1
|
Zhang L, Wu C, Liu T, Tian Y, Wang D, Wang B, Yin Y. Propofol Protects the Blood-Brain Barrier After Traumatic Brain Injury by Stabilizing the Extracellular Matrix via Prrx1: From Neuroglioma to Neurotrauma. Neurochem Res 2024; 49:2743-2762. [PMID: 38951281 DOI: 10.1007/s11064-024-04202-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
The purpose of this study is to explore the shared molecular pathogenesis of traumatic brain injury (TBI) and high-grade glioma and investigate the mechanism of propofol (PF) as a potential protective agent. By analyzing the Chinese glioma genome atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases, we compared the transcriptomic data of high-grade glioma and TBI patients to identify common pathological mechanisms. Through bioinformatics analysis, in vitro experiments and in vivo TBI model, we investigated the regulatory effect of PF on extracellular matrix (ECM)-related genes through Prrx1 under oxidative stress. The impact of PF on BBB integrity under oxidative stress was investigated using a dual-layer BBB model, and we explored the protective effect of PF on tight junction proteins and ECM-related genes in mice after TBI. The study found that high-grade glioma and TBI share ECM instability as an important molecular pathological mechanism. PF stabilizes the ECM and protects the BBB by directly binding to Prrx1 or indirectly regulating Prrx1 through miRNAs. In addition, PF reduces intracellular calcium ions and ROS levels under oxidative stress, thereby preserving BBB integrity. In a TBI mouse model, PF protected BBB integrity through up-regulated tight junction proteins and stabilized the expression of ECM-related genes. Our study reveals the shared molecular pathogenesis between TBI and glioblastoma and demonstrate the potential of PF as a protective agent of BBB. This provides new targets and approaches for the development of novel neurotrauma therapeutic drugs.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Chenrui Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Tian
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo Wang
- Department of Neurosurgery, Tianjin University Huanhu Hospital, Tianjin, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China.
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
2
|
Girkin CA, Strickland RG, Somerville MM, Anne Garner M, Grossman GH, Blake A, Kumar N, Ianov L, Fazio MA, Clark ME, Gross AK. Acute ocular hypertension in the living human eye: Model description and initial cellular responses to elevated intraocular pressure. Vision Res 2024; 223:108465. [PMID: 39173459 PMCID: PMC11444249 DOI: 10.1016/j.visres.2024.108465] [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: 06/05/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/24/2024]
Abstract
This initial methods study presents the initial immunohistochemical and transcriptomic changes in the optic nerve head and retina from three research-consented brain-dead organ donors following prolonged and transient intraocular pressure (IOP) elevation. In this initial study, research-consented brain-dead organ donors were exposed to unilateral elevation of IOP for 7.5 h (Donor 1), 30 h (Donor 2), and 1 h (Donor 3) prior to organ procurement. Optic nerve tissue and retinal tissue was obtained following organ procurement for immunohistological and transcriptomic analysis. Optic nerve sections in Donor 1 exposed to 7.5-hours of unilateral sub-ischemic IOP elevation demonstrated higher levels of protein expression of the astrocytic marker, glial fibrillary acidic protein (GFAP), within the lamina cribrosa with greatest expression inferior temporally in the treated eye compared to control. Spatial transcriptomic analysis performed on optic nerve head tissues from Donor 2 exposed to 30 h of unilateral IOP elevation demonstrated differential transcription of mRNA across laminar and scleral regions. Immunohistochemistry of retinal sections from Donor 2 exhibited higher GFAP and IBA1 expression in the treated eye compared with control, but this was not observed in Donor 3, which was exposed to only 1-hour of IOP elevation. While there were no differences in GFAP protein expression in the retina following the 1-hour IOP elevation in Donor 3, there were higher levels of transcription of GFAP in the inner nuclear layer, and CD44 in the retinal ganglion cell layer, indicative of astrocytic and Müller glial reactivity as well as an early inflammatory response, respectively. We found that transcriptomic differences can be observed across treated and control eyes following unilateral elevation of IOP in brain dead organ donors. The continued development of this model affords the unique opportunity to define the acute mechanotranscriptomic response of the optic nerve head, evaluate the injury and repair mechanisms in the retina in response to IOP elevation, and enable correlation of in vivo imaging and functional testing with ex vivo cellular responses for the first time in the living human eye.
Collapse
Affiliation(s)
- Christopher A Girkin
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ryan G Strickland
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - McKenna M Somerville
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mary Anne Garner
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Alan Blake
- Advancing Sight Network, Birmingham, AL, USA
| | - Nilesh Kumar
- IRCP-Biological Data Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lara Ianov
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; IRCP-Biological Data Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Massimo A Fazio
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mark E Clark
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alecia K Gross
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Waxman S, Schilpp H, Linton A, Jakobs TC, Sigal IA. Morphological comparison of astrocytes in the lamina cribrosa and glial lamina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.610493. [PMID: 39314351 PMCID: PMC11418941 DOI: 10.1101/2024.09.07.610493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Purpose Although the mechanisms underlying glaucomatous neurodegeneration are not yet well understood, cellular and small animal models suggest that LC astrocytes undergo early morphologic and functional changes, indicating their role as early responders to glaucomatous stress. These models, however, lack the LC found in larger animals and humans, leaving the in situ morphology of LC astrocytes and their role in glaucoma initiation underexplored. In this work, we aimed to characterize the morphology of LC astrocytes in situ and determine differences and similarities with astrocytes in the mouse glial lamina (GL), the analogous structure in a prominent glaucoma model. Methods Astrocytes in the LCs of twenty-two eyes from goats, sheep, and pigs were stochastically labeled via Multicolor DiOlistics and imaged in situ using confocal microscopy. 3D models of DiOlistically-labeled LC astrocytes and hGFAPpr-GFP mouse GL astrocytes were constructed to quantify morphological features related to astrocyte functions. LC and GL astrocyte cross-pore contacts, branching complexity, branch tortuosity, and cell and branch span were compared. Results LC astrocytes displayed distinct spatial relationships with collagen, greater branching complexity, and higher branch tortuosity compared to GL astrocytes. Despite substantial differences in their anatomical environments, LC and GL astrocytes had similar cell and branch spans. Conclusions Astrocyte morphology in the LC was characterized through Multicolor DiOlistic labeling. LC and GL astrocytes have both distinct and shared morphological features. Further research is needed to understand the potentially unique roles of LC astrocytes in glaucoma initiation and progression.
Collapse
Affiliation(s)
- Susannah Waxman
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Hannah Schilpp
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Ashley Linton
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
| | - Tatjana C. Jakobs
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary/Schepens Eye Research Institute, Harvard Medical School, Boston, MA, USA
| | - Ian A. Sigal
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh PA, USA
| |
Collapse
|
4
|
Gregucci F, Beal K, Knisely JPS, Pagnini P, Fiorentino A, Bonzano E, Vanpouille-Box CI, Cisse B, Pannullo SC, Stieg PE, Formenti SC. Biological Insights and Radiation-Immuno-Oncology Developments in Primary and Secondary Brain Tumors. Cancers (Basel) 2024; 16:2047. [PMID: 38893165 PMCID: PMC11171192 DOI: 10.3390/cancers16112047] [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: 05/08/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Malignant central nervous system (CNS) cancers include a group of heterogeneous dis-eases characterized by a relative resistance to treatments and distinguished as either primary tumors arising in the CNS or secondary tumors that spread from other organs into the brain. Despite therapeutic efforts, they often cause significant mortality and morbidity across all ages. Radiotherapy (RT) remains the main treatment for brain cancers, improving associated symptoms, improving tumor control, and inducing a cure in some. However, the ultimate goal of cancer treatment, to improve a patient's survival, remains elusive for many CNS cancers, especially primary tumors. Over the years, there have thus been many preclinical studies and clinical trials designed to identify and overcome mechanisms of resistance to improve outcomes after RT and other therapies. For example, immunotherapy delivered concurrent with RT, especially hypo-fractionated stereotactic RT, is synergistic and has revolutionized the clinical management and outcome of some brain tumors, in particular brain metastases (secondary brain tumors). However, its impact on gliomas, the most common primary malignant CNS tumors, remains limited. In this review, we provide an overview of radioresistance mechanisms, the emerging strategies to overcome radioresistance, the role of the tumor microenviroment (TME), and the selection of the most significant results of radiation-immuno-oncological investigations. We also identify novel therapeutic opportunities in primary and secondary brain tumors with the purpose of elucidating current knowledge and stimulating further research to improve tumor control and patients' survival.
Collapse
Affiliation(s)
- Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
- Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
| | - Kathryn Beal
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
| | - Jonathan P. S. Knisely
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
| | - Paul Pagnini
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
| | - Alba Fiorentino
- Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, 70021 Bari, Italy;
- Department of Medicine and Surgery, LUM University, Casamassima, 70010 Bari, Italy
| | - Elisabetta Bonzano
- Department of Radiation Oncology, IRCCS San Matteo Polyclinic Foundation, 27100 Pavia, Italy;
| | - Claire I. Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| | - Babacar Cisse
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (B.C.); (S.C.P.); (P.E.S.)
| | - Susan C. Pannullo
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (B.C.); (S.C.P.); (P.E.S.)
- Department of Biomedical Engineering, College of Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Philip E. Stieg
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY 10065, USA; (B.C.); (S.C.P.); (P.E.S.)
- Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA; (F.G.); (K.B.); (J.P.S.K.); (P.P.); (C.I.V.-B.)
