1
|
Arrigo A, Cremona O, Aragona E, Casoni F, Consalez G, Dogru RM, Hauck SM, Antropoli A, Bianco L, Parodi MB, Bandello F, Grosche A. Müller cells trophism and pathology as the next therapeutic targets for retinal diseases. Prog Retin Eye Res 2025; 106:101357. [PMID: 40254246 DOI: 10.1016/j.preteyeres.2025.101357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Müller cells are a crucial retinal cell type involved in multiple regulatory processes and functions that are essential for retinal health and functionality. Acting as structural and functional support for retinal neurons and photoreceptors, Müller cells produce growth factors, regulate ion and fluid homeostasis, and facilitate neuronal signaling. They play a pivotal role in retinal morphogenesis and cell differentiation, significantly contributing to macular development. Due to their radial morphology and unique cytoskeletal organization, Müller cells act as optical fibers, efficiently channeling photons directly to the photoreceptors. In response to retinal damage, Müller cells undergo specific gene expression and functional changes that serve as a first line of defense for neurons, but can also lead to unwarranted cell dysfunction, contributing to cell death and neurodegeneration. In some species, Müller cells can reactivate their developmental program, promoting retinal regeneration and plasticity-a remarkable ability that holds promising therapeutic potential if harnessed in mammals. The crucial and multifaceted roles of Müller cells-that we propose to collectively call "Müller cells trophism"-highlight the necessity of maintaining their functionality. Dysfunction of Müller cells, termed "Müller cells pathology," has been associated with a plethora of retinal diseases, including age-related macular degeneration, diabetic retinopathy, vitreomacular disorders, macular telangiectasia, and inherited retinal dystrophies. In this review, we outline how even subtle disruptions in Müller cells trophism can drive the pathological cascade of Müller cells pathology, emphasizing the need for targeted therapies to preserve retinal health and prevent disease progression.
Collapse
Affiliation(s)
- Alessandro Arrigo
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Ottavio Cremona
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Emanuela Aragona
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Casoni
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo Consalez
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rüya Merve Dogru
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, 80939, Germany
| | - Alessio Antropoli
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Bianco
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesco Bandello
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
2
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.23.563601. [PMID: 37961151 PMCID: PMC10634739 DOI: 10.1101/2023.10.23.563601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
|
3
|
Grosche A, Grosche J, Verkhratsky A. Physiology and pathophysiology of the retinal neuroglia. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:239-265. [PMID: 40148047 DOI: 10.1016/b978-0-443-19102-2.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Neuroglia of the retina are represented by Müller glia, parenchymal astrocytes, microglia and oligodendrocytes mainly associated with the optic nerve. Müller glia are the most numerous glia, endowed with multiple homeostatic functions and indispensable for the retinal morphofunctional organization. Müller cells integrate retinal neurons into individual functional units (known as retinal columns) and act as a living light guide, transmitting photons to photoreceptors. In pathology, retinal neuroglia undergo complex changes, which include upregulation of neuroprotection, reactive gliosis, and functional asthenia. The balance between all these changes defines the progression and outcome of retinal disorders.
Collapse
Affiliation(s)
- Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, München, Germany.
| | | | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
4
|
Segars KL, Azzari N, Cole M, Kushimi L, Rapaka S, Rich CB, Trinkaus-Randall V. Diverse calcium signaling profiles regulate migratory behavior in avascular wound healing and aberrant signal hierarchy occurs early in diabetes. Am J Physiol Cell Physiol 2024; 327:C1051-C1072. [PMID: 39129489 PMCID: PMC11482046 DOI: 10.1152/ajpcell.00249.2024] [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: 04/16/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
In avascular wound repair, calcium signaling events are the predominant mechanism cells use to transduce information about stressors in the environment into an effective and coordinated migratory response. Live cell imaging and computational analysis of corneal epithelial wound healing revealed that signal initiation and propagation at the wound edge are highly ordered, with groups of cells engaging in cyclical patterns of initiation and propagation. The cells in these groups exhibit a diverse range of signaling behavior, and dominant "conductor cells" drive activity in groups of lower-signaling neighbors. Ex vivo model systems reveal that conductor cells are present in wing cell layers of the corneal epithelium and that signaling propagates both within and between wing and basal layers. There are significant aberrations in conductor phenotype and interlayer propagation in type II diabetic murine models, indicating that signal hierarchy breakdown is an early indicator of disease. In vitro models reveal that signaling profile diversity and conductor cell phenotype is eliminated with P2X7 inhibition and is altered in Pannexin-1 or P2Y2 but not Connexin-43 inhibition. Conductor cells express significantly less P2X7 than their lower-signaling neighbors and exhibit significantly less migratory behavior after injury. Together, our results show that the postinjury calcium signaling cascade exhibits significantly more ordered and hierarchical behavior than previously thought, that proteins previously shown to be essential for regulating motility are also essential for determining signaling phenotype, and that loss of signal hierarchy integrity is an early indicator of disease state. NEW & NOTEWORTHY Calcium signaling in corneal epithelial cells after injury is highly ordered, with groups of cells engaged in cyclical patterns of event initiation and propagation driven by high-signaling cells. Signaling behavior is determined by P2X7, Pannexin-1, and P2Y2 and influences migratory behavior. Signal hierarchy is observed in healthy ex vivo models after injury and becomes aberrant in diabetes. This represents a paradigm shift, as signaling was thought to be random and determined by factors in the environment.
Collapse
Affiliation(s)
- Kristen L Segars
- Department of Pharmacology, Physiology, and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Nicholas Azzari
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Malia Cole
- STaRS Program, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Landon Kushimi
- Department of Computer Science, Boston University Center for Computing and Data Sciences, Boston, Massachusetts, United States
| | - Srikar Rapaka
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Celeste B Rich
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Vickery Trinkaus-Randall
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
5
|
Niksirat H, Siino V, Steinbach C, Levander F. The quantification of zebrafish ocular-associated proteins provides hints for sex-biased visual impairments and perception. Heliyon 2024; 10:e33057. [PMID: 38994070 PMCID: PMC11238053 DOI: 10.1016/j.heliyon.2024.e33057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Biochemical differences between sexes can also be seen in non-sexual organs and may affect organ functions and susceptibility to diseases. It has been shown that there are sex-biased visual perceptions and impairments. Abundance differences of eye proteins could provide explanations for some of these. Exploration of the ocular proteome was performed to find sex-based protein abundance differences in zebrafish Danio rerio. A label-free protein quantification workflow using high-resolution mass spectrometry was employed to find proteins with significant differences between the sexes. In total, 3740 unique master proteins were identified and quantified, and 49 proteins showed significant abundance differences between the eyes of male and female zebrafish. Those proteins belong to lipoproteins, immune system, blood coagulation, antioxidants, iron and heme-binding proteins, ion channels, pumps and exchangers, neuronal and photoreceptor proteins, and the cytoskeleton. An extensive literature review provided clues for the possible links between the sex-biased level of proteins and visual perception and impairments. In conclusion, sexual dimorphism at the protein level was discovered for the first time in the eye of zebrafish and should be accounted for in ophthalmological studies. Data are available via ProteomeXchange with identifier PXD033338.
Collapse
Affiliation(s)
- Hamid Niksirat
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Christoph Steinbach
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund, Sweden
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Mauduit O, Delcroix V, Wong A, Ivanova A, Miles L, Lee HS, Makarenkova H. A closer look into the cellular and molecular biology of myoepithelial cells across various exocrine glands. Ocul Surf 2024; 31:63-80. [PMID: 38141817 PMCID: PMC10855576 DOI: 10.1016/j.jtos.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/02/2023] [Accepted: 12/09/2023] [Indexed: 12/25/2023]
Abstract
Myoepithelial cells (MECs) are a unique subset of epithelial cells that possess several smooth muscle cell characteristics, such as a high number of actin-myosin filaments and the ability to contract. These cells are primarily located around the secretory cells of exocrine glands, including the salivary, mammary, lacrimal, and sweat glands. Their primary functions involve the construction of the basement membrane and help with secretion of gland products through contraction. So far, no comparative analysis of MECs in different exocrine glands had ever evaluated their differences. In this review, we took advantage of the various publicly available scRNAseq data from mouse exocrine glands to identify their shared and unique characteristics. The aim of this review is to compare the role of MECs in maintaining healthy glandular function, their involvement in disease states, and their regenerative capacity, with a particular emphasis on the latest research findings in these areas.
Collapse
Affiliation(s)
- Olivier Mauduit
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Vanessa Delcroix
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Andrew Wong
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Anastasiia Ivanova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Lindsey Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Hyun Soo Lee
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA; Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Helen Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
7
|
Abdalkader RK, Fujita T. Corneal epithelium models for safety assessment in drug development: Present and future directions. Exp Eye Res 2023; 237:109697. [PMID: 37890755 DOI: 10.1016/j.exer.2023.109697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
The human corneal epithelial barrier plays a crucial role in drug testing studies, including drug absorption, distribution, metabolism, and excretion (ADME), as well as toxicity testing during the preclinical stages of drug development. However, despite the valuable insights gained from animal and current in vitro models, there remains a significant discrepancy between preclinical drug predictions and actual clinical outcomes. Additionally, there is a growing emphasis on adhering to the 3R principles (refine, reduce, replace) to minimize the use of animals in testing. To tackle these challenges, there is a rising demand for alternative in vitro models that closely mimic the human corneal epithelium. Recently, remarkable advancements have been made in two key areas: microphysiological systems (MPS) or organs-on-chips (OoCs), and stem cell-derived organoids. These cutting-edge platforms integrate four major disciplines: stem cells, microfluidics, bioprinting, and biosensing technologies. This integration holds great promise in developing powerful and biomimetic models of the human cornea.
Collapse
Affiliation(s)
- Rodi Kado Abdalkader
- Ritsumeikan Global Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Takuya Fujita
- Ritsumeikan Global Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan; Department of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
8
|
Chen J, Qin G, Li L, Qi Y, Xia Y, Zhang Q, Wu Y, You Y, Yang L, Guo N, Moutari S, Moore JE, Bu S, Xu L, He W, Yu S, Pazo EE, He X. The Combined Impact of Intense Pulsed Light Combined and 3% Diquafosol Ophthalmic Solution on Evaporative Dry Eye: A Randomized Control Study. Ophthalmol Ther 2023; 12:2959-2971. [PMID: 37589932 PMCID: PMC10640412 DOI: 10.1007/s40123-023-00784-z] [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: 06/12/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023] Open
Abstract
INTRODUCTION The primary objective of this study is to assess whether the combination of intense pulsed light (IPL) with 3% diquafosol (DQS) ophthalmic solution is more effective than intense pulsed light in alleviating signs and symptoms of dry eye disease (DED). METHODS This randomized study included 66 participants with evaporative dry eye (EDE) who received IPL + DQS therapy (n = 44 eyes), IPL therapy (n = 44 eyes), or sham therapy (n = 44 eyes). All participants were examined at baseline (D0), day 14 (D14), and day 28 (D28) for non-invasive break-up time (NITBUT), tear-film lipid layer (TFLL), corneal conjunctival staining (CS), meibomian gland quality (MGQ), meibomian gland expression (MGEx), and ocular surface disease index (OSDI). RESULTS At day 28, comparison among the IPL + DQS therapy, IPL therapy, and sham therapy found significant differences in the mean NITBUT (12.03 ± 1.27 versus 10.47 ± 3.48 versus 4.57 ± 0.46; p < 0.001), TFLL (2.09 ± 0.29 versus 2.27 ± 0.45 versus 2.89 ± 0.65; p < 0.001), CS (1.43 ± 0.82 versus 1.93 ± 1.32 versus 3.52 ± 1.00; p < 0.001), MGQ (1.55 ± 0.66 versus 1.91 ± 0.77 versus 2.66 ± 0.53; p < 0.001), MGEx (1.27 ± 0.45 versus 1.75 ± 0.44 versus 2.41 ± 0.50; p < 0.001), and OSDI score (19.36 ± 7.01 versus 24.77 ± 4.68 versus 42.61 ± 7.49; p < 0.001); significant improvements in NITBUT, TFLL, CS, MGQ, MGEx, and OSDI were found in the IPL + DQS therapy and IPL therapy, while the sham therapy had no significant improvements. CONCLUSION Combining 3% diquafosol ophthalmic solution with intense pulsed light was superior to IPL therapy alone in relieving the signs and symptoms of patients with severe evaporative DED. TRIAL REGISTRATION Clinical Trials Identifier: NCT05694026.
