bone Marrow-Derived Mononuclear Cell Transplants Decrease Retinal Gliosis in Two Animal Models of Inherited Photoreceptor Degeneration. Int J Mol Sci 2020; 21:ijms21197252. [PMID: 33008136 PMCID: PMC7583887 DOI: 10.3390/ijms21197252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Inherited photoreceptor degenerations are not treatable diseases and a frequent cause of blindness in working ages. In this study we investigate the safety, integration and possible rescue effects of intravitreal and subretinal transplantation of adult human bone-marrow-derived mononuclear stem cells (hBM-MSCs) in two animal models of inherited photoreceptor degeneration, the P23H-1 and the Royal College of Surgeons (RCS) rat. Immunosuppression was started one day before the injection and continued through the study. The hBM-MSCs were injected in the left eyes and the animals were processed 7, 15, 30 or 60 days later. The retinas were cross-sectioned, and L- and S- cones, microglia, astrocytes and Müller cells were immunodetected. Transplantations had no local adverse effects and the CD45+ cells remained for up to 15 days forming clusters in the vitreous and/or a 2–3-cells-thick layer in the subretinal space after intravitreal or subretinal injections, respectively. We did not observe increased photoreceptor survival nor decreased microglial cell numbers in the injected left eyes. However, the injected eyes showed decreased GFAP immunoreactivity. We conclude that intravitreal or subretinal injection of hBM-MSCs in dystrophic P23H-1 and RCS rats causes a decrease in retinal gliosis but does not have photoreceptor neuroprotective effects, at least in the short term. However, this treatment may have a potential therapeutic effect that merits further investigation.
Collapse
|
44
|
Tauroursodeoxycholic Acid Protects Retinal Pigment Epithelial Cells from Oxidative Injury and Endoplasmic Reticulum Stress In Vitro. Biomedicines 2020; 8:biomedicines8090367. [PMID: 32967221 PMCID: PMC7555559 DOI: 10.3390/biomedicines8090367] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal degeneration is characterized by the dysfunction of retinal cells. Oxidative and endoplasmic reticulum (ER) stress play an important role in the pathogenesis and progression of retinal degeneration. Tauroursodeoxycholic acid (TUDCA) has been demonstrated to have protective effects in in vitro and in vivo retinal degeneration models. To fully understand the molecular mechanisms of TUDCA’s protection, we first treated human retinal pigment epithelial (RPE) cells, ARPE-19, with H2O2 or H2O2 plus TUDCA for 24 h. RPE cells co-exposed to TUDCA had higher cell viability and lower cell death rate compared to cells exposed to H2O2 alone. TUDCA significantly increased antioxidant capacity in H2O2-treated RPE cells by decreasing the generation of reactive oxygen species (ROS) and Malondialdehyde (MDA), upregulating the expression of antioxidant genes, and increasing the generation of glutathione (GSH). TUDCA also inhibited inflammation in H2O2-challenged RPE cells by decreasing the expression of proinflammatory cytokines. Furthermore, TUDCA suppressed thapsigargin-induced ER stress in RPE cells, as demonstrated by decreased the expression of CCAAT-enhancer-binding protein homologous protein (CHOP) and apoptosis. Our present study suggests that TUDCA can protect RPE cells against oxidative damage, inflammation, and ER stress and may benefit patients with retinal degeneration.
Collapse
|
45
|
Integrin CD11b Deficiency Aggravates Retinal Microglial Activation and RGCs Degeneration After Acute Optic Nerve Injury. Neurochem Res 2020; 45:1072-1085. [PMID: 32052258 DOI: 10.1007/s11064-020-02984-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/23/2020] [Accepted: 02/08/2020] [Indexed: 12/13/2022]
Abstract
Neuroinflammation plays a vital role in the process of a variety of retinal ganglion cells (RGCs) degenerative diseases including traumatic optic neuropathy (TON). Retinal microglial activation is believed as a harbinger of TON, and robust microglial activation can aggravate trauma-induced RGCs degeneration, which ultimately leads to RGCs loss. Toll like receptor 4 (TLR4)-triggered inflammation is of great importance in retinal inflammatory response after optic nerve injury. CD11b on macrophage and brain microglia can inhibit TLR4-triggered inflammation. However, the functional role of CD11b in retinal microglia is not well understood. Here, using an optic nerve crush model and CD11b gene deficient mice, we found that CD11b protein expression was mainly on retinal microglia, significantly increased after optic nerve injury, and still maintained at a high level till at least 28 days post crush. Compared with wild type mice, following acute optic nerve injury, CD11b deficient retinae exhibited more exacerbated microglial activation, accelerated RGCs degeneration, less growth associated protein-43 expression, as well as more proinflammatory cytokines such as interleukin-6 and tumor necrosis factor α while less anti-inflammatory factors such as arginase-1 and interleukin-10 production. We conclude that CD11b is essential in regulating retinal microglial activation and neuroinflammatory responses after acute optic nerve injury, which is critical for subsequent RGCs degeneration and loss.