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| |
Collapse
|
5
|
Fernández-Albarral JA, Ramírez AI, de Hoz R, Matamoros JA, Salobrar-García E, Elvira-Hurtado L, López-Cuenca I, Sánchez-Puebla L, Salazar JJ, Ramírez JM. Glaucoma: from pathogenic mechanisms to retinal glial cell response to damage. Front Cell Neurosci 2024; 18:1354569. [PMID: 38333055 PMCID: PMC10850296 DOI: 10.3389/fncel.2024.1354569] [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: 12/12/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Glaucoma is a neurodegenerative disease of the retina characterized by the irreversible loss of retinal ganglion cells (RGCs) leading to visual loss. Degeneration of RGCs and loss of their axons, as well as damage and remodeling of the lamina cribrosa are the main events in the pathogenesis of glaucoma. Different molecular pathways are involved in RGC death, which are triggered and exacerbated as a consequence of a number of risk factors such as elevated intraocular pressure (IOP), age, ocular biomechanics, or low ocular perfusion pressure. Increased IOP is one of the most important risk factors associated with this pathology and the only one for which treatment is currently available, nevertheless, on many cases the progression of the disease continues, despite IOP control. Thus, the IOP elevation is not the only trigger of glaucomatous damage, showing the evidence that other factors can induce RGCs death in this pathology, would be involved in the advance of glaucomatous neurodegeneration. The underlying mechanisms driving the neurodegenerative process in glaucoma include ischemia/hypoxia, mitochondrial dysfunction, oxidative stress and neuroinflammation. In glaucoma, like as other neurodegenerative disorders, the immune system is involved and immunoregulation is conducted mainly by glial cells, microglia, astrocytes, and Müller cells. The increase in IOP produces the activation of glial cells in the retinal tissue. Chronic activation of glial cells in glaucoma may provoke a proinflammatory state at the retinal level inducing blood retinal barrier disruption and RGCs death. The modulation of the immune response in glaucoma as well as the activation of glial cells constitute an interesting new approach in the treatment of glaucoma.
Collapse
Affiliation(s)
- Jose A. Fernández-Albarral
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Ana I. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José A. Matamoros
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Lidia Sánchez-Puebla
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Juan J. Salazar
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid (UCM), Grupo UCM 920105, IdISSC, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
6
|
Hörberg CJ, Englund Johansson U, Johansson F, O'Carroll D. Spontaneous Cell Cluster Formation in Human iPSC-Derived Neuronal Spheroid Networks Influences Network Activity. eNeuro 2022; 9:ENEURO.0143-22.2022. [PMID: 36216508 PMCID: PMC9581577 DOI: 10.1523/eneuro.0143-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/07/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Three-dimensional neuronal culture systems such as spheroids, organoids, and assembloids constitute a branch of neuronal tissue engineering that has improved our ability to model the human brain in the laboratory. However, the more elaborate the brain model, the more difficult it becomes to study functional properties such as electrical activity at the neuronal level, similar to the challenges of studying neurophysiology in vivo We describe a simple approach to generate self-assembled three-dimensional neuronal spheroid networks with defined human cell composition on microelectrode arrays. Such spheroid networks develop a highly three-dimensional morphology with cell clusters up to 60 µm in thickness and are interconnected by pronounced bundles of neuronal fibers and glial processes. We could reliably record from up to hundreds of neurons simultaneously per culture for ≤90 d. By quantifying the formation of these three-dimensional structures over time, while regularly monitoring electrical activity, we were able to establish a strong link between spheroid morphology and network activity. In particular, the formation of cell clusters accelerates formation and maturation of correlated network activity. Astrocytes both influence electrophysiological network activity as well as accelerate the transition from single cell layers to cluster formation. Higher concentrations of astrocytes also have a strong effect of modulating synchronized network activity. This approach thus represents a practical alternative to often complex and heterogeneous organoids, providing easy access to activity within a brain-like 3D environment.Significance StatementNeuronal "organoid" cultures with multiple cell types grown on elaborate three-dimensional scaffolds have become popular tools to generate brain-like properties in vitro but bring with them similar problems concerning access to physiological function as real brain tissue. Here, we developed a new approach to form simple brain-like spheroid networks from human neurons, but using the normal supporting cells of the brain, astrocytes, as the scaffold. By growing these cultures on conventional microelectrode arrays, we were able to observe development of complex patterns of electrical activity for months. Our results highlight how formation of three-dimensional structures accelerated the formation of synchronized neuronal network activity and provide a promising new simple model system for studying interactions between known human cell types in vitro.
Collapse
|
7
|
Strickland RG, Garner MA, Gross AK, Girkin CA. Remodeling of the Lamina Cribrosa: Mechanisms and Potential Therapeutic Approaches for Glaucoma. Int J Mol Sci 2022; 23:8068. [PMID: 35897642 PMCID: PMC9329908 DOI: 10.3390/ijms23158068] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022] Open
Abstract
Glaucomatous optic neuropathy is the leading cause of irreversible blindness in the world. The chronic disease is characterized by optic nerve degeneration and vision field loss. The reduction of intraocular pressure remains the only proven glaucoma treatment, but it does not prevent further neurodegeneration. There are three major classes of cells in the human optic nerve head (ONH): lamina cribrosa (LC) cells, glial cells, and scleral fibroblasts. These cells provide support for the LC which is essential to maintain healthy retinal ganglion cell (RGC) axons. All these cells demonstrate responses to glaucomatous conditions through extracellular matrix remodeling. Therefore, investigations into alternative therapies that alter the characteristic remodeling response of the ONH to enhance the survival of RGC axons are prevalent. Understanding major remodeling pathways in the ONH may be key to developing targeted therapies that reduce deleterious remodeling.
Collapse
Affiliation(s)
- Ryan G. Strickland
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Mary Anne Garner
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Alecia K. Gross
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.G.S.); (M.A.G.); (A.K.G.)
| | - Christopher A. Girkin
- Department of Ophthalmology and Vision Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
8
|
Molecular regulation of neuroinflammation in glaucoma: Current knowledge and the ongoing search for new treatment targets. Prog Retin Eye Res 2022; 87:100998. [PMID: 34348167 PMCID: PMC8803988 DOI: 10.1016/j.preteyeres.2021.100998] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022]
Abstract
Neuroinflammation relying on the inflammatory responses of glial cells has emerged as an impactful component of the multifactorial etiology of neurodegeneration in glaucoma. It has become increasingly evident that despite early adaptive and reparative features of glial responses, prolonged reactivity of the resident glia, along with the peripheral immune cells, create widespread toxicity to retinal ganglion cell (RGC) axons, somas, and synapses. As much as the synchronized responses of astrocytes and microglia to glaucoma-related stress or neuron injury, their bi-directional interactions are critical to build and amplify neuroinflammation and to dictate the neurodegenerative outcome. Although distinct molecular programs regulate somatic and axonal degeneration in glaucoma, inhibition of neurodegenerative inflammation can provide a broadly beneficial treatment strategy to rescue RGC integrity and function. Since inflammatory toxicity and mitochondrial dysfunction are converging etiological paths that can boost each other and feed into a vicious cycle, anti-inflammatory treatments may also offer a multi-target potential. This review presents an overview of the current knowledge on neuroinflammation in glaucoma with particular emphasis on the cell-intrinsic and cell-extrinsic factors involved in the reciprocal regulation of glial responses, the interdependence between inflammatory and mitochondrial routes of neurodegeneration, and the research aspects inspiring for prospective immunomodulatory treatments. With the advent of powerful technologies, ongoing research on molecular and functional characteristics of glial responses is expected to accumulate more comprehensive and complementary information and to rapidly move the field forward to safe and effective modulation of the glial pro-inflammatory activities, while restoring or augmenting the glial immune-regulatory and neurosupport functions.
Collapse
|
9
|
Kirschner A, Strat AN, Yablonski J, Yoo H, Bagué T, Li H, Zhao J, Bollinger KE, Herberg S, Ganapathy PS. Mechanosensitive channel inhibition attenuates TGFβ2-induced actin cytoskeletal remodeling and reactivity in mouse optic nerve head astrocytes. Exp Eye Res 2021; 212:108791. [PMID: 34656548 DOI: 10.1016/j.exer.2021.108791] [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: 06/07/2021] [Revised: 09/22/2021] [Accepted: 10/11/2021] [Indexed: 11/27/2022]
Abstract
Astrocytes within the optic nerve head undergo actin cytoskeletal rearrangement early in glaucoma, which coincides with astrocyte reactivity and extracellular matrix (ECM) deposition. Elevated transforming growth factor beta 2 (TGFβ2) levels within astrocytes have been described in glaucoma, and TGFβ signaling induces actin cytoskeletal remodeling and ECM deposition in many tissues. A key mechanism by which astrocytes sense and respond to external stimuli is via mechanosensitive ion channels. Here, we tested the hypothesis that inhibition of mechanosensitive channels will attenuate TGFβ2-mediated optic nerve head astrocyte actin cytoskeletal remodeling, reactivity, and ECM deposition. Primary optic nerve head astrocytes were isolated from C57BL/6J mice and cell purity was confirmed by immunostaining. Astrocytes were treated with vehicle control, TGFβ2 (5 ng/ml), GsMTx4 (a mechanosensitive channel inhibitor; 500 nM), or TGFβ2 (5 ng/ml) + GsMTx4 (500 nM) for 48 h. FITC-phalloidin staining was used to assess the formation of f-actin stress fibers and to quantify the presence of crosslinked actin networks (CLANs). Cell reactivity was determined by immunostaining and immunoblotting for GFAP. Levels of fibronectin and collagen IV deposition were also quantified. Primary optic nerve head astrocytes were positive for the astrocyte marker GFAP and negative for markers for microglia (F4/80) and oligodendrocytes (OSP1). Significantly increased %CLAN-positive cells were observed after 48-h treatment with TGFβ2 vs. control in a dose-dependent manner. Co-treatment with GsMTx4 significantly decreased %CLAN-positive cells vs. TGFβ2 treatment and the presence of f-actin stress fibers. TGFβ2 treatment significantly increased GFAP, fibronectin, and collagen IV levels, and GsMTx4 co-treatment ameliorated GFAP immunoreactivity. Our data suggest inhibition of mechanosensitive channel activity as a potential therapeutic strategy to modulate actin cytoskeletal remodeling within the optic nerve head in glaucoma.