Collapse
Affiliation(s)
- Jiayan Chen
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Guanghao Qin
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Liangzhe Li
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Yifan Qi
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Yang Xia
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Qing Zhang
- Tianjin Medical University, Tianjin, China
| | - Yi Wu
- China Medical University, Shenyang, China
| | - Yue You
- Sinqi Eye Hospital, Shenyang, China
| | | | - Naici Guo
- University of St Andrews, St Andrews, UK
| | | | | | | | - Ling Xu
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Wei He
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Sile Yu
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China
| | - Emmanuel Eric Pazo
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China.
| | - Xingru He
- Department of Ophthalmology, He Eye Specialist Hospital, Shenyang, 110034, China.
- School of Public Health, He University, Shenyang, 110034, China.
| |
Collapse
|
9
|
Molcak H, Jiang K, Campbell CJ, Matsubara JA. Purinergic signaling via P2X receptors and mechanisms of unregulated ATP release in the outer retina and age-related macular degeneration. Front Neurosci 2023; 17:1216489. [PMID: 37496736 PMCID: PMC10366617 DOI: 10.3389/fnins.2023.1216489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
Age-related macular degeneration (AMD) is a chronic and progressive inflammatory disease of the retina characterized by photoceptor loss and significant central visual impairment due to either choroidal neovascularization or geographic atrophy. The pathophysiology of AMD is complex and multifactorial, driven by a combination of modifiable and non-modifiable risk factors, molecular mechanisms, and cellular processes that contribute to overall disease onset, severity, and progression. Unfortunately, due to the structural, cellular, and pathophysiologic complexity, therapeutic discovery is challenging. While purinergic signaling has been investigated for its role in the development and treatment of ocular pathologies including AMD, the potential crosstalk between known contributors to AMD, such as the complement cascade and inflammasome activation, and other biological systems, such as purinergic signaling, have not been fully characterized. In this review, we explore the interactions between purinergic signaling, ATP release, and known contributors to AMD pathogenesis including complement dysregulation and inflammasome activation. We begin by identifying what is known about purinergic receptors in cell populations of the outer retina and potential sources of extracellular ATP required to trigger purinergic receptor activation. Next, we examine evidence in the literature that the purinergic system accelerates AMD pathogenesis leading to apoptotic and pyroptotic cell death in retinal cells. To fully understand the potential role that purinergic signaling plays in AMD, more research is needed surrounding the expression, distribution, functions, and interactions of purinergic receptors within cells of the outer retina as well as potential crosstalk with other systems. By determining how these processes are affected in the context of purinergic signaling, it will improve our understanding of the mechanisms that drive AMD pathogenesis which is critical in developing treatment strategies that prevent or slow progression of the disease.
Collapse
Affiliation(s)
- Haydn Molcak
- Matsubara Lab, Faculty of Medicine, Department of Ophthalmology and Visual Sciences, Eye Care Centre, Vancouver, BC, Canada
| | - Kailun Jiang
- Matsubara Lab, Faculty of Medicine, Department of Ophthalmology and Visual Sciences, Eye Care Centre, Vancouver, BC, Canada
| | | | - Joanne A. Matsubara
- Matsubara Lab, Faculty of Medicine, Department of Ophthalmology and Visual Sciences, Eye Care Centre, Vancouver, BC, Canada
| |
Collapse
|
10
|
Markitantova Y, Simirskii V. Endogenous and Exogenous Regulation of Redox Homeostasis in Retinal Pigment Epithelium Cells: An Updated Antioxidant Perspective. Int J Mol Sci 2023; 24:10776. [PMID: 37445953 DOI: 10.3390/ijms241310776] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The retinal pigment epithelium (RPE) performs a range of necessary functions within the neural layers of the retina and helps ensure vision. The regulation of pro-oxidative and antioxidant processes is the basis for maintaining RPE homeostasis and preventing retinal degenerative processes. Long-term stable changes in the redox balance under the influence of endogenous or exogenous factors can lead to oxidative stress (OS) and the development of a number of retinal pathologies associated with RPE dysfunction, and can eventually lead to vision loss. Reparative autophagy, ubiquitin-proteasome utilization, the repair of damaged proteins, and the maintenance of their conformational structure are important interrelated mechanisms of the endogenous defense system that protects against oxidative damage. Antioxidant protection of RPE cells is realized as a result of the activity of specific transcription factors, a large group of enzymes, chaperone proteins, etc., which form many signaling pathways in the RPE and the retina. Here, we discuss the role of the key components of the antioxidant defense system (ADS) in the cellular response of the RPE against OS. Understanding the role and interactions of OS mediators and the components of the ADS contributes to the formation of ideas about the subtle mechanisms in the regulation of RPE cellular functions and prospects for experimental approaches to restore RPE functions.
Collapse
Affiliation(s)
- Yuliya Markitantova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Vladimir Simirskii
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
11
|
From lead to clinic: A review of the structural design of P2X7R antagonists. Eur J Med Chem 2023; 251:115234. [PMID: 36893624 DOI: 10.1016/j.ejmech.2023.115234] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023]
Abstract
P2X7R, which is a member of the purinergic P2 receptor family, is widely expressed in many immune cells, such as macrophages, lymphocytes, monocytes, and neutrophils. P2X7R is upregulated in response to proinflammatory stimulation, which is closely related to a variety of inflammatory diseases. The inhibition of P2X7 receptors has resulted in the elimination or reduction of symptoms in animal models of arthritis, depression, neuropathic pain, multiple sclerosis, and Alzheimer's disease. Therefore, the development of P2X7R antagonists is of great significance for the treatment of various inflammatory diseases. This review classifies the reported P2X7R antagonists according to their different cores, focuses on the structure-activity relationship (SAR) of the compounds, and analyzes some common substituents and strategies in the design of lead compounds, with the hope of providing valuable information for the development of new and efficient P2X7R antagonists.
Collapse
|
12
|
Wang JN, Fan H, Song JT. Targeting purinergic receptors to attenuate inflammation of dry eye. Purinergic Signal 2023; 19:199-206. [PMID: 35218451 PMCID: PMC9984584 DOI: 10.1007/s11302-022-09851-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/04/2022] [Indexed: 11/28/2022] Open
Abstract
Inflammation is one of the potential factors to cause the damage of ocular surface in dry eye disease (DED). Increasing evidence indicated that purinergic A1, A2A, A3, P2X4, P2X7, P2Y1, P2Y2, and P2Y4 receptors play an important role in the regulation of inflammation in DED: A1 adenosine receptor (A1R) is a systemic pro-inflammatory factor; A2AR is involved in the activation of the MAPK/NF-kB pathway; A3R combined with inhibition of adenylate cyclase and regulation of the mitogen-activated protein kinase (MAPK) pathway leads to regulation of transcription; P2X4 promotes receptor-associated activation of pro-inflammatory cytokines and inflammatory vesicles; P2X7 promotes inflammasome activation and release of pro-inflammatory cytokines IL-1β and IL-18; P2Y receptors affect the phospholipase C(PLC)/IP3/Ca2+ signaling pathway and mucin secretion. These suggested that purinergic receptors would be promising targets to control the inflammation of DED in the future.
Collapse
Affiliation(s)
- Jia-Ning Wang
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hua Fan
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian-Tao Song
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
13
|
Mechanical Stretch Activates TRPV4 and Hemichannel Responses in the Nonpigmented Ciliary Epithelium. Int J Mol Sci 2023; 24:ijms24021673. [PMID: 36675184 PMCID: PMC9865367 DOI: 10.3390/ijms24021673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Previously, we reported a mechanosensitive ion channel, TRPV4, along with functional connexin hemichannels on the basolateral surface of the ocular nonpigmented ciliary epithelium (NPE). In the lens, TRPV4-mediated hemichannel opening is part of a feedback loop that senses and respond to swelling. The present study was undertaken to test the hypothesis that TRPV4 and hemichannels in the NPE respond to a mechanical stimulus. Porcine NPE cells were cultured on flexible membranes to study effects of cyclic stretch and ATP release was determined by a luciferase assay. The uptake of propidium iodide (PI) was measured as an indicator of hemichannel opening. NPE cells subjected to cyclic stretch for 1-10 min (10%, 0.5 Hz) displayed a significant increase in ATP release into the bathing medium. In studies where PI was added to the bathing medium, the same stretch stimulus increased cell PI uptake. The ATP release and PI uptake responses to stretch both were prevented by a TRPV4 antagonist, HC067047 (10 µM), and a connexin mimetic peptide, Gap 27 (200µm). In the absence of a stretch stimulus, qualitatively similar ATP release and PI uptake responses were observed in cells exposed to the TRPV4 agonist GSK1016790A (10 nM), and Gap 27 prevented the responses. Cells subjected to an osmotic swelling stimulus (hypoosmotic medium: 200 mOsm) also displayed a significant increase in ATP release and PI uptake and the responses were abolished by TRPV4 inhibition. The findings point to TRPV4-dependent connexin hemichannel opening in response to mechanical stimulus. The TRPV4-hemichannel mechanism may act as a mechanosensor that facilitates the release of ATP and possibly other autocrine or paracrine signaling molecules that influence fluid (aqueous humor) secretion by the NPE.
Collapse
|
14
|
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
|
15
|
Wu P, Wang Y, Liu Y, Liu Y, Zhou G, Wu X, Wen Q. Emerging roles of the P2X7 receptor in cancer pain. Purinergic Signal 2022:10.1007/s11302-022-09902-1. [DOI: 10.1007/s11302-022-09902-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/03/2022] [Indexed: 11/21/2022] Open
Abstract
AbstractCancer pain is the most prevalent symptom experienced by cancer patients. It substantially impacts a patient’s long-term physical and emotional health, making it a pressing issue that must be addressed. Purinergic receptor P2X7 (P2X7R) is a widely distributed and potent non-selective ATP-gated ion channel that regulates tumor proliferation, chronic pain, and the formation of inflammatory lesions in the central nervous system. P2X7R plays an essential role in cancer pain and complications related to cancer pain including depression and opioid tolerance. This review focuses on the structure and distribution of P2X7R, its role in diverse tissues in cancer pain, and the application of P2X7R antagonists in the treatment of cancer pain to propose new ideas for cancer pain management.