Collapse
|
46
|
Roche SL, Kutsyr O, Cuenca N, Cotter TG. Norgestrel, a Progesterone Analogue, Promotes Significant Long-Term Neuroprotection of Cone Photoreceptors in a Mouse Model of Retinal Disease. Invest Ophthalmol Vis Sci 2019; 60:3221-3235. [PMID: 31335948 DOI: 10.1167/iovs.19-27246] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) refers to a group of inherited blinding retinal diseases, whereby the death of mutated rod photoreceptors is followed closely by the death of cone photoreceptors. Cone cell death can be hugely debilitating as color/daytime vision becomes impaired. Thus, treatments that are effective against cone cell death are urgently needed. Our research has been working toward development of a neuroprotective treatment for RP. We have previously demonstrated significant neuroprotective properties of norgestrel, a progesterone analogue, in the mouse retina. The current study further investigates the potential of norgestrel as a treatment for RP, with a focus on long-term preservation of cone photoreceptors. Methods Using the well-established rd10 mouse model of RP, we administered a norgestrel-supplemented diet at postnatal day (P)30, following widespread loss of rod photoreceptors and at the outset of cone degeneration. We subsequently assessed cone cell morphology and retinal function at P50, P60, and P80, using immunohistochemistry, electroretinograph recordings, and optomotor testing. Results While cone cell degeneration was widespread in the untreated rd10 retina, we observed profound preservation of cone photoreceptor morphology in the norgestrel-treated mice for at least 50 days, out to P80. This was demonstrated by up to 28-fold more cone arrestin-positive photoreceptors. This protection transpired to functional preservation at all ages. Conclusions This work presents norgestrel as an incredibly promising long-term neuroprotective compound for the treatment of RP. Crucially, norgestrel could be used in the mid-late stages of the disease to protect remaining cone cells and help preserve color/daytime vision.
Collapse
Affiliation(s)
- Sarah L Roche
- Cell Development and Disease Laboratory, Biochemistry Department, Biosciences Institute, University College Cork, Cork, Ireland
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, San Vicente del Raspeig, Spain
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, San Vicente del Raspeig, Spain
| | - Thomas G Cotter
- Cell Development and Disease Laboratory, Biochemistry Department, Biosciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
47
|
Kusaczuk M. Tauroursodeoxycholate-Bile Acid with Chaperoning Activity: Molecular and Cellular Effects and Therapeutic Perspectives. Cells 2019; 8:E1471. [PMID: 31757001 PMCID: PMC6952947 DOI: 10.3390/cells8121471] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/15/2019] [Accepted: 11/19/2019] [Indexed: 12/11/2022] Open
Abstract
Tauroursodeoxycholic acid (TUDCA) is a naturally occurring hydrophilic bile acid that has been used for centuries in Chinese medicine. Chemically, TUDCA is a taurine conjugate of ursodeoxycholic acid (UDCA), which in contemporary pharmacology is approved by Food and Drug Administration (FDA) for treatment of primary biliary cholangitis. Interestingly, numerous recent studies demonstrate that mechanisms of TUDCA functioning extend beyond hepatobiliary disorders. Thus, TUDCA has been demonstrated to display potential therapeutic benefits in various models of many diseases such as diabetes, obesity, and neurodegenerative diseases, mostly due to its cytoprotective effect. The mechanisms underlying this cytoprotective activity have been mainly attributed to alleviation of endoplasmic reticulum (ER) stress and stabilization of the unfolded protein response (UPR), which contributed to naming TUDCA as a chemical chaperone. Apart from that, TUDCA has also been found to reduce oxidative stress, suppress apoptosis, and decrease inflammation in many in-vitro and in-vivo models of various diseases. The latest research suggests that TUDCA can also play a role as an epigenetic modulator and act as therapeutic agent in certain types of cancer. Nevertheless, despite the massive amount of evidence demonstrating positive effects of TUDCA in pre-clinical studies, there are certain limitations restraining its wide use in patients. Here, molecular and cellular modes of action of TUDCA are described and therapeutic opportunities and limitations of this bile acid are discussed.
Collapse
Affiliation(s)
- Magdalena Kusaczuk
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Mickiewicza 2A, 15-222 Białystok, Poland
| |
Collapse
|
48
|
Daruich A, Picard E, Boatright JH, Behar-Cohen F. Review: The bile acids urso- and tauroursodeoxycholic acid as neuroprotective therapies in retinal disease. Mol Vis 2019; 25:610-624. [PMID: 31700226 PMCID: PMC6817734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/12/2019] [Indexed: 10/28/2022] Open
Abstract
Bile acids are produced in the liver and excreted into the intestine, where their main function is to participate in lipid digestion. Ursodeoxycholic acid (UDCA) and tauroursodeoxycholic acid (TUDCA) have shown antiapoptotic, anti-inflammatory, and antioxidant effects in various models of neurodegenerative diseases. However, little is known about signaling pathways and molecular mechanisms through which these bile acids act as neuroprotectors, delaying translation to the clinical setting. We review evidence supporting a potentially therapeutic role for bile acids in retinal disorders, and the mechanisms and pathways involved in the cytoprotective effects of bile acids from the liver and the enterohepatic circulation to the central nervous system and the retina. As secondary bile acids are generated by the microbiota metabolism, bile acids might be a link between neurodegenerative retinal diseases and microbiota.