Collapse
Affiliation(s)
- Alexander Kirschner
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Ana N Strat
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - John Yablonski
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Hannah Yoo
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Tyler Bagué
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Haiyan Li
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Jing Zhao
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta, GA, 30912, USA
| | - Kathryn E Bollinger
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA; Culver Vision Discovery Institute, Augusta, GA, 30912, USA
| | - Samuel Herberg
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
| | - Preethi S Ganapathy
- Department of Ophthalmology & Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA.
| |
Collapse
|
10
|
VanderWall KB, Lu B, Alfaro JS, Allsop AR, Carr AS, Wang S, Meyer JS. Differential susceptibility of retinal ganglion cell subtypes in acute and chronic models of injury and disease. Sci Rep 2020; 10:17359. [PMID: 33060618 PMCID: PMC7566630 DOI: 10.1038/s41598-020-71460-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
Retinal ganglion cells (RGCs) are a heterogeneous population of neurons, comprised of numerous subtypes that work synchronously to transmit visual information to the brain. In blinding disorders such as glaucoma, RGCs are the main cell type to degenerate and lead to loss of vision. Previous studies have identified and characterized a variety of RGC subtypes in animal models, although only a handful of studies demonstrate the differential loss of these RGC subtypes in response to disease or injury. Thus, efforts of the current study utilized both chronic (bead occlusion) and acute (optic nerve crush, ONC) rat models to characterize disease response and differential loss of RGC subtypes. Bead occlusion and ONC retinas demonstrated significant RGC loss, glial reactivity and apoptosis compared to control retinas. Importantly, bead occlusion and ONC retinas resulted in differential subtype-specific loss of RGCs, with a high susceptibility for alpha- and direction selective-RGCs and preferential survival of ipRGCs. Results of this study serve as an important foundation for future experiments focused on the mechanisms resulting in the loss of RGCs in optic neuropathies, as well as the development of targeted therapeutics for RGC subtype-specific neuroprotection.
Collapse
Affiliation(s)
- Kirstin B VanderWall
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Bin Lu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Regenerative Medicine Institute, Los Angeles, CA, 90048, USA
| | - Jorge S Alfaro
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Regenerative Medicine Institute, Los Angeles, CA, 90048, USA
| | - Anna R Allsop
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Alexa S Carr
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Shaomei Wang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Regenerative Medicine Institute, Los Angeles, CA, 90048, USA.
| | - Jason S Meyer
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Deparment of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
11
|
Nafian F, Kamali Doust Azad B, Yazdani S, Rasaee MJ, Daftarian N. A lab-on-a-chip model of glaucoma. Brain Behav 2020; 10:e01799. [PMID: 32803874 PMCID: PMC7559618 DOI: 10.1002/brb3.1799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 06/14/2020] [Accepted: 07/28/2020] [Indexed: 01/13/2023] Open
Abstract
AIMS We developed a glaucoma-on-a-chip model to evaluate the viability of retinal ganglion cells (RGCs) against high pressure and the potential effect of neuroprotection. METHODS A three-layered chip consisting of interconnecting microchannels and culture wells was designed and fabricated from poly-methyl methacrylate sheets. The bottom surface of the wells was modified by air plasma and coated with different membranes to provide a suitable extracellular microenvironment. RGCs were purified from postnatal Wistar rats by magnetic assisted cell sorting up to 70% and characterized by flow cytometry and immunocytochemistry. The cultured RGCs were exposed to normal (15 mmHg) or elevated pressure (33 mmHg) for 6, 12, 24, 36, and 48 hr, with and without adding brain-derived neurotrophic factor (BDNF) or a novel BDNF mimetic (RNYK). RESULTS Multiple inlet ports allow culture media and gas into the wells under elevated hydrostatic pressure. PDL/laminin formed the best supporting membrane. RGC survival rates were 85%, 78%, 70%, 67%, and 61% under normal pressure versus 40%, 22%, 18%, 12%, and 10% under high pressure at 6, 12, 24, 36, and 48 hr, respectively. BDNF and RNYK separately reduced RGC death rates about twofold under both normal and elevated pressures. CONCLUSION This model recapitulated the effects of elevated pressure over relatively short time periods and demonstrated the neuroprotective effects of BDNF and RNYK.
Collapse
Affiliation(s)
- Fatemeh Nafian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Babak Kamali Doust Azad
- Department of Electronics, School of Electrical and Computer Engineering, Tehran University, Tehran, Iran
| | - Shahin Yazdani
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narsis Daftarian
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Pustchi SE, Avci NG, Akay YM, Akay M. Astrocytes Decreased the Sensitivity of Glioblastoma Cells to Temozolomide and Bay 11-7082. Int J Mol Sci 2020; 21:E7154. [PMID: 32998285 PMCID: PMC7583902 DOI: 10.3390/ijms21197154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant type of astrocytic tumors. GBM patients have a poor prognosis with a median survival of approximately 15 months despite the "Stupp" Regimen and high tumor recurrence due to the tumor resistance to chemotherapy. In this study, we co-cultured GBM cells with human astrocytes in three-dimensional (3D) poly(ethylene glycol) dimethyl acrylate (PEGDA) microwells to mimic the tumor microenvironment. We treated 3D co- and mono-cultured cells with Temozolomide (TMZ) and the nuclear factor-κB (NF-κB) inhibitor Bay 11-7082 and investigated the combined effect of the drugs. We assessed the expressions of glial fibrillary acidic protein (GFAP) and vimentin that play a role in the tumor malignancy and activation of the astrocytes as well as Notch-1 and survivin that play a role in GBM malignancy after the drug treatment to understand how astrocytes induced GBM drug response. Our results showed that in the co-culture, astrocytes increased GBM survival and resistance after combined drug treatment compared to mono-cultures. These data restated the importance of 3D cell culture to mimic the tumor microenvironment for drug screening.
Collapse
MESH Headings
- Antineoplastic Agents, Alkylating/pharmacology
- Astrocytes/cytology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell Survival/genetics
- Coculture Techniques/methods
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Glial Fibrillary Acidic Protein/genetics
- Glial Fibrillary Acidic Protein/metabolism
- Humans
- Models, Biological
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neuroglia/pathology
- Nitriles/pharmacology
- Primary Cell Culture
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Signal Transduction
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Sulfones/pharmacology
- Survivin/genetics
- Survivin/metabolism
- Temozolomide/pharmacology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Vimentin/genetics
- Vimentin/metabolism
Collapse
Affiliation(s)
| | | | | | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (S.E.P.); (N.G.A.); (Y.M.A.)
| |
Collapse
|
13
|
Lee EJ, Han JC, Park DY, Kee C. A neuroglia-based interpretation of glaucomatous neuroretinal rim thinning in the optic nerve head. Prog Retin Eye Res 2020; 77:100840. [PMID: 31982595 DOI: 10.1016/j.preteyeres.2020.100840] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/02/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Neuroretinal rim thinning (NRR) is a characteristic glaucomatous optic disc change. However, the precise mechanism of the rim thinning has not been completely elucidated. This review focuses on the structural role of the glioarchitecture in the formation of the glaucomatous NRR thinning. The NRR is a glia-framed structure, with honeycomb geometry and mechanically reinforced astrocyte processes along the transverse plane. When neural damage selectively involves the neuron and spares the glia, the gross structure of the tissue is preserved. The disorganization and loss of the glioarchitecture are the two hallmarks of optic nerve head (ONH) remodeling in glaucoma that leads to the thinning of NRR tissue upon axonal loss. This is in contrast to most non-glaucomatous optic neuropathies with optic disc pallor where hypertrophy of the glioarchitecture is associated with the seemingly absent optic disc cupping. Arteritic anterior ischemic optic neuropathy is an exception where pan-necrosis of ONH tissue leads to NRR thinning. Milder ischemia indicates selective neuronal loss that spares glia in non-arteritic anterior ischemic optic neuropathy. The biological reason is the heterogeneous glial response determined by the site, type, and severity of the injury. The neuroglial interpretation explains how the cellular changes underlie the clinical findings. Updated understandings on glial responses illustrate the mechanical, microenvironmental, and microglial modulation of activated astrocytes in glaucoma. Findings relevant to the possible mechanism of the astrocyte death in advanced glaucoma are also emerging. Ultimately, a better understanding of glaucomatous glial response may lead to glia-targeting neuroprotection in the future.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Jong Chul Han
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Do Young Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Changwon Kee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
14
|
Fachel FNS, Dal Prá M, Azambuja JH, Endres M, Bassani VL, Koester LS, Henriques AT, Barschak AG, Teixeira HF, Braganhol E. Glioprotective Effect of Chitosan-Coated Rosmarinic Acid Nanoemulsions Against Lipopolysaccharide-Induced Inflammation and Oxidative Stress in Rat Astrocyte Primary Cultures. Cell Mol Neurobiol 2020; 40:123-139. [PMID: 31446560 DOI: 10.1007/s10571-019-00727-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/12/2019] [Indexed: 01/11/2023]
Abstract
Rosmarinic acid (RA) is a natural polyphenolic compound with a well-documented neuroprotective effect mainly associated with its anti-inflammatory and antioxidant activities. Recently, our research group developed and optimized chitosan-coated RA nanoemulsions (RA CNE) intended to be used for nasal delivery as a new potential neuroprotective therapy. In this sense, the present study aimed to evaluate the protective and/or therapeutic potential of RA CNE in inflammation/oxidative stress induced by LPS (1 μg mL-1) in rat astrocyte primary cultures. In summary, pre-treatment with RA CNE before exposure to LPS (protective protocol) reduced significantly the LPS-induced alterations in astrocyte cell viability, proliferation, and cell death by necrosis, which was not observed in therapeutic protocol. RA CNE protective protocol also enhanced anti-oxidative status by ~ 50% by decreasing oxygen reactive species production and nitric oxide levels and preventing total thiol content decrease. Finally, our results demonstrate the protective effect of RA CNE in migratory activation and GFAP expression of reactive astrocytes. Overall, our findings indicate for the first time the RA CNE glioprotective potential, associated with an increase in cell viability and proliferation, a preventive effect on cellular death by necrosis, migratory ability and hypertrophic reactive astrocytes, and the reparation of astrocyte redox state.