Collapse
|
16
|
Faure C, Chassery M, Ores R, Audo I. Didanosine-induced Retinopathy: New Insights with Long-term Follow-up. Ocul Immunol Inflamm 2022; 30:1625-1632. [PMID: 34255599 DOI: 10.1080/09273948.2021.1927117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Didanosine is an adenosine analog, part of the nucleoside reverse-transcriptase inhibitor family. Since the description of didanosine-induced retinopathy in the early 1990s, little is known about the progression of this toxic retinopathy and the putative underlying mitochondrial defect. OBJECTIVES We report long-term follow-up for cases of didanosine-induced retinopathy and discuss a new hypothesis for pathophysiology based on the alteration of endogenous adenosine on the photoreceptor outer segment turnover and phagocytosis by the retinal pigment epithelium. METHODS Ophthalmic data from six cases (12 eyes) of didanosine-induced retinopathy from a single institution were retrospectively analyzed. RESULTS All patients displayed bilateral retinal alterations in the mid-periphery. Despite didanosine discontinuation, patients with advanced areas of patchy chorioretinal atrophy appeared to have a faster progression than those with limited lesions. Full-field electroretinogram revealed generalized rod-cone dysfunction in most cases that remained stable over time. CONCLUSION We propose new guidelines including early screening and long-term observations.
Collapse
Affiliation(s)
- Céline Faure
- Unité d'électrophysiologie, Centre Hospitalier National des Quinze-Vingts, Paris, France.,Hôpital Privé Saint Martin, Département d'ophtalmologie, Ramsay Générale de Santé, Caen, France
| | - Maxime Chassery
- Unité d'électrophysiologie, Centre Hospitalier National des Quinze-Vingts, Paris, France
| | - Raphaëlle Ores
- Unité d'électrophysiologie, Centre Hospitalier National des Quinze-Vingts, Paris, France
| | - Isabelle Audo
- Unité d'électrophysiologie, Centre Hospitalier National des Quinze-Vingts, Paris, France.,INSERM-DHOS CIC1423, CHNO des Quinze-Vingts, DHU Sight Restore, Paris, France.,Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
17
|
Li L, Jasmer KJ, Camden JM, Woods LT, Martin AL, Yang Y, Layton M, Petris MJ, Baker OJ, Weisman GA, Petris CK. Early Dry Eye Disease Onset in a NOD.H-2h4 Mouse Model of Sjögren's Syndrome. Invest Ophthalmol Vis Sci 2022; 63:18. [PMID: 35727180 PMCID: PMC9233292 DOI: 10.1167/iovs.63.6.18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To develop a mouse model of human dry eye disease (DED) for investigation of sex differences in autoimmune-associated dry eye pathology. Methods Ocular surface disease was assessed by quantifying corneal epithelial damage with lissamine green stain in the NOD.H-2h4,IFNγ−/−,CD28−/− (NOD.H-2h4 DKO) mouse model of Sjögren's syndrome (SS). Lacrimal gland function was assessed by tear volume quantification with phenol red thread and lacrimal gland inflammation (i.e., dacryoadenitis) was assessed by quantification of immune cell foci, flow cytometric analysis of immune cell composition, and expression of proinflammatory markers. Results The NOD.H-2h4 DKO mouse model of SS exhibits greater age-dependent increases in corneal damage than in NOD.H-2h4 parental mice and demonstrates an earlier disease onset in females compared to males. The severity of ocular surface disease correlates with loss of goblet cell density, increased conjunctivitis, and dacryoadenitis that is more pronounced in NOD.H-2h4 DKO than NOD.H-2h4 mice. B cells dominate lacrimal infiltrates in 16-week-old NOD.H-2h4 and NOD.H-2h4 DKO mice, but T helper cells and macrophages are also present. Lacrimal gland expression of proinflammatory genes, including the P2X7 and P2Y2 purinergic receptors, is greater in NOD.H-2h4 DKO than NOD.H-2h4 mice and correlates with dacryoadenitis. Conclusions Our results demonstrate for the first time that autoimmune dry eye disease occurs in both sexes of NOD.H-2h4 DKO and NOD.H-2h4 mice, with earlier onset in female NOD.H-2h4 DKO mice when compared to males of the same strain. This study demonstrates that both NOD.H-2h4 and NOD.H-2h4 DKO mice are novel models that closely resemble SS-related and sex-dependent DED.
Collapse
Affiliation(s)
- Lili Li
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Visual Science and Optometry Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Kimberly J Jasmer
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Jean M Camden
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Lucas T Woods
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Adam L Martin
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Yong Yang
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Ophthalmology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Maria Layton
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States
| | - Michael J Petris
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Ophthalmology, University of Missouri, Columbia, Missouri, United States
| | - Olga J Baker
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri, United States
| | - Gary A Weisman
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Carisa K Petris
- Department of Ophthalmology, University of Missouri, Columbia, Missouri, United States.,Mason Eye Institute, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
18
|
Pinilla I, Maneu V, Campello L, Fernández-Sánchez L, Martínez-Gil N, Kutsyr O, Sánchez-Sáez X, Sánchez-Castillo C, Lax P, Cuenca N. Inherited Retinal Dystrophies: Role of Oxidative Stress and Inflammation in Their Physiopathology and Therapeutic Implications. Antioxidants (Basel) 2022; 11:antiox11061086. [PMID: 35739983 PMCID: PMC9219848 DOI: 10.3390/antiox11061086] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a large group of genetically and clinically heterogeneous diseases characterized by the progressive degeneration of the retina, ultimately leading to loss of visual function. Oxidative stress and inflammation play fundamental roles in the physiopathology of these diseases. Photoreceptor cell death induces an inflammatory state in the retina. The activation of several molecular pathways triggers different cellular responses to injury, including the activation of microglia to eliminate debris and recruit inflammatory cells from circulation. Therapeutical options for IRDs are currently limited, although a small number of patients have been successfully treated by gene therapy. Many other therapeutic strategies are being pursued to mitigate the deleterious effects of IRDs associated with oxidative metabolism and/or inflammation, including inhibiting reactive oxygen species’ accumulation and inflammatory responses, and blocking autophagy. Several compounds are being tested in clinical trials, generating great expectations for their implementation. The present review discusses the main death mechanisms that occur in IRDs and the latest therapies that are under investigation.
Collapse
Affiliation(s)
- Isabel Pinilla
- Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa, University Hospital, 50009 Zaragoza, Spain
- Department of Surgery, University of Zaragoza, 50009 Zaragoza, Spain
- Correspondence: (I.P.); (V.M.)
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain;
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Correspondence: (I.P.); (V.M.)
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Laura Fernández-Sánchez
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain;
| | - Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Pedro Lax
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Nicolás Cuenca
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| |
Collapse
|
19
|
Combined drug triads for synergic neuroprotection in retinal degeneration. Biomed Pharmacother 2022; 149:112911. [DOI: 10.1016/j.biopha.2022.112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/23/2022] Open
|
20
|
Fernández-Albarral JA, de Hoz R, Matamoros JA, Chen L, López-Cuenca I, Salobrar-García E, Sánchez-Puebla L, Ramírez JM, Triviño A, Salazar JJ, Ramírez AI. Retinal Changes in Astrocytes and Müller Glia in a Mouse Model of Laser-Induced Glaucoma: A Time-Course Study. Biomedicines 2022; 10:biomedicines10050939. [PMID: 35625676 PMCID: PMC9138377 DOI: 10.3390/biomedicines10050939] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 11/28/2022] Open
Abstract
Macroglia (astrocytes and Müller glia) may play an important role in the pathogenesis of glaucoma. In a glaucoma mouse model, we studied the effects of unilateral laser-induced ocular hypertension (OHT) on macroglia in OHT and contralateral eyes at different time points after laser treatment (1, 3, 5, 8 and 15 days) using anti-GFAP and anti-MHC-II, analyzing the morphological changes, GFAP-labelled retinal area (GFAP-PA), and GFAP and MHC-II immunoreactivity intensities ((GFAP-IRI and MHC-II-IRI)). In OHT and contralateral eyes, with respect to naïve eyes, at all the time points, we found the following: (i) astrocytes with thicker somas and more secondary processes, mainly in the intermediate (IR) and peripheral retina (PR); (ii) astrocytes with low GFAP-IRI and only primary processes near the optic disc (OD); (iii) an increase in total GFAP-RA, which was higher at 3 and 5 days, except for at 15 days; (iv) an increase in GFAP-IRI in the IR and especially in the PR; (v) a decrease in GFAP-IRI near the OD, especially at 1 and 5 days; (vi) a significant increase in MHC-II-IRI, which was higher in the IR and PR; and (vii) the Müller glia were GFAP+ and MHC-II+. In conclusion, in this model of glaucoma, there is a bilateral macroglial activation maintained over time involved in the inflammatory glaucoma process.
Collapse
Affiliation(s)
- Jose A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - José A. Matamoros
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Lejing Chen
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Lidia Sánchez-Puebla
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Medicina, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Alberto Triviño
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Medicina, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
- Correspondence: (J.J.S.); (A.I.R.)
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
- Correspondence: (J.J.S.); (A.I.R.)
| |
Collapse
|
21
|
Losenkova K, Takeda A, Ragauskas S, Cerrada-Gimenez M, Vähätupa M, Kaja S, Paul ML, Schmies CC, Rolshoven G, Müller CE, Sandholm J, Jalkanen S, Kalesnykas G, Yegutkin GG. CD73 controls ocular adenosine levels and protects retina from light-induced phototoxicity. Cell Mol Life Sci 2022; 79:152. [PMID: 35212809 PMCID: PMC8881442 DOI: 10.1007/s00018-022-04187-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 01/28/2022] [Accepted: 02/04/2022] [Indexed: 01/03/2023]
Abstract
ATP and adenosine have emerged as important signaling molecules involved in vascular remodeling, retinal functioning and neurovascular coupling in the mammalian eye. However, little is known about the regulatory mechanisms of purinergic signaling in the eye. Here, we used three-dimensional multiplexed imaging, in situ enzyme histochemistry, flow cytometric analysis, and single cell transcriptomics to characterize the whole pattern of purine metabolism in mouse and human eyes. This study identified ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39), NTPDase2, and ecto-5′-nucleotidase/CD73 as major ocular ecto-nucleotidases, which are selectively expressed in the photoreceptor layer (CD73), optic nerve head, retinal vasculature and microglia (CD39), as well as in neuronal processes and cornea (CD39, NTPDase2). Specifically, microglial cells can create a spatially arranged network in the retinal parenchyma by extending and retracting their branched CD39high/CD73low processes and forming local “purinergic junctions” with CD39low/CD73− neuronal cell bodies and CD39high/CD73− retinal blood vessels. The relevance of the CD73–adenosine pathway was confirmed by flash electroretinography showing that pharmacological inhibition of adenosine production by injection of highly selective CD73 inhibitor PSB-12489 in the vitreous cavity of dark-adapted mouse eyes rendered the animals hypersensitive to prolonged bright light, manifested as decreased a-wave and b-wave amplitudes. The impaired electrical responses of retinal cells in PSB-12489-treated mice were not accompanied by decrease in total thickness of the retina or death of photoreceptors and retinal ganglion cells. Our study thus defines ocular adenosine metabolism as a complex and spatially integrated network and further characterizes the critical role of CD73 in maintaining the functional activity of retinal cells.