Collapse
Affiliation(s)
- Alejandra Daruich
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, Paris, France,Ophthalmology Department, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Emilie Picard
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, Paris, France
| | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA,Center of Excellence, Atlanta Veterans Administration Medical Center, Decatur, GA
| | - Francine Behar-Cohen
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, Paris, France,Ophtalmopole, Cochin Hospital, AP-HP, Assistance Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
49
|
Arnold E, Thébault S, Aroña RM, Martínez de la Escalera G, Clapp C. Prolactin mitigates deficiencies of retinal function associated with aging. Neurobiol Aging 2019; 85:38-48. [PMID: 31698287 DOI: 10.1016/j.neurobiolaging.2019.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 01/17/2023]
Abstract
Aging causes the progressive degeneration of retinal cells leading to the eventual loss of vision. The hormone prolactin (PRL) is a neurotrophic factor able to compensate for photoreceptor cell death and electroretinogram deficits induced by light retinal damage. Here, we used adult 4-month old and aged 20-month old pigmented mice, null or not for the PRL receptor to explore whether PRL provides trophic support against age-related retinal dysfunction. Retinal functionality, apoptosis, glia activation, and neurotrophin expression were assessed by electroretinogram, TUNEL, glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1 immunohistochemistry, and real-time PCR, respectively. Lack of PRL signaling in aged mice, but not in adult mice, correlated with photosensitive retinal dysfunction, increased photoreceptor apoptosis, differential expression of proapoptotic mediators, and microglia activation. We conclude that PRL is required for maintaining retinal functionality in both female and male mice during aging and has potential therapeutic value against age-related retinal disorders.
Collapse
Affiliation(s)
- Edith Arnold
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México; CONACYT-Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Stéphanie Thébault
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Rodrigo M Aroña
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | | | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México.
| |
Collapse
|
50
|
Chen TF, Chen KW, Chien Y, Lai YH, Hsieh ST, Ma HY, Wang KC, Shiau CY. Dental Pulp Stem Cell-Derived Factors Alleviate Subarachnoid Hemorrhage-Induced Neuroinflammation and Ischemic Neurological Deficits. Int J Mol Sci 2019; 20:ijms20153747. [PMID: 31370244 PMCID: PMC6695587 DOI: 10.3390/ijms20153747] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 12/15/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH), characterized by the extravasation of blood into the subarachnoid space caused by an intracranial aneurysm rupture, may lead to neurocognitive impairments and permanent disability and usually carries poor outcome. Dental or gingiva-derived stem cells have been shown to contribute to immune modulation and neuroregeneration, but the underlying mechanisms are unclear. In the present study, we sought to investigate whether dental pulp stem cells (DPSCs) secrete certain factor(s) that can ameliorate the neural damage and other manifestations in a rat aSAH model. Twenty-four hours after the induction of aSAH, microthrombosis, cortical vasoconstriction, and the decrease in microcirculation and tissue oxygen pressure were detected. Intrathecal administration of DPSC-derived conditioned media (DPSC-CM) ameliorated aSAH-induced vasoconstriction, neuroinflammation, and improved the oxygenation in the injured brain. Rotarod test revealed that the aSAH-induced cognitive and motor impairments were significantly improved by this DPSC-CM administration. Cytokine array indicated the major constituent of DPSC-CM was predominantly insulin growth factor-1 (IGF-1). Immunohistochemistry staining of injured brain tissue revealed the robust increase in Iba1-positive cells that were also ameliorated by DPSC-CM administration. Antibody-mediated neutralization of IGF-1 moderately deteriorated the rescuing effect of DPSC-CM on microcirculation, Iba1-positive cells in the injured brain area, and the cognitive/motor impairments. Taken together, the DPSC-derived secretory factors showed prominent therapeutic potential for aSAH. This therapeutic efficacy may include improvement of microcirculation, alleviation of neuroinflammation, and microglial activation; partially through IGF-1-dependent mechanisms.
Collapse
Affiliation(s)
- Te-Fu Chen
- Department of surgery, Division of Neurosurgery, National Taiwan University Hospital, Taipei 100, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Department of Neurosurgery, Tri-Service General Hospital, Taipei 115, Taiwan
- Non-invasive Cancer Therapy Research Institute - Taiwan, Taipei 104, Taiwan
| | - Kuo-We Chen
- Department of surgery, Division of Neurosurgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Yueh Chien
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Ying-Hsiu Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 102, Taiwan
| | - Hsin-Yi Ma
- Department of Neurosurgery, Tri-Service General Hospital, Taipei 115, Taiwan
| | - Kou-Chung Wang
- Department of surgery, Division of Neurosurgery, National Taiwan University Hospital, Taipei 100, Taiwan.
| | - Chia-Yang Shiau
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan.
| |
Collapse
|