Collapse
Affiliation(s)
- Flávia Nathiely Silveira Fachel
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Morgana Dal Prá
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Juliana Hofstätter Azambuja
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Marcelo Endres
- Programa de Pós-Graduação em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Valquíria Linck Bassani
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Letícia Scherer Koester
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Amelia Teresinha Henriques
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alethea Gatto Barschak
- Programa de Pós-Graduação em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Helder Ferreira Teixeira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Elizandra Braganhol
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
15
|
Tehrani S, Davis L, Cepurna WO, Delf RK, Lozano DC, Choe TE, Johnson EC, Morrison JC. Optic Nerve Head Astrocytes Display Axon-Dependent and -Independent Reactivity in Response to Acutely Elevated Intraocular Pressure. Invest Ophthalmol Vis Sci 2019; 60:312-321. [PMID: 30665231 PMCID: PMC6343680 DOI: 10.1167/iovs.18-25447] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Purpose Optic nerve head (ONH) astrocytes provide support for axons, but exhibit structural and functional changes (termed reactivity) in a number of glaucoma models. The purpose of this study was to determine if ONH astrocyte structural reactivity is axon-dependent. Methods Using rats, we combine retrobulbar optic nerve transection (ONT) with acute controlled elevation of intraocular pressure (CEI), to induce total optic nerve axon loss and ONH astrocyte reactivity, respectively. Animals were euthanized immediately or 1 day post CEI, in the presence or absence of ONT. ONH sections were labeled with fluorescent-tagged phalloidin and antibodies against β3 tubulin, phosphorylated cortactin, phosphorylated paxillin, or complement C3. ONH label intensities were quantified after confocal microscopy. Retrobulbar nerves were assessed for axon injury by light microscopy. Results While ONT alone had no effect on ONH astrocyte structural orientation, astrocytes demonstrated significant reorganization of cellular extensions within hours after CEI, even when combined with ONT. However, ONH astrocytes displayed differential intensities of actin (phosphorylated cortactin) and focal adhesion (phosphorylated paxillin) mediators in response to CEI alone, ONT alone, or the combination of CEI and ONT. Lastly, label intensities of complement C3 within the ONH were unchanged in eyes subjected to CEI alone, ONT alone, or the combination of CEI and ONT, relative to controls. Conclusions Early ONH astrocyte structural reactivity to elevated IOP is multifaceted, displaying both axon dependent and independent responses. These findings have important implications for pursuing astrocytes as diagnostic and therapeutic targets in neurodegenerative disorders with fluctuating levels of axon injury.
Collapse
Affiliation(s)
- Shandiz Tehrani
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| | - Lauren Davis
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| | - William O Cepurna
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| | - R Katherine Delf
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| | - Diana C Lozano
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| | - Tiffany E Choe
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| | - Elaine C Johnson
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| | - John C Morrison
- Casey Eye Institute, Department of Ophthalmology, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
16
|
Vroemen PAMM, Gorgels TGMF, Webers CAB, de Boer J. Modeling the Mechanical Parameters of Glaucoma. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:412-428. [PMID: 31088331 DOI: 10.1089/ten.teb.2019.0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Glaucoma is a major eye disease characterized by a progressive loss of retinal ganglion cells (RGCs). Biomechanical forces as a result of hydrostatic pressure and strain play a role in this disease. Decreasing intraocular pressure is the only available therapy so far, but is not always effective and does not prevent blindness in many cases. There is a need for drugs that protect RGCs from dying in glaucoma; to develop these, we need valid glaucoma and drug screening models. Since in vivo models are unsuitable for screening purposes, we focus on in vitro and ex vivo models in this review. Many groups have studied pressure and strain model systems to mimic glaucoma, to investigate the molecular and cellular events leading to mechanically induced RGC death. Therefore, the focus of this review is on the different mechanical model systems used to mimic the biomechanical forces in glaucoma. Most models use either cell or tissue strain, or fluid- or gas-controlled hydrostatic pressure application and apply it to the relevant cell types such as trabecular meshwork cells, optic nerve head astrocytes, and RGCs, but also to entire eyes. New model systems are warranted to study concepts and test experimental compounds for the development of new drugs to protect vision in glaucoma patients. Impact Statement The outcome of currently developed models to investigate mechanically induced retinal ganglion cell death by applying different mechanical strains varies widely. This suggests that a robust glaucoma model has not been developed yet. However, a comprehensive overview of current developments is not available. In this review, we have therefore assessed what has been done before and summarized the available knowledge in the field, which can be used to develop improved models for glaucoma research.
Collapse
Affiliation(s)
- Pascal A M M Vroemen
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Complex Tissue Regeneration (CTR), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Theo G M F Gorgels
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Carroll A B Webers
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Institute for Complex Molecular Structures, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
17
|
Lauzi J, Anders F, Liu H, Pfeiffer N, Grus F, Thanos S, Arnhold S, Prokosch V. Neuroprotective and neuroregenerative effects of CRMP-5 on retinal ganglion cells in an experimental in vivo and in vitro model of glaucoma. PLoS One 2019; 14:e0207190. [PMID: 30673694 PMCID: PMC6343933 DOI: 10.1371/journal.pone.0207190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/28/2018] [Indexed: 01/15/2023] Open
Abstract
Purpose To analyze the potential neuro-protective and neuro-regenerative effects of Collapsin-response-mediator-protein-5 (CRMP-5) on retinal ganglion cells (RGCs) using in vitro and in vivo animal models of glaucoma. Methods Elevated intraocular pressure (IOP) was induced in adult female Sprague-Dawley (SD) rats by cauterization of three episcleral veins. Changes in CRMP-5 expression within the retinal proteome were analyzed via label-free mass spectrometry. In vitro, retinal explants were cultured under elevated pressure (60 mmHg) within a high-pressure incubation chamber with and without addition of different concentrations of CRMP-5 (4 μg/l, 200 μg/l and 400 μg/l). In addition, retinal explants were cultured under regenerative conditions with and without application of 200 μg/l CRMP-5 after performing an optic nerve crush (ONC). Thirdly, an antibody against Protein Kinase B (PKB) was added to examine the possible effects of CRMP-5. RGC count was performed. Number and length of the axons were determined and compared. To undermine a signal-transduction pathway via CRMP-5 and PKB microarray and immunohistochemistry were performed. Results CRMP-5 was downregulated threefold in animals showing chronically elevated IOP. The addition of CRMP-5 to retinal culture significantly increased RGC numbers under pressure in a dose-dependent manner and increased and elongated outgrowing axons in retinal explants significantly which could be blocked by PKB. Especially the number of neurites longer than 400 μm significantly increased after application of CRMP-5. CRMP-5 as well as PKB were detected higher in the experimental than in the control group. Conclusion CRMP-5 seems to play an important role in an animal model of glaucoma. Addition of CRMP-5 exerts neuro-protective and neuro-regenerative effects in vitro. This effect could be mediated via activation of PKB affecting intra-cellular apoptosis pathways.