Collapse
Affiliation(s)
- Karolina Losenkova
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Akira Takeda
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | | | | | | | - Simon Kaja
- Experimentica Ltd., Kuopio, Finland.,Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Marius L Paul
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.,Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Constanze C Schmies
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Georg Rolshoven
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Christa E Müller
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Sirpa Jalkanen
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | | | - Gennady G Yegutkin
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.
| |
Collapse
|
22
|
Bianca Maria Platania C, Drago F, Bucolo C. The P2X7 receptor as a new pharmacological target for retinal diseases. Biochem Pharmacol 2022; 198:114942. [PMID: 35134386 DOI: 10.1016/j.bcp.2022.114942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 11/02/2022]
|
23
|
Mugisho OO, Green CR. The NLRP3 inflammasome in age-related eye disease: Evidence-based connexin hemichannel therapeutics. Exp Eye Res 2021; 215:108911. [PMID: 34958779 DOI: 10.1016/j.exer.2021.108911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/25/2021] [Accepted: 12/21/2021] [Indexed: 12/21/2022]
Abstract
The inflammasome pathway is a fundamental component of the innate immune system, playing a key role especially in chronic age-related eye diseases (AREDs). The inflammasome is of particular interest because it is a common disease pathway that once instigated, can amplify and perpetuate itself leading to chronic inflammation. With aging, it becomes more difficult to shut down inflammation after an insult but the common pathway means that a shared solution may be feasible that could be effective across multiple disease indications. This review focusses on the NLRP3 inflammasome, the most studied and characterized inflammasome in the eye. It describes the two-step signalling required for NLRP3 inflammasome complex activation, and provides evidence for its role in AREDs. In the final section, the article gives an overview of potential NLRP3 inflammasome targeting therapies, before presenting evidence for connexin hemichannel regulators as upstream blockers of inflammasome activation. These have shown therapeutic efficacy in multiple ocular disease models.
Collapse
Affiliation(s)
- Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
| | - Colin R Green
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, New Zealand
| |
Collapse
|
24
|
Markitantova YV, Simirskii VN. The Role of the Purinergic Signaling System in the Control of Histogenesis, Homeostasis, and Pathogenesis of the Vertebrate Retina. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421060084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
Shestopalov VI, Spurlock M, Gramlich OW, Kuehn MH. Immune Responses in the Glaucomatous Retina: Regulation and Dynamics. Cells 2021; 10:1973. [PMID: 34440742 PMCID: PMC8391899 DOI: 10.3390/cells10081973] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/27/2022] Open
Abstract
Glaucoma is a multifactorial disease resulting in progressive vision loss due to retinal ganglion cell (RGC) dysfunction and death. Early events in the pathobiology of the disease include oxidative, metabolic, or mechanical stress that acts upon RGC, causing these to rapidly release danger signals, including extracellular ATP, resulting in micro- and macroglial activation and neuroinflammation. Danger signaling also leads to the formation of inflammasomes in the retina that enable maturation of proinflammatory cytokines such IL-1β and IL-18. Chronic neuroinflammation can have directly damaging effects on RGC, but it also creates a proinflammatory environment and compromises the immune privilege of the retina. In particular, continuous synthesis of proinflammatory mediators such as TNFα, IL-1β, and anaphylatoxins weakens the blood-retina barrier and recruits or activates T-cells. Recent data have demonstrated that adaptive immune responses strongly exacerbate RGC loss in animal models of the disease as T-cells appear to target heat shock proteins displayed on the surface of stressed RGC to cause their apoptotic death. It is possible that dysregulation of these immune responses contributes to the continued loss of RGC in some patients.
Collapse
Affiliation(s)
- Valery I. Shestopalov
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA;
- Department of Cell and Developmental Biology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA;
- Graduate Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Kharkevich Institute for Information Transmission Problems, RAS, 127051 Moscow, Russia
| | - Markus Spurlock
- Department of Cell and Developmental Biology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA;
- Graduate Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| | - Oliver W. Gramlich
- Department of Veterans Affairs, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA;
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Markus H. Kuehn
- Department of Veterans Affairs, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA;
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
26
|
Jassim AH, Inman DM, Mitchell CH. Crosstalk Between Dysfunctional Mitochondria and Inflammation in Glaucomatous Neurodegeneration. Front Pharmacol 2021; 12:699623. [PMID: 34366851 PMCID: PMC8334009 DOI: 10.3389/fphar.2021.699623] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction and excessive inflammatory responses are both sufficient to induce pathology in age-dependent neurodegenerations. However, emerging evidence indicates crosstalk between damaged mitochondrial and inflammatory signaling can exacerbate issues in chronic neurodegenerations. This review discusses evidence for the interaction between mitochondrial damage and inflammation, with a focus on glaucomatous neurodegeneration, and proposes that positive feedback resulting from this crosstalk drives pathology. Mitochondrial dysfunction exacerbates inflammatory signaling in multiple ways. Damaged mitochondrial DNA is a damage-associated molecular pattern, which activates the NLRP3 inflammasome; priming and activation of the NLRP3 inflammasome, and the resulting liberation of IL-1β and IL-18 via the gasdermin D pore, is a major pathway to enhance inflammatory responses. The rise in reactive oxygen species induced by mitochondrial damage also activates inflammatory pathways, while blockage of Complex enzymes is sufficient to increase inflammatory signaling. Impaired mitophagy contributes to inflammation as the inability to turnover mitochondria in a timely manner increases levels of ROS and damaged mtDNA, with the latter likely to stimulate the cGAS-STING pathway to increase interferon signaling. Mitochondrial associated ER membrane contacts and the mitochondria-associated adaptor molecule MAVS can activate NLRP3 inflammasome signaling. In addition to dysfunctional mitochondria increasing inflammation, the corollary also occurs, with inflammation reducing mitochondrial function and ATP production; the resulting downward spiral accelerates degeneration. Evidence from several preclinical models including the DBA/2J mouse, microbead injection and transient elevation of IOP, in addition to patient data, implicates both mitochondrial damage and inflammation in glaucomatous neurodegeneration. The pressure-dependent hypoxia and the resulting metabolic vulnerability is associated with mitochondrial damage and IL-1β release. Links between mitochondrial dysfunction and inflammation can occur in retinal ganglion cells, microglia cells and astrocytes. In summary, crosstalk between damaged mitochondria and increased inflammatory signaling enhances pathology in glaucomatous neurodegeneration, with implications for other complex age-dependent neurodegenerations like Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Assraa Hassan Jassim
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Denise M. Inman
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Claire H. Mitchell
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, United States
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, United States
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
27
|
Hallaj S, Mirza-Aghazadeh-Attari M, Arasteh A, Ghorbani A, Lee D, Jadidi-Niaragh F. Adenosine: The common target between cancer immunotherapy and glaucoma in the eye. Life Sci 2021; 282:119796. [PMID: 34245774 DOI: 10.1016/j.lfs.2021.119796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/20/2022]
Abstract
Adenosine, an endogenous purine nucleoside, is a well-known actor of the immune system and the inflammatory response both in physiologic and pathologic conditions. By acting upon particular, G-protein coupled adenosine receptors, i.e., A1, A2- a & b, and A3 receptors mediate a variety of intracellular and immunomodulatory actions. Several studies have elucidated Adenosine's effect and its up-and downstream molecules and enzymes on the anti-tumor response against several types of cancers. We have also targeted a couple of molecules to manipulate this pathway and get the immune system's desired response in our previous experiences. Besides, the outgrowth of the studies on ocular Adenosine in recent years has significantly enhanced the knowledge about Adenosine and its role in ocular immunology and the inflammatory response of the eye. Glaucoma is the second leading cause of blindness globally, and the recent application of Adenosine and its derivatives has shown the critical role of the adenosine pathway in its pathophysiology. However, despite a very promising background, the phase III clinical trial of Trabodenoson failed to achieve the non-inferiority goals of the study. In this review, we discuss different aspects of the abovementioned pathway in ophthalmology and ocular immunology; following a brief evaluation of the current immunotherapeutic strategies, we try to elucidate the links between cancer immunotherapy and glaucoma in order to introduce novel therapeutic targets for glaucoma.
Collapse
Affiliation(s)
- Shahin Hallaj
- Wills Eye Hospital, Glaucoma Research Center, Philadelphia, PA 19107, USA
| | | | - Amin Arasteh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Anahita Ghorbani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daniel Lee
- Wills Eye Hospital, Glaucoma Research Center, Philadelphia, PA 19107, USA.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Kiessling E, Nötzli S, Todorova V, Forny M, Baumgartner MR, Samardzija M, Krijt J, Kožich V, Grimm C, Froese DS. Absence of MMACHC in peripheral retinal cells does not lead to an ocular phenotype in mice. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166201. [PMID: 34147638 DOI: 10.1016/j.bbadis.2021.166201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/25/2021] [Accepted: 06/08/2021] [Indexed: 01/10/2023]
Abstract
Combined methylmalonic aciduria with homocystinuria (cblC type) is a rare disease caused by mutations in the MMACHC gene. MMACHC encodes an enzyme crucial for intracellular vitamin B12 metabolism, leading to the accumulation of toxic metabolites e.g. methylmalonic acid (MMA) and homocysteine (Hcy), and secondary disturbances in folate and one-carbon metabolism when not fully functional. Patients with cblC deficiency often present in the neonatal or early childhood period with a severe multisystem pathology, which comprises a broad spectrum of treatment-resistant ophthalmological phenotypes, including retinal degeneration, impaired vision, and vascular changes. To examine the potential function of MMACHC in the retina and how its loss may impact disease, we performed gene expression studies in human and mouse, which showed that local expression of MMACHC in the retina and retinal pigment epithelium is relatively stable over time. To study whether functional MMACHC is required for retinal function and tissue integrity, we generated a transgenic mouse lacking Mmachc expression in cells of the peripheral retina. Characterization of this mouse revealed accumulation of cblC disease related metabolites, including MMA and the folate-dependent purine synthesis intermediates AICA-riboside and SAICA-riboside in the retina. Nevertheless, fundus appearance, morphology, vasculature, and cellular composition of the retina, as well as ocular function, remained normal in mice up to 6 or 12 months of age. Our data indicates that peripheral retinal neurons do not require intrinsic expression of Mmachc for survival and function and questions whether a local MMACHC deficiency is responsible for the retinal phenotypes in patients.
Collapse
Affiliation(s)
- Eva Kiessling
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Sarah Nötzli
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Vyara Todorova
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Merima Forny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland
| | - Marijana Samardzija
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Jakub Krijt
- Dept. of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Viktor Kožich
- Dept. of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Christian Grimm
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland.
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland.
| |
Collapse
|
29
|
Ye SS, Tang Y, Song JT. ATP and Adenosine in the Retina and Retinal Diseases. Front Pharmacol 2021; 12:654445. [PMID: 34211393 PMCID: PMC8239296 DOI: 10.3389/fphar.2021.654445] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular ATP and its ultimate degradation product adenosine are potent extracellular signaling molecules that elicit a variety of pathophysiological pathways in retina through the activation of P2 and P1 purinoceptors, respectively. Excessive build-up of extracellular ATP accelerates pathologic responses in retinal diseases, whereas accumulation of adenosine protects retinal cells against degeneration or inflammation. This mini-review focuses on the roles of ATP and adenosine in three types of blinding diseases including age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR). Several agonists and antagonists of ATP receptors and adenosine receptors (ARs) have been developed for the potential treatment of glaucoma, DR and AMD: antagonists of P2X7 receptor (P2X7R) (BBG, MRS2540) prevent ATP-induced neuronal apoptosis in glaucoma, DR, and AMD; A1 receptor (A1R) agonists (INO-8875) lower intraocular pressure in glaucoma; A2A receptor (A2AR) agonists (CGS21680) or antagonists (SCH58261, ZM241385) reduce neuroinflammation in glaucoma, DR, and AMD; A3 receptor (A3R) agonists (2-Cl-lB-MECA, MRS3558) protect retinal ganglion cells (RGCs) from apoptosis in glaucoma.
Collapse
Affiliation(s)
- Shan-Shan Ye
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Jian-Tao Song
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
30
|
Harsing LG, Szénási G, Zelles T, Köles L. Purinergic-Glycinergic Interaction in Neurodegenerative and Neuroinflammatory Disorders of the Retina. Int J Mol Sci 2021; 22:ijms22126209. [PMID: 34201404 PMCID: PMC8228622 DOI: 10.3390/ijms22126209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative–neuroinflammatory disorders of the retina seriously hamper human vision. In searching for key factors that contribute to the development of these pathologies, we considered potential interactions among purinergic neuromodulation, glycinergic neurotransmission, and microglia activity in the retina. Energy deprivation at cellular levels is mainly due to impaired blood circulation leading to increased release of ATP and adenosine as well as glutamate and glycine. Interactions between these modulators and neurotransmitters are manifold. First, P2Y purinoceptor agonists facilitate reuptake of glycine by glycine transporter 1, while its inhibitors reduce reverse-mode operation; these events may lower extracellular glycine levels. The consequential changes in extracellular glycine concentration can lead to parallel changes in the activity of NR1/NR2B type NMDA receptors of which glycine is a mandatory agonist, and thereby may reduce neurodegenerative events in the retina. Second, P2Y purinoceptor agonists and glycine transporter 1 inhibitors may indirectly inhibit microglia activity by decreasing neuronal or glial glycine release in energy-compromised retina. These inhibitions may have a role in microglia activation, which is present during development and progression of neurodegenerative disorders such as glaucomatous and diabetic retinopathies and age-related macular degeneration or loss of retinal neurons caused by thromboembolic events. We have hypothesized that glycine transporter 1 inhibitors and P2Y purinoceptor agonists may have therapeutic importance in neurodegenerative–neuroinflammatory disorders of the retina by decreasing NR1/NR2B NMDA receptor activity and production and release of a series of proinflammatory cytokines from microglial cells.
Collapse
Affiliation(s)
- Laszlo G. Harsing
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Correspondence: ; Tel.: +36-1-210-4416
| | - Gábor Szénási
- Institute of Translational Medicine, Semmelweis University, H-1089 Budapest, Hungary;
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
| | - László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary; (T.Z.); (L.K.)
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary
| |
Collapse
|
31
|
Sabirov RZ, Islam MR, Okada T, Merzlyak PG, Kurbannazarova RS, Tsiferova NA, Okada Y. The ATP-Releasing Maxi-Cl Channel: Its Identity, Molecular Partners and Physiological/Pathophysiological Implications. Life (Basel) 2021; 11:life11060509. [PMID: 34073084 PMCID: PMC8229958 DOI: 10.3390/life11060509] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/18/2022] Open
Abstract
The Maxi-Cl phenotype accounts for the majority (app. 60%) of reports on the large-conductance maxi-anion channels (MACs) and has been detected in almost every type of cell, including placenta, endothelium, lymphocyte, cardiac myocyte, neuron, and glial cells, and in cells originating from humans to frogs. A unitary conductance of 300-400 pS, linear current-to-voltage relationship, relatively high anion-to-cation selectivity, bell-shaped voltage dependency, and sensitivity to extracellular gadolinium are biophysical and pharmacological hallmarks of the Maxi-Cl channel. Its identification as a complex with SLCO2A1 as a core pore-forming component and two auxiliary regulatory proteins, annexin A2 and S100A10 (p11), explains the activation mechanism as Tyr23 dephosphorylation at ANXA2 in parallel with calcium binding at S100A10. In the resting state, SLCO2A1 functions as a prostaglandin transporter whereas upon activation it turns to an anion channel. As an efficient pathway for chloride, Maxi-Cl is implicated in a number of physiologically and pathophysiologically important processes, such as cell volume regulation, fluid secretion, apoptosis, and charge transfer. Maxi-Cl is permeable for ATP and other small signaling molecules serving as an electrogenic pathway in cell-to-cell signal transduction. Mutations at the SLCO2A1 gene cause inherited bone and gut pathologies and malignancies, signifying the Maxi-Cl channel as a perspective pharmacological target.
Collapse
Affiliation(s)
- Ravshan Z. Sabirov
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
- Correspondence: (R.Z.S.); (Y.O.); Tel.: +81-46-858-1501 (Y.O.); Fax: +81-46-858-1542 (Y.O.)
| | - Md. Rafiqul Islam
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Department of Biochemistry and Molecular Biology, Jagannath University, Dhaka 1100, Bangladesh
| | - Toshiaki Okada
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Veneno Technologies Co. Ltd., Tsukuba 305-0031, Japan
| | - Petr G. Merzlyak
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
| | - Ranokhon S. Kurbannazarova
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
| | - Nargiza A. Tsiferova
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent 100174, Uzbekistan
| | - Yasunobu Okada
- Division of Cell Signaling, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan; (M.R.I.); (T.O.); (P.G.M.); (R.S.K.); (N.A.T.)
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Physiology, School of Medicine, Aichi Medical University, Nagakute 480-1195, Japan
- Correspondence: (R.Z.S.); (Y.O.); Tel.: +81-46-858-1501 (Y.O.); Fax: +81-46-858-1542 (Y.O.)
| |
Collapse
|
32
|
Fletcher EL. Advances in understanding the mechanisms of retinal degenerations. Clin Exp Optom 2021; 103:723-732. [PMID: 33090561 DOI: 10.1111/cxo.13146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 01/13/2023] Open
Abstract
Photoreceptor death is an important contributor to irreversible vision loss worldwide. In this review, I outline our work examining the role that purines, such as adenosine triphosphate (ATP), have in normal retinal function and in retinal disease. Our work shows that the actions of ATP, mediated by P2X receptors, are expressed in various retinal layers including photoreceptor terminals, and when stimulated by excessive levels of ATP is associated with rapid death of neurons. Treatment with a compound that blocks the action of P2X and some P2Y receptors reduces photoreceptor death in a mouse model of retinal degeneration. Our observations not only provide a means for developing a potential treatment for reducing photoreceptor death, but also provides a novel way of studying the neural plasticity effects that develop in the inner retina following photoreceptor death. There are a range of inner retinal changes that could influence the effectiveness of retinal prostheses. Indeed, using an ATP-induced degeneration model, we established that the amount of electrical stimulation required to elicit a response in the visual cortex was affected by the level of glial scarring. However, changes in P2X7 receptor expression by OFF ganglion cells during retinal degeneration can also be exploited by photoswitches to restore light sensitivity to degenerated retinae. Finally, our work has also considered how P2X7 expression by innate immune cells, and its role as a scavenger receptor, contributes to age-related macular degeneration (AMD). Our results show that loss of P2X7 function is associated with thickening of Bruch's membrane as well as increased risk of advanced disease in people with AMD. Overall, our work over the last 20 years highlights the importance of purinergic signalling in normal retinal function and retinal disease and suggest that developing therapies that target P2X7 function could be of benefit for these diseases.
Collapse
Affiliation(s)
- Erica L Fletcher
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
33
|
Husain S, Zaidi SAH, Singh S, Guzman W, Mehrotra S. Reduction of Neuroinflammation by δ-Opioids Via STAT3-Dependent Pathway in Chronic Glaucoma Model. Front Pharmacol 2021; 12:601404. [PMID: 33628191 PMCID: PMC7898062 DOI: 10.3389/fphar.2021.601404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/05/2021] [Indexed: 12/25/2022] Open
Abstract
The main objective of this study was to determine the inhibition of pro-inflammatory cytokines and their associated signaling molecules by δ-opioid receptor activation by a selective ligand, SNC-121 in chronic rat glaucoma model. Intraocular pressure was raised in rat eyes by injecting 2 M hypertonic saline into the limbal veins. SNC-121 (1 mg/kg; i. p) or Stattic (5 mg/kg; i. p) was administered in Brown Norway rats daily for 7 days. The mRNA expression of IL-1β, TNF-α, Fas, IL-6, leukemia inhibitory factor, and IFN-γ was increased significantly in the retina of ocular hypertensive animals at day 7, post injury. Administration of SNC-121 (1 mg/kg; i. p. injection) for 7 days (once a day) completely inhibited the increase in the mRNA and protein expression of pro-inflammatory cytokines. Mechanistically, we provide data showing a significant increase in the phosphorylation of STAT3 at tyrosine 705 whereas a moderate but significant increase in the total STAT3 protein expression was also seen in the retina of ocular hypertensive animals. Data illustrated that SNC-121 administration completely abrogated ocular hypertension-induced increase in STAT3Y705 phosphorylation. Interestingly, acetylation of STAT3 at lysine 685 (AcK685) was reduced in ocular hypertensive animals and subsequently increased significantly by SNC-121 treatment. Stattic, a selective STAT3 inhibitor, administration resulted in a complete attenuation in the production of IL-1β and IL-6 in ocular hypertensive animals. In conclusion, δ-opioid receptor activation suppressed the phosphorylation of STAT3 at tyrosine 705 and increased acetylation at lysine 686 and these posttranslational modifications can regulate the production of some but not all pro-inflammatory cytokines in response to glaucomatous injury.
Collapse
Affiliation(s)
- Shahid Husain
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, United States
| | - Syed A H Zaidi
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, United States
| | - Sudha Singh
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, United States
| | - Wendy Guzman
- Department of Ophthalmology, Storm Eye Institute, Medical University of South Carolina, Charleston, SC, United States
| | - Shikhar Mehrotra
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
34
|
Smith EL, Hung LF, She Z, Beach K, Ostrin LA, Jong M. Topically instilled caffeine selectively alters emmetropizing responses in infant rhesus monkeys. Exp Eye Res 2021; 203:108438. [PMID: 33428866 DOI: 10.1016/j.exer.2021.108438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 11/30/2022]
Abstract
Oral administration of the adenosine receptor (ADOR) antagonist, 7-methylxanthine (7-MX), reduces both form-deprivation and lens-induced myopia in mammalian animal models. We investigated whether topically instilled caffeine, another non-selective ADOR antagonist, retards vision-induced axial elongation in monkeys. Beginning at 24 days of age, a 1.4% caffeine solution was instilled in both eyes of 14 rhesus monkeys twice each day until the age of 135 days. Concurrent with the caffeine regimen, the monkeys were fitted with helmets that held either -3 D (-3D/pl caffeine, n = 8) or +3 D spectacle lenses (+3D/pl caffeine, n = 6) in front of their lens-treated eyes and zero-powered lenses in front of their fellow-control eyes. Refractive errors and ocular dimensions were measured at baseline and periodically throughout the lens-rearing period. Control data were obtained from 8 vehicle-treated animals also reared with monocular -3 D spectacles (-3D/pl vehicle). In addition, historical comparison data were available for otherwise untreated lens-reared controls (-3D/pl controls, n = 20; +3D/pl controls, n = 9) and 41 normal monkeys. The vehicle controls and the untreated lens-reared controls consistently developed compensating axial anisometropias (-3D/pl vehicle = -1.44 ± 1.04 D; -3D/pl controls = -1.85 ± 1.20 D; +3D/pl controls = +1.92 ± 0.56 D). The caffeine regime did not interfere with hyperopic compensation in response to +3 D of anisometropia (+1.93 ± 0.82 D), however, it reduced the likelihood that animals would compensate for -3 D of anisometropia (+0.58 ± 1.82 D). The caffeine regimen also promoted hyperopic shifts in both the lens-treated and fellow-control eyes; 26 of the 28 caffeine-treated eyes became more hyperopic than the median normal monkey (mean (±SD) relative hyperopia = +2.27 ± 1.65 D; range = +0.31 to +6.37 D). The effects of topical caffeine on refractive development, which were qualitatively similar to those produced by oral administration of 7-MX, indicate that ADOR antagonists have potential in treatment strategies for preventing and/or reducing myopia progression.