Collapse
Affiliation(s)
- Jasmin Lauzi
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Fabian Anders
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Hanhan Liu
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Norbert Pfeiffer
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Franz Grus
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
| | - Solon Thanos
- Department of Experimental Ophthalmology, School of Medicine, University of Münster, Münster, Germany
| | - Stefan Arnhold
- Institute of Veterinary-Anatomy, -Histology and–Embryology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Verena Prokosch
- Experimental Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz
- * E-mail:
| |
Collapse
|
18
|
Ma K, Deng X, Xia X, Fan Z, Qi X, Wang Y, Li Y, Ma Y, Chen Q, Peng H, Ding J, Li C, Huang Y, Tian C, Zheng JC. Direct conversion of mouse astrocytes into neural progenitor cells and specific lineages of neurons. Transl Neurodegener 2018; 7:29. [PMID: 30410751 PMCID: PMC6217767 DOI: 10.1186/s40035-018-0132-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/11/2018] [Indexed: 12/26/2022] Open
Abstract
Background Cell replacement therapy has been envisioned as a promising treatment for neurodegenerative diseases. Due to the ethical concerns of ESCs-derived neural progenitor cells (NPCs) and tumorigenic potential of iPSCs, reprogramming of somatic cells directly into multipotent NPCs has emerged as a preferred approach for cell transplantation. Methods Mouse astrocytes were reprogrammed into NPCs by the overexpression of transcription factors (TFs) Foxg1, Sox2, and Brn2. The generation of subtypes of neurons was directed by the force expression of cell-type specific TFs Lhx8 or Foxa2/Lmx1a. Results Astrocyte-derived induced NPCs (AiNPCs) share high similarities, including the expression of NPC-specific genes, DNA methylation patterns, the ability to proliferate and differentiate, with the wild type NPCs. The AiNPCs are committed to the forebrain identity and predominantly differentiated into glutamatergic and GABAergic neuronal subtypes. Interestingly, additional overexpression of TFs Lhx8 and Foxa2/Lmx1a in AiNPCs promoted cholinergic and dopaminergic neuronal differentiation, respectively. Conclusions Our studies suggest that astrocytes can be converted into AiNPCs and lineage-committed AiNPCs can acquire differentiation potential of other lineages through forced expression of specific TFs. Understanding the impact of the TF sets on the reprogramming and differentiation into specific lineages of neurons will provide valuable strategies for astrocyte-based cell therapy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Kangmu Ma
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Xiaobei Deng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xiaohuan Xia
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Zhaohuan Fan
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xinrui Qi
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yongxiang Wang
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Yuju Li
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Yizhao Ma
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Qiang Chen
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Hui Peng
- 3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Jianqing Ding
- 4Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Chunhong Li
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yunlong Huang
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Changhai Tian
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Jialin C Zheng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,2Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA.,5Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| |
Collapse
|
19
|
Zhang X, Ding K, Wang J, Li X, Zhao P. Chemoresistance caused by the microenvironment of glioblastoma and the corresponding solutions. Biomed Pharmacother 2018; 109:39-46. [PMID: 30391707 DOI: 10.1016/j.biopha.2018.10.063] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary human brain tumor. Although comprehensive therapies combining radiotherapy and chemotherapy after surgery can prolong survival, the prognosis is still poor with a median survival of only 14.6 months. Chemoresistance is one of the major causes of relapse as well as poor survival in glioma patients. Therefore, novel strategies to overcome chemoresistance are desperately needed for improved treatment of human GBM. Recent studies have demonstrated that the tumor microenvironment plays a critical role in the chemoresistance of various tumor types, which makes it a suitable target in anti-cancer therapies, as well as a valuable biomarker for prognostic purposes. This review focuses on chemoresistance in GBM induced by stromal cells, including the endothelium of blood vessels, astrocytes, and myeloid cells, as well as non-cellular factors in the tumor microenvironment. Corresponding therapies are discussed, including progressive strategies involving 3-dimensional models integrating engineering as well as biological advances.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Kaikai Ding
- Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, PR China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Peng Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China.
| |
Collapse
|
20
|
Orr MB, Gensel JC. Spinal Cord Injury Scarring and Inflammation: Therapies Targeting Glial and Inflammatory Responses. Neurotherapeutics 2018; 15:541-553. [PMID: 29717413 PMCID: PMC6095779 DOI: 10.1007/s13311-018-0631-6] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Deficits in neuronal function are a hallmark of spinal cord injury (SCI) and therapeutic efforts are often focused on central nervous system (CNS) axon regeneration. However, secondary injury responses by astrocytes, microglia, pericytes, endothelial cells, Schwann cells, fibroblasts, meningeal cells, and other glia not only potentiate SCI damage but also facilitate endogenous repair. Due to their profound impact on the progression of SCI, glial cells and modification of the glial scar are focuses of SCI therapeutic research. Within and around the glial scar, cells deposit extracellular matrix (ECM) proteins that affect axon growth such as chondroitin sulfate proteoglycans (CSPGs), laminin, collagen, and fibronectin. This dense deposition of material, i.e., the fibrotic scar, is another barrier to endogenous repair and is a target of SCI therapies. Infiltrating neutrophils and monocytes are recruited to the injury site through glial chemokine and cytokine release and subsequent upregulation of chemotactic cellular adhesion molecules and selectins on endothelial cells. These peripheral immune cells, along with endogenous microglia, drive a robust inflammatory response to injury with heterogeneous reparative and pathological properties and are targeted for therapeutic modification. Here, we review the role of glial and inflammatory cells after SCI and the therapeutic strategies that aim to replace, dampen, or alter their activity to modulate SCI scarring and inflammation and improve injury outcomes.
Collapse
Affiliation(s)
- Michael B Orr
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky College of Medicine, 741 S. Limestone, B463 BBSRB, Lexington, Kentucky, 40536, USA
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky College of Medicine, 741 S. Limestone, B463 BBSRB, Lexington, Kentucky, 40536, USA.
| |
Collapse
|
21
|
Tworkoski E, Glucksberg MR, Johnson M. The effect of the rate of hydrostatic pressure depressurization on cells in culture. PLoS One 2018; 13:e0189890. [PMID: 29315329 PMCID: PMC5760025 DOI: 10.1371/journal.pone.0189890] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/04/2017] [Indexed: 01/07/2023] Open
Abstract
Changes in hydrostatic pressure, at levels as low as 10 mm Hg, have been reported in some studies to alter cell function in vitro; however, other studies have found no detectable changes using similar methodologies. We here investigate the hypothesis that the rate of depressurization, rather than elevated hydrostatic pressure itself, may be responsible for these reported changes. Hydrostatic pressure (100 mm Hg above atmospheric pressure) was applied to bovine aortic endothelial cells (BAECs) and PC12 neuronal cells using pressurized gas for periods ranging from 3 hours to 9 days, and then the system was either slowly (~30 minutes) or rapidly (~5 seconds) depressurized. Cell viability, apoptosis, proliferation, and F-actin distribution were then assayed. Our results did not show significant differences between rapidly and slowly depressurized cells that would explain differences previously reported in the literature. Moreover, we found no detectable effect of elevated hydrostatic pressure (with slow depressurization) on any measured variables. Our results do not confirm the findings of other groups that modest increases in hydrostatic pressure affect cell function, but we are not able to explain their findings.
Collapse
Affiliation(s)
- Ellen Tworkoski
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Matthew R. Glucksberg
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
| | - Mark Johnson
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States of America
- Department of Mechanical Engineering, Northwestern University, Evanston, Illinois, United States of America
- Department of Ophthalmology, Northwestern University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
22
|
Liu B, McNally S, Kilpatrick JI, Jarvis SP, O'Brien CJ. Aging and ocular tissue stiffness in glaucoma. Surv Ophthalmol 2017; 63:56-74. [PMID: 28666629 DOI: 10.1016/j.survophthal.2017.06.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 12/27/2022]
Abstract
Glaucoma is a progressive and chronic neurodegenerative disorder characterized by damage to the inner layers of the retina and deformation of the optic nerve head. The degeneration of retinal ganglion cells and their axons results in an irreversible loss of vision and is correlated with increasing age. Extracellular matrix changes related to natural aging generate a stiffer extracellular environment throughout the body. Altered age-associated ocular tissue stiffening plays a major role in a significant number of ophthalmic pathologies. In glaucoma, both the trabecular meshwork and the optic nerve head undergo extensive extracellular matrix remodeling, characterized by fibrotic changes associated with cellular and molecular events (including myofibroblast activation) that drive further tissue fibrosis and stiffening. Here, we review the literature concerning the role of age-related ocular stiffening in the trabecular meshwork, lamina cribrosa, sclera, cornea, retina, and Bruch membrane/choroid and discuss their potential role in glaucoma progression. Because both trabecular meshwork and lamina cribrosa cells are mechanosensitive, we then describe molecular mechanisms underlying tissue stiffening and cell mechanotransduction and how these cellular activities can drive further fibrotic changes within ocular tissues. An improved understanding of the interplay between age-related tissue stiffening and biological responses in the trabecular meshwork and optic nerve head could potentially lead to novel therapeutic strategies for glaucoma treatment.
Collapse
Affiliation(s)
- Baiyun Liu
- School of Physics, Conway Institute, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Sara McNally
- Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Jason I Kilpatrick
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Suzanne P Jarvis
- School of Physics, Conway Institute, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Colm J O'Brien
- Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland; School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
23
|
Quigley HA. Understanding Glaucomatous Optic Neuropathy: The Synergy Between Clinical Observation and Investigation. Annu Rev Vis Sci 2016; 2:235-254. [PMID: 28532352 DOI: 10.1146/annurev-vision-111815-114417] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glaucoma is a complex disorder of aging defined by the death of retinal ganglion cells and remodeling of connective tissues at the optic nerve head. Intraocular pressure-induced axonal injury at the optic nerve head leads to apoptosis. Loss of retinal ganglion cells follows a slowly progressive sequence. Clinical features of the disease have suggested and corroborated pathological events. The death of retinal ganglion cells causes secondary loss of neurons in the brain, but only as a by-product of injury to the retinal ganglion cells. Although therapy to lower intraocular pressure is moderately effective, new treatments are being developed to alter the remodeling of ocular connective tissue, to interrupt the injury signal from axon to soma, and to upregulate a variety of survival mechanisms.