Collapse
Affiliation(s)
- Earl L Smith
- College of Optometry, University of Houston, Houston, TX, United States; Brien Holden Vision Institute, Sydney, Australia.
| | - Li-Fang Hung
- College of Optometry, University of Houston, Houston, TX, United States; Brien Holden Vision Institute, Sydney, Australia
| | - Zhihui She
- College of Optometry, University of Houston, Houston, TX, United States
| | - Krista Beach
- College of Optometry, University of Houston, Houston, TX, United States
| | - Lisa A Ostrin
- College of Optometry, University of Houston, Houston, TX, United States
| | - Monica Jong
- Brien Holden Vision Institute, Sydney, Australia; Discipline of Optometry and Vision Science, University of Canberra, Canberra, Australia
| |
Collapse
|
35
|
Fernandez-Abascal J, Graziano B, Encalada N, Bianchi L. Glial Chloride Channels in the Function of the Nervous System Across Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:195-223. [PMID: 35138616 PMCID: PMC11247392 DOI: 10.1007/978-981-16-4254-8_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the nervous system, the concentration of Cl- in neurons that express GABA receptors plays a key role in establishing whether these neurons are excitatory, mostly during early development, or inhibitory. Thus, much attention has been dedicated to understanding how neurons regulate their intracellular Cl- concentration. However, regulation of the extracellular Cl- concentration by other cells of the nervous system, including glia and microglia, is as important because it ultimately affects the Cl- equilibrium potential across the neuronal plasma membrane. Moreover, Cl- ions are transported in and out of the cell, via either passive or active transporter systems, as counter ions for K+ whose concentration in the extracellular environment of the nervous system is tightly regulated because it directly affects neuronal excitability. In this book chapter, we report on the Cl- channel types expressed in the various types of glial cells focusing on the role they play in the function of the nervous system in health and disease. Furthermore, we describe the types of stimuli that these channels are activated by, the other solutes that they may transport, and the involvement of these channels in processes such as pH regulation and Regulatory Volume Decrease (RVD). The picture that emerges is one of the glial cells expressing a variety of Cl- channels, encoded by members of different gene families, involved both in short- and long-term regulation of the nervous system function. Finally, we report data on invertebrate model organisms, such as C. elegans and Drosophila, that are revealing important and previously unsuspected functions of some of these channels in the context of living and behaving animals.
Collapse
Affiliation(s)
- Jesus Fernandez-Abascal
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Bianca Graziano
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Nicole Encalada
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Laura Bianchi
- Department Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
36
|
Yang K, Liu J, Zhang X, Ren Z, Gao L, Wang Y, Lin W, Ma X, Hao M, Kuang H. H3 Relaxin Alleviates Migration, Apoptosis and Pyroptosis Through P2X7R-Mediated Nucleotide Binding Oligomerization Domain-Like Receptor Protein 3 Inflammasome Activation in Retinopathy Induced by Hyperglycemia. Front Pharmacol 2020; 11:603689. [PMID: 33584279 PMCID: PMC7873867 DOI: 10.3389/fphar.2020.603689] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022] Open
Abstract
Introduction: P2X7R excitation-interrelated NLRP3 inflammasome activation induced by high glucose contributes to the pathogenesis of diabetic retinopathy (DR). Relaxin-3 is a bioactive peptide with a structure similar to insulin, which has been reported to be effective in diabetic cardiomyopathy models in vivo and in vitro. However, it is not known whether relaxin-3 has a beneficial impact on DR, and the underlying mechanisms of the effect are also remain unknown. Methods and Results: The retinas of male streptozotocin (STZ)-induced diabetic Sprague-Dawley (SD) rats were characterized. Human retinal microvascular endothelial cells (HRMECs) were used to evaluate the anti-inflammatory, antiapoptotic, antipyroptotic and anti-migration effects of H3 relaxin by transmission electron microscopy, wound-healing assay, transwell assay, flow cytometry, cytokine assays and western-blot analysis. After H3 relaxin treatment, changes of the ultrastructure and expression of NLRP3 inflammasome related proteins in the retinas of rats were compared with those in the diabetic group. In vitro, H3 relaxin played a beneficial role that decreased cell inflammation, apoptosis, pyroptosis and migration stimulated by advanced glycation end products (AGEs). Moreover, inhibition of P2X7R and NLRP3 inflammasome activation decreased NLRP3 inflammasome-mediated injury that similar to the effects of H3 relaxin. H3 relaxin suppressed the stimulation of apoptosis, pyroptosis and migration of HRMECs in response to AGEs mediated by P2X7R activation of the NLRP3 inflammasome. Conclusion: Our findings provide new insights into the mechanisms of the inhibitory effect of H3 relaxin on AGE-induced retinal injury, including migration, apoptosis and pyroptosis, mediated by P2X7R-dependent activation of the NLRP3 inflammasome in HRMECs.
Collapse
Affiliation(s)
- Kelaier Yang
- The Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiannan Liu
- The Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaohui Zhang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziqi Ren
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Gao
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Wang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjian Lin
- The Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuefei Ma
- The Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ming Hao
- The Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongyu Kuang
- The Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Dihydrotanshinone, a Natural Diterpenoid, Preserves Blood-Retinal Barrier Integrity via P2X7 Receptor. Int J Mol Sci 2020; 21:ijms21239305. [PMID: 33291318 PMCID: PMC7730037 DOI: 10.3390/ijms21239305] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/23/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Activation of P2X7 signaling, due to high glucose levels, leads to blood retinal barrier (BRB) breakdown, which is a hallmark of diabetic retinopathy (DR). Furthermore, several studies report that high glucose (HG) conditions and the related activation of the P2X7 receptor (P2X7R) lead to the over-expression of pro-inflammatory markers. In order to identify novel P2X7R antagonists, we carried out virtual screening on a focused compound dataset, including indole derivatives and natural compounds such as caffeic acid phenethyl ester derivatives, flavonoids, and diterpenoids. Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) rescoring and structural fingerprint clustering of docking poses from virtual screening highlighted that the diterpenoid dihydrotanshinone (DHTS) clustered with the well-known P2X7R antagonist JNJ47965567. A human-based in vitro BRB model made of retinal pericytes, astrocytes, and endothelial cells was used to assess the potential protective effect of DHTS against HG and 2′(3′)-O-(4-Benzoylbenzoyl)adenosine-5′-triphosphate (BzATP), a P2X7R agonist, insult. We found that HG/BzATP exposure generated BRB breakdown by enhancing barrier permeability (trans-endothelial electrical resistance (TEER)) and reducing the levels of ZO-1 and VE-cadherin junction proteins as well as of the Cx-43 mRNA expression levels. Furthermore, HG levels and P2X7R agonist treatment led to increased expression of pro-inflammatory mediators (TLR-4, IL-1β, IL-6, TNF-α, and IL-8) and other molecular markers (P2X7R, VEGF-A, and ICAM-1), along with enhanced production of reactive oxygen species. Treatment with DHTS preserved the BRB integrity from HG/BzATP damage. The protective effects of DHTS were also compared to the validated P2X7R antagonist, JNJ47965567. In conclusion, we provided new findings pointing out the therapeutic potential of DHTS, which is an inhibitor of P2X7R, in terms of preventing and/or counteracting the BRB dysfunctions elicited by HG conditions.
Collapse
|
38
|
Shao X, Guha S, Lu W, Campagno KE, Beckel JM, Mills JA, Yang W, Mitchell CH. Polarized Cytokine Release Triggered by P2X7 Receptor from Retinal Pigmented Epithelial Cells Dependent on Calcium Influx. Cells 2020; 9:cells9122537. [PMID: 33255431 PMCID: PMC7760537 DOI: 10.3390/cells9122537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
Cytokine release from non-inflammatory cells is a key step in innate immunity, and agonists triggering cytokine release are central in coordinating responses. P2X7 receptor (P2X7R) stimulation by extracellular ATP is best known to active the NLRP3 inflammasome and release IL-1β, but stimulation also leads to release of other cytokines. As cytokine signaling by retinal pigmented epithelial (RPE) cells is implicated in retinal neurodegeneration, the role of P2X7R in release of cytokine IL-6 from RPE cells was investigated. P2X7R stimulation triggered IL-6 release from primary mouse RPE, human iPS-RPE and human ARPE-19 cells. IL-6 release was polarized, with predominant rise across apical membranes. IL-6 release was inhibited by P2X7R antagonists A438079, A839977, and AZ10606120, but not the NRTI lamivudine (3TC), P2X1R antagonist NF279, or P2Y1R antagonist MRS2179. P2X7R-mediated IL-6 release required extracellular Ca2+ and was blocked by Ca2+ chelator BAPTA. IL-6 release and Ca2+ elevation occurred rapidly, consistent with vesicular IL-6 staining in unstimulated cells. P2X7R stimulation did not trigger IL-1β release in these unprimed cells. P2X7R-mediated IL-6 release was enhanced in RPE cells from the ABCA4−/− mouse model of retinal degeneration. In summary, P2X7R stimulation triggers rapid Ca2+-dependent IL-6 release across the apical membrane of RPE cells.
Collapse
Affiliation(s)
- Xiaolei Shao
- Shenzhen Eye Hospital, School of Optometry, Shenzhen University, Shenzhen 518060, China;
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.G.); (J.M.B.)
| | - Sonia Guha
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.G.); (J.M.B.)
- Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Wennan Lu
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA 19104, USA; (W.L.); (K.E.C.)
| | - Keith E. Campagno
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA 19104, USA; (W.L.); (K.E.C.)
| | - Jonathan M. Beckel
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.G.); (J.M.B.)
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jason A. Mills
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenli Yang
- Department of Medicine and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Claire H. Mitchell
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.G.); (J.M.B.)
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA 19104, USA; (W.L.); (K.E.C.)