Collapse
Affiliation(s)
- Harry A Quigley
- Glaucoma Center of Excellence, Wilmer Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287;
| |
Collapse
|
24
|
Mac Nair CE, Schlamp CL, Montgomery AD, Shestopalov VI, Nickells RW. Retinal glial responses to optic nerve crush are attenuated in Bax-deficient mice and modulated by purinergic signaling pathways. J Neuroinflammation 2016; 13:93. [PMID: 27126275 PMCID: PMC4850653 DOI: 10.1186/s12974-016-0558-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 04/20/2016] [Indexed: 01/14/2023] Open
Abstract
Background Retinal ganglion cell (RGC) soma death is a consequence of optic nerve damage, including in optic neuropathies like glaucoma. The activation of the innate immune network in the retina after nerve damage has been linked to RGC pathology. Since the eye is immune privileged, innate immune functions are the responsibility of the glia, specifically the microglia, astrocytes, and Müller cells that populate the retina. Glial activation, leading to the production of inflammatory cytokines, is a hallmark feature of retinal injury resulting from optic nerve damage and purported to elicit secondary degeneration of RGC somas. Methods A mouse model of optic nerve crush (ONC) was used to study retinal glial activation responses. RGC apoptosis was blocked using Bax-deficient mice. Glial activation responses were monitored by quantitative PCR and immunofluorescent labeling in retinal sections of activation markers. ATP signaling pathways were interrogated using P2X receptor agonists and antagonists and Pannexin 1 (Panx1)-deficient mice with RGC-specific deletion. Results ONC induced activation of both macroglia and microglia in the retina, and both these responses were dramatically muted if RGC death was blocked by deletion of the Bax gene. Macroglial, but not microglial, activation was modulated by purinergic receptor activation. Release of ATP after optic nerve damage was not mediated by PANX1 channels in RGCs. Conclusions RGC death in response to ONC plays a principal stimulatory role in the retinal glial activation response. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0558-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caitlin E Mac Nair
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, 571A Medical Sciences-1300 University Ave, Madison, WI, 53706, USA.,Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, 3170-10K/L MFCB, 1685 Highland Avenue, Madison, WI, 53705, USA
| | - Cassandra L Schlamp
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, 571A Medical Sciences-1300 University Ave, Madison, WI, 53706, USA
| | - Angela D Montgomery
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, 571A Medical Sciences-1300 University Ave, Madison, WI, 53706, USA
| | - Valery I Shestopalov
- Department of Ophthalmology, University of Miami Miller School of Medicine, 900 N.W. 17th Street, Miami, FL, 33136, USA.,Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, 900 N.W. 17th Street, Miami, FL, 33136, USA
| | - Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, 571A Medical Sciences-1300 University Ave, Madison, WI, 53706, USA.
| |
Collapse
|
25
|
Russo R, Varano GP, Adornetto A, Nucci C, Corasaniti MT, Bagetta G, Morrone LA. Retinal ganglion cell death in glaucoma: Exploring the role of neuroinflammation. Eur J Pharmacol 2016; 787:134-42. [PMID: 27044433 DOI: 10.1016/j.ejphar.2016.03.064] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 03/10/2016] [Accepted: 03/31/2016] [Indexed: 01/06/2023]
Abstract
In clinical glaucoma, as well as in experimental models, the loss of retinal ganglion cells occurs by apoptosis. This final event is preceded by inflammatory responses involving the activation of innate and adaptive immunity, with retinal and optic nerve resident glial cells acting as major players. Here we review the current literature on the role of neuroinflammation in neurodegeneration, focusing on the inflammatory molecular mechanisms involved in the pathogenesis and progression of the optic neuropathy.
Collapse
Affiliation(s)
- Rossella Russo
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy.
| | - Giuseppe Pasquale Varano
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Annagrazia Adornetto
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Carlo Nucci
- Ophthalmology Unit, Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome Italy
| | | | - Giacinto Bagetta
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy; University Center for Adaptive Disorders and Head Pain, Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, Arcavacata di Rende, Italy
| | - Luigi Antonio Morrone
- Department of Pharmacy, Nutritional and Health Sciences, University of Calabria, Arcavacata di Rende, Italy; University Center for Adaptive Disorders and Head Pain, Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
26
|
Nickells RW, Pelzel HR. Tools and resources for analyzing gene expression changes in glaucomatous neurodegeneration. Exp Eye Res 2015; 141:99-110. [PMID: 25999234 DOI: 10.1016/j.exer.2015.05.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/10/2015] [Accepted: 05/17/2015] [Indexed: 01/12/2023]
Abstract
Evaluating gene expression changes presents one of the most powerful interrogative approaches to study the molecular, biochemical, and cellular pathways associated with glaucomatous disease pathology. Technologies to study gene expression profiles in glaucoma are wide ranging. Qualitative techniques provide the power of localizing expression changes to individual cells, but are not robust to evaluate differences in expression changes. Alternatively, quantitative changes provide a high level of stringency to quantify changes in gene expression. Additionally, advances in high throughput analysis and bioinformatics have dramatically improved the number of individual genes that can be evaluated in a single experiment, while dramatically reducing amounts of input tissue/starting material. Together, gene expression profiling and proteomics have yielded new insights on the roles of neuroinflammation, the complement cascade, and metabolic shutdown as important players in the pathology of the optic nerve head and retina in this disease.
Collapse
Affiliation(s)
- Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin - Madison, Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin - Madison, Madison, WI, USA.
| | - Heather R Pelzel
- Department of Biological Sciences, University of Wisconsin - Whitewater, Whitewater, WI, USA
| |
Collapse
|
27
|
|
28
|
Kaja S, Payne AJ, Patel KR, Naumchuk Y, Koulen P. Differential subcellular Ca2+ signaling in a highly specialized subpopulation of astrocytes. Exp Neurol 2014; 265:59-68. [PMID: 25542978 DOI: 10.1016/j.expneurol.2014.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/26/2014] [Accepted: 12/12/2014] [Indexed: 11/24/2022]
Abstract
Recent evidence suggests that astrocytes do not serve a mere buffering function, but exhibit complex signaling pathways, disturbance of which contributes significantly to the pathophysiology of CNS diseases. Little is known regarding the intracellular signaling pathways in the specialized optic nerve head astrocytes (ONHAs), the major glia cell type in non-myelinated optic nerve head. Here we show the differential subcellular expression of intracellular Ca(2+) channels in ONHAs. Expression of type 1 and type 3 inositol-1-4-5,-trisphosphate receptors (IP3Rs) in the endoplasmic reticulum and type 2 IP3Rs in the nuclear envelope causes differential Ca(2+) release from intracellular stores in nuclear vs. cytosolic compartments. Our study identifies differential distribution and activity of Ca(2+) channels as molecular substrate and mechanism by which astrocytes independently regulate Ca(2+) transients in both cytoplasm and nucleoplasm, thereby controlling genomic and non-genomic cellular signaling, respectively. This provides excellent targets for therapeutics restoring pathological disturbances of intracellular Ca(2+) signaling present in glaucoma and other neurodegenerative disorders with astrocyte involvement.
Collapse
Affiliation(s)
- Simon Kaja
- Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City, School of Medicine, 2411 Holmes St., Kansas City, MO 64108, USA
| | - Andrew J Payne
- Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City, School of Medicine, 2411 Holmes St., Kansas City, MO 64108, USA
| | - Krupa R Patel
- Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City, School of Medicine, 2411 Holmes St., Kansas City, MO 64108, USA
| | - Yuliya Naumchuk
- Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City, School of Medicine, 2411 Holmes St., Kansas City, MO 64108, USA
| | - Peter Koulen
- Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City, School of Medicine, 2411 Holmes St., Kansas City, MO 64108, USA.
| |
Collapse
|
29
|
Tehrani S, Johnson EC, Cepurna WO, Morrison JC. Astrocyte processes label for filamentous actin and reorient early within the optic nerve head in a rat glaucoma model. Invest Ophthalmol Vis Sci 2014; 55:6945-52. [PMID: 25257054 DOI: 10.1167/iovs.14-14969] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine if astrocyte processes label for actin and to quantify the orientation of astrocytic processes within the optic nerve head (ONH) in a rat glaucoma model. METHODS Chronic intraocular pressure (IOP) elevation was produced by episcleral hypertonic saline injection and tissues were collected after 5 weeks. For comparison, eyes with optic nerve transection were collected at 2 weeks. Fellow eyes served as controls. Axonal degeneration in retrobulbar optic nerves was graded on a scale of 1 to 5. Optic nerve head sections (n ≥ 4 eyes per group) were colabeled with phalloidin (actin marker) and antibodies to astrocytic glial fibrillary acidic protein and aquaporin 4, or axonal tubulin βIII. Confocal microscopy and FIJI software were used to quantify the orientation of actin bundles. RESULTS Control ONHs showed stereotypically arranged actin bundles within astrocyte processes. Optic nerve head actin bundle orientation was nearly perpendicular to axons (82.9° ± 6.3° relative to axonal axis), unlike the retrobulbar optic nerve (45.4° ± 28.7°, P < 0.05). With IOP elevation, ONH actin bundle orientation became less perpendicular to axons, even in eyes with no perceivable axonal injury (i.e., 38.8° ± 15.1° in grade 1, P < 0.05 in comparison to control ONHs). With severe injury, ONH actin bundle orientation became more parallel to the axonal axis (24.1° ± 28.4°, P < 0.05 in comparison to control ONHs). Optic nerve head actin bundle orientation in transected optic nerves was unchanged. CONCLUSIONS Actin labeling identifies fine astrocyte processes within the ONH. Optic nerve head astrocyte process reorientation occurs early in response to elevated IOP.
Collapse
Affiliation(s)
- Shandiz Tehrani
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Elaine C Johnson
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - William O Cepurna
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - John C Morrison
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
30
|
Ho KW, Lambert WS, Calkins DJ. Activation of the TRPV1 cation channel contributes to stress-induced astrocyte migration. Glia 2014; 62:1435-51. [PMID: 24838827 DOI: 10.1002/glia.22691] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 04/25/2014] [Accepted: 04/29/2014] [Indexed: 01/13/2023]
Abstract
Astrocytes provide metabolic, structural, and synaptic support to neurons in normal physiology and also contribute widely to pathogenic processes in response to stress or injury. Reactive astrocytes can undergo cytoskeletal reorganization and increase migration through changes in intracellular Ca(2+) mediated by a variety of potential modulators. Here we tested whether migration of isolated retinal astrocytes following mechanical injury (scratch wound) involves the transient receptor potential vanilloid-1 channel (TRPV1), which contributes to Ca(2+)-mediated cytoskeletal rearrangement and migration in other systems. Application of the TRPV1-specific antagonists, capsazepine (CPZ) or 5'-iodoresiniferatoxin (IRTX), slowed migration by as much as 44%, depending on concentration. In contrast, treatment with the TRPV1-specific agonists, capsaicin (CAP) or resiniferatoxin (RTX) produced only a slight acceleration over a range of concentrations. Chelation of extracellular Ca(2+) with EGTA (1 mM) slowed astrocyte migration by 35%. Ratiometric imaging indicated that scratch wound induced a sharp 20% rise in astrocyte Ca(2+) that dissipated with distance from the wound. Treatment with IRTX both slowed and dramatically reduced the scratch-induced Ca(2+) increase. Both CPZ and IRTX influenced astrocyte cytoskeletal organization, especially near the wound edge. Taken together, our results indicate that astrocyte mobilization in response to mechanical stress involves influx of extracellular Ca(2+) and cytoskeletal changes in part mediated by TRPV1 activation.