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence: ; Tel.: +1-215-573-2176
| |
Collapse
|
39
|
Calzaferri F, Ruiz-Ruiz C, de Diego AMG, de Pascual R, Méndez-López I, Cano-Abad MF, Maneu V, de Los Ríos C, Gandía L, García AG. The purinergic P2X7 receptor as a potential drug target to combat neuroinflammation in neurodegenerative diseases. Med Res Rev 2020; 40:2427-2465. [PMID: 32677086 DOI: 10.1002/med.21710] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 06/22/2020] [Accepted: 06/27/2020] [Indexed: 12/25/2022]
Abstract
Neurodegenerative diseases (NDDs) represent a huge social burden, particularly in Alzheimer's disease (AD) in which all proposed treatments investigated in murine models have failed during clinical trials (CTs). Thus, novel therapeutic strategies remain crucial. Neuroinflammation is a common pathogenic feature of NDDs. As purinergic P2X7 receptors (P2X7Rs) are gatekeepers of inflammation, they could be developed as drug targets for NDDs. Herein, we review this challenging hypothesis and comment on the numerous studies that have investigated P2X7Rs, emphasizing their molecular structure and functions, as well as their role in inflammation. Then, we elaborate on research undertaken in the field of medicinal chemistry to determine potential P2X7R antagonists. Subsequently, we review the state of neuroinflammation and P2X7R expression in the brain, in animal models and patients suffering from AD, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, multiple sclerosis, and retinal degeneration. Next, we summarize the in vivo studies testing the hypothesis that by mitigating neuroinflammation, P2X7R blockers afford neuroprotection, increasing neuroplasticity and neuronal repair in animal models of NDDs. Finally, we reviewed previous and ongoing CTs investigating compounds directed toward targets associated with NDDs; we propose that CTs with P2X7R antagonists should be initiated. Despite the high expectations for putative P2X7Rs antagonists in various central nervous system diseases, the field is moving forward at a relatively slow pace, presumably due to the complexity of P2X7Rs. A better pharmacological approach to combat NDDs would be a dual strategy, combining P2X7R antagonism with drugs targeting a selective pathway in a given NDD.
Collapse
Affiliation(s)
- Francesco Calzaferri
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Ruiz-Ruiz
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio M G de Diego
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ricardo de Pascual
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Iago Méndez-López
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - María F Cano-Abad
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Victoria Maneu
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, San Vicente del Raspeig, Spain
| | - Cristóbal de Los Ríos
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Gandía
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio G García
- Departamento de Farmacología, Instituto-Fundación Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
40
|
Jackson EK, Gillespie DG, Cheng D, Mi Z, Menshikova EV. Characterization of the N 6-etheno-bridge method to assess extracellular metabolism of adenine nucleotides: detection of a possible role for purine nucleoside phosphorylase in adenosine metabolism. Purinergic Signal 2020; 16:187-211. [PMID: 32367441 PMCID: PMC7367995 DOI: 10.1007/s11302-020-09699-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
The goal of this study was to determine the validity of using N6-etheno-bridged adenine nucleotides to evaluate ecto-nucleotidase activity. We observed that the metabolism of N6-etheno-ATP versus ATP was quantitatively similar when incubated with recombinant CD39, ENTPD2, ENTPD3, or ENPP-1, and the quantitative metabolism of N6-etheno-AMP versus AMP was similar when incubated with recombinant CD73. This suggests that ecto-nucleotidases process N6-etheno-bridged adenine nucleotides similarly to endogenous adenine nucleotides. Four cell types rapidly (t1/2, 0.21 to 0.66 h) metabolized N6-etheno-ATP. Applied N6-etheno-ATP was recovered in the medium as N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and surprisingly N6-etheno-adenine; intracellular N6-etheno compounds were undetectable. This suggests minimal cellular uptake, intracellular metabolism, or deamination of these compounds. N6-etheno-ATP, N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and N6-etheno-adenine had little affinity for recombinant A1, A2A, or A2B receptors, for a subset of P2X receptors (3H-α,β-methylene-ATP binding to rat bladder membranes), or for a subset of P2Y receptors (35S-ATP-αS binding to rat brain membranes), suggesting minimal pharmacological activity. N6-etheno-adenosine was partially converted to N6-etheno-adenine in four different cell types; this was blocked by purine nucleoside phosphorylase (PNPase) inhibition. Intravenous N6-etheno-ATP was quickly metabolized, with N6-etheno-adenine being the main product in naïve rats, but not in rats pretreated with a PNPase inhibitor. PNPase inhibition reduced the urinary excretion of endogenous adenine and attenuated the conversion of exogenous adenosine to adenine in the renal cortex. The N6-etheno-bridge method is a valid technique to assess extracellular metabolism of adenine nucleotides by ecto-nucleotidases. Also, rats express an enzyme with PNPase-like activity that metabolizes N6-etheno-adenosine to N6-etheno-adenine.
Collapse
Affiliation(s)
- Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Delbert G. Gillespie
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Dongmei Cheng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Elizabeth V. Menshikova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| |
Collapse
|
41
|
Ali AA, Avakian GA, Von Gall C. The Role of Purinergic Receptors in the Circadian System. Int J Mol Sci 2020; 21:E3423. [PMID: 32408622 PMCID: PMC7279285 DOI: 10.3390/ijms21103423] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/24/2022] Open
Abstract
The circadian system is an internal time-keeping system that synchronizes the behavior and physiology of an organism to the 24 h solar day. The master circadian clock, the suprachiasmatic nucleus (SCN), resides in the hypothalamus. It receives information about the environmental light/dark conditions through the eyes and orchestrates peripheral oscillators. Purinergic signaling is mediated by extracellular purines and pyrimidines that bind to purinergic receptors and regulate multiple body functions. In this review, we highlight the interaction between the circadian system and purinergic signaling to provide a better understanding of rhythmic body functions under physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | - Charlotte Von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany; (A.A.H.A.); (G.A.A.)
| |
Collapse
|
42
|
Kim JW, Lee JB, Lee SH. Effect and Mechanism of Phosphodiesterase Inhibitors on Trabecular Outflow. KOREAN JOURNAL OF OPHTHALMOLOGY 2019; 33:414-421. [PMID: 31612651 PMCID: PMC6791954 DOI: 10.3341/kjo.2019.0057] [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: 05/21/2019] [Revised: 06/10/2019] [Accepted: 07/15/2019] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Phosphodiesterase (PDE) inhibitors increase matrix metalloproteinase (MMP) production by inhibiting re-uptake of adenosine and may potentiate nitric oxide (NO) activity. This study was performed to investigate the effects and mechanisms of PDE inhibitors on trabecular outflow in cultured human trabecular meshwork cells (HTMCs). METHODS Primary HTMC cultures were exposed to 0, 20, and 50 μM dipyridamole (DPD) or theophylline (TPN). Permeability through the HTMC monolayer was assessed using carboxyfluorescein. The production of NO was assessed using the Griess assay and MMP-2 levels were measured via Western blotting. RESULTS DPD significantly increased permeability accompanied with increased nitrite concentration and MMP-2 levels (all p < 0.05). TPN increased nitrite but did not affect permeability or MMP-2 levels significantly (p > 0.05). When treated with DPD and TPN together, both permeability and nitrite production were increased; however, MMP-2 levels showed no difference compared to DPD exposure alone (p > 0.05). CONCLUSIONS DPD increased trabecular permeability accompanied with increased nitrite production and MMP-2 levels. PDE inhibitors may increase trabecular outflow by increasing MMP-2 levels and by potentiating NO activity through cyclic GMP in HTMC.
Collapse
Affiliation(s)
- Jae Woo Kim
- Department of Ophthalmology, Daegu Catholic University School of Medicine, Daegu, Korea.
| | - Jong Been Lee
- Department of Ophthalmology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - So Hyung Lee
- Department of Ophthalmology, Daegu Catholic University School of Medicine, Daegu, Korea
| |
Collapse
|
43
|
Clapp C, Diaz-Lezama N, Adan-Castro E, Ramirez-Hernandez G, Moreno-Carranza B, Sarti AC, Falzoni S, Solini A, Di Virgilio F. Pharmacological blockade of the P2X7 receptor reverses retinal damage in a rat model of type 1 diabetes. Acta Diabetol 2019; 56:1031-1036. [PMID: 30982154 DOI: 10.1007/s00592-019-01343-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/05/2019] [Indexed: 02/07/2023]
Abstract
AIMS Retinopathy is a leading cause of vision impairment in diabetes. Its pathogenesis involves inflammation, pathological angiogenesis, neuronal and glial dysfunction. The purinergic P2X7 receptor (P2X7R) has a leading role in inflammation and angiogenesis. Potent and selective P2X7R blockers have been synthesized and tested in Phase I/II clinical studies. We hypothesize that P2X7R blockade will ameliorate diabetes-related pathological retinal changes. METHODS Streptozotocin (STZ)-treated rats were intraperitoneally inoculated with either of two small molecule P2X7R receptor inhibitors, A740003 and AZ10606120, and after blood glucose levels increased to above 400 mg/dL, retinae were analyzed for P2X7R expression, vascular permeability, VEGF, and IL-6 expression. RESULTS STZ administration caused a near fourfold increase in blood glucose, a large increase in retinal microvasculature permeability, as well as in retinal P2X7R, VEGF, and IL-6 expression. P2X7R blockade fully reversed retinal vascular permeability increase, VEGF accumulation, and IL-6 expression, with no effect on blood glucose. CONCLUSION P2X7R blockade might be promising strategy for the treatment of microvascular changes observed in the early phases of diabetic retinopathy.
Collapse
Affiliation(s)
- Carmen Clapp
- Institute of Neurobiology, National University of Mexico [UNAM], Querétaro, Mexico
| | - Nundehui Diaz-Lezama
- Institute of Neurobiology, National University of Mexico [UNAM], Querétaro, Mexico
| | - Elva Adan-Castro
- Institute of Neurobiology, National University of Mexico [UNAM], Querétaro, Mexico
| | | | | | - Alba Clara Sarti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Borsari 46, 44121, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Borsari 46, 44121, Ferrara, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Via Borsari 46, 44121, Ferrara, Italy.
| |
Collapse
|
44
|
Mastrangelo M, Alfonsi C, Screpanti I, Masuelli L, Tavazzi B, Mei D, Giannotti F, Guerrini R, Leuzzi V. Broadening phenotype of adenylosuccinate lyase deficiency: A novel clinical pattern resembling neuronal ceroid lipofuscinosis. Mol Genet Metab Rep 2019; 21:100502. [PMID: 31467849 PMCID: PMC6713842 DOI: 10.1016/j.ymgmr.2019.100502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 11/30/2022] Open
Abstract
We describe a 7-year-old boy presenting with a developmental encephalopathy, severe epilepsy, retinopathy with salt and pepper fundus, and ultrastructural skin alterations resembling a neuronal ceroid lipofuscinosis. Whole exome-sequencing detected biallelic variants in the ADSL gene (c.65C > T [p.(Ala22Val)] and c.340 T > C [p.(Tyr114His)]). The increase of SAICAR and S-Ado in blood and urine was consistent with the pattern of adenylosuccinate lyase deficiency (OMIM 103050). An unusual increase of AICAR, that was due to a residual ADSL enzyme activity of about 28%, was also detected. Neither salt and pepper retinopathy nor ultrastructural skin alterations had been reported in ADSL deficiency before. Impaired purinergic signaling inside the retina is probably involved in visual failure. Ultrastructural alterations in fibroblasts suggest a possible damage of autophagic processes, whose role in the pathogenesis of neurological dysfunction deserves further study.