Collapse
Affiliation(s)
- Karen W Ho
- Vanderbilt Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | |
Collapse
|
31
|
Hussain AA, Lee Y, Zhang JJ, Marshall J. Characterization of the gelatinase system of the laminar human optic nerve, and surrounding annulus of Bruch's membrane, choroid, and sclera. Invest Ophthalmol Vis Sci 2014; 55:2358-64. [PMID: 24609626 DOI: 10.1167/iovs.13-12503] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE We determined the presence and levels of gelatinase matrix metalloproteinases (MMPs) in the optic nerve and surrounding rim region of the human fundus. METHODS Samples of optic nerve, rim region, and Bruch's membrane-choroid from macular and peripheral regions were isolated from 9 pairs of human donor eyes. The MMPs were extracted and separated by gelatin zymography. Individual gelatinase species were identified by their respective molecular weights and levels quantified by standard densitometric techniques. Ratios of active/latent MMPs were calculated as representative indicators of the degree of proteolytic activity at each of the locations examined. RESULTS All of the gelatinase species normally found in Bruch's membrane also were present in the optic nerve region. The presence of the high molecular weight MMP species (HMW1 and HMW2) was indicative of the age-related accumulation of polymerized MMPs 2 and 9. Level of activated MMPs was considerably raised in comparison with their latent forms at the optic nerve and surrounding region indicative of greater ongoing turnover of the matrix (P < 0.005). CONCLUSIONS The components of the gelatinase pathway mediating matrix turnover in Bruch's membrane also were present in the optic nerve region. The presence of high levels of active MMPs 2 and 9 in comparison with the latent forms in the optic nerve and rim area is indicative of a high rate of matrix remodeling in these regions. Enhanced matrix turnover within the optic nerve region may represent an important mechanism for maintaining the plasticity of the lamina cribrosa.
Collapse
Affiliation(s)
- Ali A Hussain
- Department of Genetics, University College London (UCL) Institute of Ophthalmology, University of London, London, United Kingdom
| | | | | | | |
Collapse
|
32
|
Shih GC, Calkins DJ. Secondary neuroprotective effects of hypotensive drugs and potential mechanisms of action. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 7:161-175. [PMID: 22737176 DOI: 10.1586/eop.12.13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Primary open-angle glaucoma, a long-term degenerative ocular neuropathy, remains a significant cause of vision impairment worldwide. While many risk factors have been correlated with increased risk for primary open-angle glaucoma, intraocular pressure (IOP) remains the only modifiable risk factor and primary therapeutic target. Pharmacologic therapies are administered topically; these include α(2)-agonists, β-antagonists, prostaglandin analogs and carbonic anhydrase inhibitors. Some of these topical medications exhibit secondary neuroprotective effects independent of their effect on IOP. This review covers the possible mechanisms of neuroprotection stimulated by drugs currently marketed for the lowering of IOP, based on known literature. While the neuroprotective properties of many glaucoma pharmaceuticals are promising from an experimental standpoint, key challenges for the development of new clinical practices include unknown systemic side effects, limited methods of drug delivery to the retina and optic nerve, and development of extended-release formulations.
Collapse
Affiliation(s)
- Grace C Shih
- The Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University, School of Medicine, 11435 MRB IV, 2215B Garland Avenue, Nashville, TN 37232, USA
| | | |
Collapse
|
33
|
Yang C, Rahimpour S, Yu ACH, Lonser RR, Zhuang Z. Regulation and dysregulation of astrocyte activation and implications in tumor formation. Cell Mol Life Sci 2013; 70:4201-11. [PMID: 23420481 PMCID: PMC11113190 DOI: 10.1007/s00018-013-1274-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 01/11/2013] [Accepted: 01/22/2013] [Indexed: 01/06/2023]
Abstract
Astrocytic activation is a cellular response to disturbances of the central nervous system (CNS). Recent advances in cellular and molecular biology have demonstrated the remarkable changes in molecular signaling, morphology, and metabolism that occur during astrocyte activation. Based on these studies, it has become clear that the astrocyte activation process is regulated by a variety of signaling pathways, which result in metabolic support, wound healing and scar formation. While normal astrocyte activation pathways drive homeostasis and/or repair in the CNS, dysregulation of these pathways can lead to astrocyte abnormalities, including glioma formation with similar phenotypes as reactive astrocytes. We review the principle pathways responsible for astrocytic activation, as well as their potential contribution to tumor formation in the CNS.
Collapse
Affiliation(s)
- Chunzhang Yang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 10, Room 3D20, Bethesda, Maryland 20892-1414 USA
| | - Shervin Rahimpour
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 10, Room 3D20, Bethesda, Maryland 20892-1414 USA
| | - Albert C. H. Yu
- Neuroscience Research Institute, Key Laboratory of Neuroscience (Ministry of Education), Key Laboratory for Neuroscience (Ministry of Public Health), Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Russell R. Lonser
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 10, Room 3D20, Bethesda, Maryland 20892-1414 USA
| | - Zhengping Zhuang
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 10, Room 3D20, Bethesda, Maryland 20892-1414 USA
| |
Collapse
|
34
|
Nussenblatt RB, Liu B, Wei L, Sen HN. The immunological basis of degenerative diseases of the eye. Int Rev Immunol 2013; 32:97-112. [PMID: 23360161 DOI: 10.3109/08830185.2012.740536] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
It has become clear that disorders that were once considered "degenerative" have complex mechanisms, with many having been shown to have immune mediation as part of the disease process. These include arteriosclerotic heart disease and Alzheimer's disease. Indeed, several ocular disorders that once fell into the "degenerative" category meet this criterion as well. Immune mediation has been shown to be a part of many of the most common ocular disorders, and not just that of uveitis, or ocular inflammatory disease.
Collapse
Affiliation(s)
- Robert B Nussenblatt
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20814, USA.
| | | | | | | |
Collapse
|
35
|
Lye-Barthel M, Sun D, Jakobs TC. Morphology of astrocytes in a glaucomatous optic nerve. Invest Ophthalmol Vis Sci 2013; 54:909-17. [PMID: 23322566 DOI: 10.1167/iovs.12-10109] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE To establish the morphologic changes of astrocytes in the glial lamina of glaucomatous mice. METHODS A strain of mice that expresses GFP in individual astrocytes (hGFAPpr-GFP) was crossed into the DBA/2J strain that develops glaucoma. In the resulting strain (D2.hGFAPpr-GFP) we assessed the severity of glaucoma by staining the retina for neurofilaments and counting the neurons of the retinal ganglion cell layer. We observed the morphology of astrocytes in the glial lamina of the optic nerves. RESULTS D2.hGFAPpr-GFP mice developed glaucoma in an age-dependent manner. Astrocytes in the glial lamina showed morphologic changes that correlated with the severity of glaucoma. The cells showed thickening of processes from 1.3 ± 0.28 μm in nondiseased animals to 1.71 ± 0.46 μm in eyes with moderate glaucoma and 2.1 ± 0.42 μm in those with severe glaucoma. Their spatial coverage, as determined by their convex polygon area, was reduced in eyes with severe glaucoma. The astrocytes in severely glaucomatous optic nerves also showed simplification of their processes. In 6-month-old mice with no obvious signs of degeneration in the retina, we found astrocytes with appendages growing out of primary astrocyte processes into the axon bundles. This localized hypertrophy of processes was never observed in the hGFAPpr-GFP strain. CONCLUSIONS Confirming results after optic nerve crush, astrocytes in glaucomatous optic nerves had thickened and simplified processes, and reduced spatial coverage. We also found evidence of localized sprouting of new processes in early stages of the disease, before detectable changes in ganglion cell number.
Collapse
Affiliation(s)
- Ming Lye-Barthel
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, USA
| | | | | |
Collapse
|
36
|
Scarsella G, Nebbioso M, Stefanini S, Pescosolido N. Degenerative effects in rat eyes after experimental ocular hypertension. Eur J Histochem 2012; 56:e42. [PMID: 23361238 PMCID: PMC3567761 DOI: 10.4081/ejh.2012.e42] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 09/17/2012] [Accepted: 09/12/2012] [Indexed: 11/23/2022] Open
Abstract
This study was used to evaluate the degenerative effects on the retina and eye-cup sections after experimental induction of acute ocular hypertension on animal models. In particular, vascular events were directly focused in this research in order to assess the vascular remodeling after transient ocular hypertension on rat models. After local anaesthesia by administration of eye drops of 0.4% oxibuprocaine, 16 male adult Wistar rats were injected in the anterior chamber of the right eye with 15 µL of methylcellulose (MTC) 2% in physiological solution. The morphology and the vessels of the retina and eye-cup sections were examined in animals sacrificed 72 h after induction of ocular hypertension. In retinal fluorescein angiographies (FAGs), by means of fluorescein isothiocyanate-coniugated dextran (FITC), the radial venules showed enlargements and increased branching, while the arterioles appeared focally thickened. The length and size of actually perfused vessels appeared increased in the whole superficial plexus. In eye-cup sections of MTC-injected animals, in deep plexus and connecting layer there was a bigger increase of vessels than in controls. Moreover, the immunolocalization of astrocytic marker glial fibrillary acidic protein (GFAP) revealed its increased expression in internal limiting membrane and ganglion cell layer, as well as its presence in Müller cells. Finally, the pro-angiogenic factor vascular endothelial growth factor (VEGF) was found to be especially expressed by neurones of ganglion cell layer, both in control and in MTC-injected eyes. The data obtained in this experimental model on the interactions among glia, vessels and neurons should be useful to evaluate if also in glaucomatous patients the activation of vessel-adjacent glial cells might play key roles in following neuronal dysfunction.