Collapse
Affiliation(s)
- Mario Mastrangelo
- Division of Child Neurology and Infantile Psychiatry, Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Chiara Alfonsi
- Division of Child Neurology and Infantile Psychiatry, Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | | | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Barbara Tavazzi
- Institute of Biochemistry and Clinical Biochemistry Catholic University, Rome
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Italy
| | - Davide Mei
- Neuroscience and Neurogenetics Excellence Centre, Children's Hospital Anna Meyer, Florence, Italy
| | - Flavia Giannotti
- Division of Child Neurology and Infantile Psychiatry, Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Renzo Guerrini
- Neuroscience and Neurogenetics Excellence Centre, Children's Hospital Anna Meyer, Florence, Italy
| | - Vincenzo Leuzzi
- Division of Child Neurology and Infantile Psychiatry, Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
- Corresponding author at: Division of Child Neurology and Infantile Psychiatry, Department of Human Neurosciences, Sapienza- University of Rome, Via dei Sabelli 108, 00141 Roma, Italy.
| |
Collapse
|
45
|
Blood-retinal barrier protection against high glucose damage: The role of P2X7 receptor. Biochem Pharmacol 2019; 168:249-258. [PMID: 31302133 DOI: 10.1016/j.bcp.2019.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022]
Abstract
Blood retinal barrier (BRB) breakdown is a hallmark of diabetic retinopathy, whose occurrence in early or later phases of the disease has not yet been completely clarified. Recent evidence suggests that hyperglycemia induces activation of the P2X7 receptor (P2X7R) leading to pericyte cell death. We herein investigated the role of P2X7R on retinal endothelial cells viability and expression of tight- and adherens-junctions following high glucose (HG) exposure. We found that HG elicited P2X7R activation and expression and release of the pro-inflammatory cytokine IL-1β in human retinal endothelial cells (HRECs). Furthermore, HG exposure caused a decrease in HRECs viability and a damage of the BRB. JNJ47965567, a P2X7R antagonist, protected HRECs from HG-induced damage (LDH release) and preserved the BRB, as shown by transendothelial electrical resistance and cell junction morphology (ZO-1, claudin-5 and VE-cadherin). Moreover, JNJ47965567 treatment significantly decreased IL-1β expression and release, elicited by HG. These data indicate that P2X7R plays an important role to regulate BRB integrity, in particular the block of this receptor was useful to counteract the damage elicited by HG in HRECs, and warranting further clinical evaluation of P2X7R antagonists for the treatment of diabetic macular edema.
Collapse
|
46
|
Lu W, Campagno KE, Tso HY, Cenaj A, Laties AM, Carlsson LG, Mitchell CH. Oral Delivery of the P2Y12 Receptor Antagonist Ticagrelor Prevents Loss of Photoreceptors in an ABCA4-/- Mouse Model of Retinal Degeneration. Invest Ophthalmol Vis Sci 2019; 60:3046-3053. [PMID: 31319418 PMCID: PMC6640265 DOI: 10.1167/iovs.19-27241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/10/2019] [Indexed: 01/22/2023] Open
Abstract
Purpose Accumulation of lysosomal waste is linked to neurodegeneration in multiple diseases, and pharmacologic enhancement of lysosomal activity is hypothesized to reduce pathology. An excessive accumulation of lysosomal-associated lipofuscin waste and an elevated lysosomal pH occur in retinal pigment epithelial cells of the ABCA4-/- mouse model of Stargardt's retinal degeneration. As treatment with the P2Y12 receptor antagonist ticagrelor was previously shown to lower lysosomal pH and lipofuscin-like autofluorescence in these cells, we asked whether oral delivery of ticagrelor also prevented photoreceptor loss. Methods Moderate light exposure was used to accelerate photoreceptor loss in albino ABCA4-/- mice as compared to BALB/c controls. Ticagrelor (0.1%-0.15%) was added to mouse chow for between 1 and 10 months. Photoreceptor function was determined with electroretinograms, while cell survival was determined using optical coherence tomography and histology. Results Protection by ticagrelor was demonstrated functionally by using the electroretinogram, as ticagrelor-treated ABCA4-/- mice had increased a- and b-waves compared to untreated mice. Mice receiving ticagrelor treatment had a thicker outer nuclear layer, as measured with both optical coherence tomography and histologic sections. Ticagrelor decreased expression of LAMP1, implicating enhanced lysosomal function. No signs of retinal bleeding were observed after prolonged treatment with ticagrelor. Conclusions Oral treatment with ticagrelor protected photoreceptors in the ABCA4-/- mouse, which is consistent with enhanced lysosomal function. As mouse ticagrelor exposure levels were clinically relevant, the drug may be of benefit in preventing the loss of photoreceptors in Stargardt's disease and other neurodegenerations associated with lysosomal dysfunction.
Collapse
Affiliation(s)
- Wennan Lu
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Keith E. Campagno
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Huen-Yee Tso
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Aurora Cenaj
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Alan M. Laties
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Leif G. Carlsson
- Bioscience Cardiovascular Research and Early Development Cardiovascular, Renal and Metabolism BioPhamaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Claire H. Mitchell
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
47
|
Fernández-Albarral JA, Salobrar-García E, Martínez-Páramo R, Ramírez AI, de Hoz R, Ramírez JM, Salazar JJ. Retinal glial changes in Alzheimer's disease - A review. JOURNAL OF OPTOMETRY 2019; 12:198-207. [PMID: 30377086 PMCID: PMC6612028 DOI: 10.1016/j.optom.2018.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 05/17/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative dementia characterized by the deposition of extracellular β-amyloid (Aβ) plaques and the presence of neurofibrillary tangles. Until now, the techniques used to analyze these deposits have been difficult to access, invasive, and expensive. This leads us to consider new access routes to the central nervous system (CNS), allowing us to diagnose the disease before the first symptoms appear. Recent studies have shown that microglial and macroglial cell activation could play a role in the development of this disease. Glial cells in the CNS can respond to various damages, such as neurodegenerative pathologies, with morphological and functional changes. These changes are a common feature in neurodegenerative diseases, including AD. The retina is considered an extension of the CNS and has a population of glial cells similar to that of the CNS. When glial cells are activated, various molecules are released and changes in glial cell expression occur, which can be indicators of neuronal damage. The objective of this review is to compile the most relevant findings in the last 10 years relating to alterations in the eye in AD, and the role that glial cells play in the degenerative process in the retina in the context of neurodegeneration.
Collapse
Affiliation(s)
- José A Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Spain
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Spain
| | - Rebeca Martínez-Páramo
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Spain
| | - Ana I Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Spain; Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Spain; Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, Spain
| | - José M Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Spain; Facultad de Medicina, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, Spain.
| | - Juan J Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid, Spain; Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, Spain.
| |
Collapse
|
48
|
Akhtar-Schäfer I, Wang L, Krohne TU, Xu H, Langmann T. Modulation of three key innate immune pathways for the most common retinal degenerative diseases. EMBO Mol Med 2019; 10:emmm.201708259. [PMID: 30224384 PMCID: PMC6180304 DOI: 10.15252/emmm.201708259] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review highlights the role of three key immune pathways in the pathophysiology of major retinal degenerative diseases including diabetic retinopathy, age‐related macular degeneration, and rare retinal dystrophies. We first discuss the mechanisms how loss of retinal homeostasis evokes an unbalanced retinal immune reaction involving responses of local microglia and recruited macrophages, activity of the alternative complement system, and inflammasome assembly in the retinal pigment epithelium. Presenting these key mechanisms as complementary targets, we specifically emphasize the concept of immunomodulation as potential treatment strategy to prevent or delay vision loss. Promising molecules are ligands for phagocyte receptors, specific inhibitors of complement activation products, and inflammasome inhibitors. We comprehensively summarize the scientific evidence for this strategy from preclinical animal models, human ocular tissue analyses, and clinical trials evolving in the last few years.
Collapse
Affiliation(s)
- Isha Akhtar-Schäfer
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Luping Wang
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Tim U Krohne
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Heping Xu
- Centre for Experimental Medicine, The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany .,Center for Molecular Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
49
|
Effect of Ocular Hypertension on D- β-Aspartic Acid-Containing Proteins in the Retinas of Rats. J Ophthalmol 2019; 2019:2431481. [PMID: 31240134 PMCID: PMC6556240 DOI: 10.1155/2019/2431481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/28/2019] [Accepted: 05/07/2019] [Indexed: 12/31/2022] Open
Abstract
Purpose To investigate the effect of ocular hypertension-induced isomerization of aspartic acid in retinal proteins. Methods Adult Wistar rats with ocular hypertension were used as an experimental model. D-β-aspartic acid-containing proteins were isolated by SDS-PAGE and western blot with an anti-D-β-aspartic acid antibody and identified by liquid chromatography-mass spectrometry analysis. The concentration of ATP was measured by ELISA. Results D-β-aspartic acid was expressed in a protein band at around 44.5 kDa at much higher quantities in the retinas of rats with ocular hypertension than in those of normotensive rats. The 44.5 kDa protein band was mainly composed of α-enolase, S-arrestin, and ATP synthase subunits α and β, in both the ocular hypertensive and normotensive retinas. Moreover, increasing intraocular pressure was correlated with increasing ATP concentrations in the retinas of rats. Conclusion Ocular hypertension affected the expression of proteins containing D-β-aspartic acid, including ATP synthase subunits, and up-regulation of ATP in the retinas of rats.
Collapse
|
50
|
Abstract
Adenylate kinase is a small, usually monomeric, enzyme found in every living thing due to its crucial role in energetic metabolism. This paper outlines the most relevant data about adenylate kinases isoforms, and the connection between dysregulation or mutation of human adenylate kinase and medical conditions. The following datadases were consulted: National Centre for Biotechnology Information, Protein Data Bank, and Mouse Genomic Informatics. The SmartBLAST tool, EMBOSS Needle Program, and Clustal Omega Program were used to analyze the best protein match, and to perform pairwise sequence alignment and multiple sequence alignment. Human adenylate kinase genes are located on different chromosomes, six of them being on the chromosomes 1 and 9. The adenylate kinases' intracellular localization and organ distribution explain their dysregulation in many diseases. The cytosolic isoenzyme 1 and the mitochondrial isoenzyme 2 are the main adenylate kinases that are integrated in the vast network of inflammatory modulators. The cytosolic isoenzyme 5 is correlated with limbic encephalitis and Leu673Pro mutation of the isoenzyme 7 leads to primary male infertility due to impairment of the ciliary function. The impairment of the mitochondrial isoenzymes 2 and 4 is demonstrated in neuroblastoma or glioma. The adenylate kinases are disease modifier that can assess the risk of diseases where oxidative stress plays a crucial role in pathogenesis like metabolic syndrome or neurodegenerative diseases. Because adenylate kinases has ATP as substrate, they are integrated in the global network of energetic process of any organism therefore are valid target for new pharmaceutical compounds.
Collapse
Affiliation(s)
- Mihaela Ileana Ionescu
- Department of Microbiology, Faculty of Medicine, Iuliu Haţieganu University of Medicine and Pharmacy, 6 Louis Pasteur, Cluj-Napoca, 400349, Romania.
- County Emergency Clinical Hospital, Cluj-Napoca, Romania.
| |
Collapse
|