Collapse
Affiliation(s)
- G Scarsella
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome
| | | | | | | |
Collapse
|
37
|
Li X, Lin WJ, Chen CY, Si Y, Zhang X, Lu L, Suswam E, Zheng L, King PH. KSRP: a checkpoint for inflammatory cytokine production in astrocytes. Glia 2012; 60:1773-84. [PMID: 22847996 DOI: 10.1002/glia.22396] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 06/29/2012] [Indexed: 11/08/2022]
Abstract
Chronic inflammation in the central nervous system (CNS) is a central feature of many neurodegenerative and autoimmune diseases. As an immunologically competent cell, the astrocyte plays an important role in CNS inflammation. It is capable of expressing a number of cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) that promote inflammation directly and through the recruitment of immune cells. Checkpoints are therefore in place to keep tight control over cytokine production. Adenylate/uridylate-rich elements (ARE) in the 3' untranslated region of cytokine mRNAs serve as a major checkpoint by regulating mRNA stability and translational efficiency. Here, we examined the impact of KH-type splicing regulatory protein (KSRP), an RNA binding protein which destabilizes mRNAs via the ARE, on cytokine expression and paracrine phenotypes of primary astrocytes. We identified a network of inflammatory mediators, including TNF-α and IL-1β, whose expression increased 2 to 4-fold at the RNA level in astrocytes isolated from KSRP(-/-) mice compared to littermate controls. Upon activation, KSRP(-/-) astrocytes produced TNF-α and IL-1β at levels that exceeded control cells by 15-fold or more. Conditioned media from KSRP(-/-) astrocytes induced chemotaxis and neuronal cell death in vitro. Surprisingly, we observed a prolongation of half-life in only a subset of mRNA targets and only after selective astrocyte activation. Luciferase reporter studies indicated that KSRP regulates cytokine gene expression at both transcriptional and post-transcriptional levels. Our results outline a critical role for KSRP in regulating pro-inflammatory mediators and have implications for a wide range of CNS inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Xuelin Li
- Department of Neurology, University of Alabama, Birmingham, Alabama 35233-0017, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Human anterior lens capsule serving as a substrate for human trabecular meshwork cells cultivation. Cell Tissue Bank 2012; 14:407-12. [DOI: 10.1007/s10561-012-9332-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 07/18/2012] [Indexed: 10/28/2022]
|
39
|
Terai N, Spoerl E, Haustein M, Hornykewycz K, Haentzschel J, Pillunat LE. Diabetes mellitus affects biomechanical properties of the optic nerve head in the rat. Ophthalmic Res 2011; 47:189-94. [PMID: 22156545 DOI: 10.1159/000331990] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 03/22/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND To investigate the effect of diabetes on the biomechanical behavior of the optic nerve head (ONH) and the peripapillary sclera (ppSc) in streptozocine-induced diabetic rats. METHODS Diabetes mellitus was induced in 20 Wistar rats using streptozocine. Twenty-five nondiabetic rats served as controls. Eyes were enucleated after 12 weeks and 2 strips of one eye were prepared containing ONH or ppSc. The stress-strain relation was measured in the stress range of 0.05-10 MPa using a biomaterial tester. RESULTS At 5% strain the stress of the ONH in diabetic rats was 897±295 kPa and in the control group it was 671±246 kPa; there was a significant difference between both groups (p=0.011). The stress of the diabetic ppSc (574±185 kPa) increased compared to that of the nondiabetic ppSc (477±171 kPa), but this did not reach statistical significance (p=0.174). The calculated tangent modulus at 5% strain was 11.79 MPa in the diabetic ONH and 8.77 MPa in the nondiabetic ONH; there was a significant difference between both groups (p=0.006). The calculated tangent modulus at 5% strain was 7.17 MPa in the diabetic ppSc and 6.12 MPa in the nondiabetic ppSc, without a statistically significant difference (p=0.09). CONCLUSION In contrast to the ppSc, the ONH of diabetic rats showed a significant increase in stiffness compared to nondiabetic rats, which might be explained by nonenzymatic collagen cross-linking mediated by advanced glycation end products due to high blood glucose levels in diabetes. Further studies are needed to investigate if these biomechanical changes represent a detrimental risk factor for intraocular pressure regulation in diabetic glaucoma patients.
Collapse
Affiliation(s)
- Naim Terai
- Department of Ophthalmology, Carl Gustav Carus University Hospital, Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
40
|
Tribute to Rosario Hernandez. Exp Eye Res 2011; 93:116-9. [DOI: 10.1016/j.exer.2011.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 05/09/2011] [Accepted: 05/10/2011] [Indexed: 11/22/2022]
|
41
|
Burgoyne CF. A biomechanical paradigm for axonal insult within the optic nerve head in aging and glaucoma. Exp Eye Res 2011; 93:120-32. [PMID: 20849846 PMCID: PMC3128181 DOI: 10.1016/j.exer.2010.09.005] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 09/04/2010] [Accepted: 09/07/2010] [Indexed: 12/27/2022]
Abstract
This article is dedicated to Rosario Hernandez for her warm support of my own work and her genuine enthusiasm for the work of her colleagues throughout her career. I first met Rosario as a research fellow in Harry Quigley's laboratory between 1991 and 1993. Along with Harry, John Morrison, Elaine Johnson, Abe Clark, Colm O'Brien and many others, Rosario's work has provided lamina cribrosa astrocyte cellular mechanisms that are biomechanically plausible and in so doing provided credibility to early notions of the optic nerve head (ONH) as a biomechanical structure. We owe a large intellectual debt to Rosario for her dogged persistence in the characterization of the ONH astrocyte and lamina cribrosacyte in age and disease. Two questions run through her work and remain of central importance today. First, how do astrocytes respond to and alter the biomechanical environment of the ONH and the physiologic stresses created therein? Second, how do these physiologic demands on the astrocyte influence their ability to deliver the support to retinal ganglion cell axon transport and flow against the translaminar pressure gradient? The purpose of this article is to summarize what is known about the biomechanical determinants of retinal ganglion cell axon physiology within the ONH in the optic neuropathy of aging and Glaucoma. My goal is to provide a biomechanical framework for this discussion. This framework assumes that the ONH astrocytes and glia fundamentally support and influence both the lamina cribrosa extracellular matrix and retinal ganglion cell axon physiology. Rosario Hernandez was one of the first investigators to recognize the implications of this unique circumstance. Many of the ideas contained herein have been initially presented within or derived from her work (Hernandez, M.R., 2000. The optic nerve head in glaucoma: role of astrocytes in tissue remodeling. Prog Retin Eye Res. 19, 297-321.; Hernandez, M.R., Pena, J.D., 1997. The optic nerve head in glaucomatous optic neuropathy. Arch Ophthalmol. 115, 389-395.).
Collapse
Affiliation(s)
- Claude F Burgoyne
- Optic Nerve Head Research Laboratory, Part of the Discoveries in Sight Research Laboratories of the Devers Eye Institute, Legacy Health System, 1225 NE 2nd Ave, Portland, OR 97232, USA.
| |
Collapse
|
42
|
Sato K, Horiuchi Y, Jin Y, Malchinkhuu E, Komachi M, Kondo T, Okajima F. Unmasking of LPA1 receptor-mediated migration response to lysophosphatidic acid by interleukin-1β-induced attenuation of Rho signaling pathways in rat astrocytes. J Neurochem 2011; 117:164-74. [PMID: 21244430 DOI: 10.1111/j.1471-4159.2011.07188.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Action mechanism of lipopolysaccharide (LPS), interleukin-1β (IL-1β), and lysophosphatidic acid (LPA) to regulate motility, an important process of astrogliosis, was investigated in rat astrocytes. While LPA exerted no significant effect on the cell migration, the prior treatment of the cells with LPS or IL-1β resulted in the appearance of migration activity in response to LPA. The LPS induction of the migration response to LPA was associated with the production of IL-1β precursor protein and inhibited by the IL-1 receptor antagonist. The IL-1β treatment also allowed LPA to activate Rac1. The LPA-induced Rac1 activation and migration were inhibited by pertussis toxin, a small interfering RNA specific to LPA(1) receptors, and LPA(1) receptor antagonists, including Ki16425. However, the IL-1β treatment had no appreciable effect on LPA(1) receptor mRNA expression and LPA-induced activation of ERK, Akt, and proliferation. The induction of the migration response to LPA by IL-1β was inhibited by a constitutively active RhoA. Moreover, LPA significantly activated RhoA through the LPA(1) receptor in the control cells but not in the IL-1β-treated cells. These results suggest that IL-1β inhibits the LPA(1) receptor-mediated Rho signaling through the IL-1 receptor, thereby disclosing the LPA(1) receptor-mediated G(i) protein/Rac/migration pathway.
Collapse
Affiliation(s)
- Koichi Sato
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Crish SD, Calkins DJ. Neurodegeneration in glaucoma: progression and calcium-dependent intracellular mechanisms. Neuroscience 2010; 176:1-11. [PMID: 21187126 DOI: 10.1016/j.neuroscience.2010.12.036] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/16/2010] [Accepted: 12/18/2010] [Indexed: 01/18/2023]
Abstract
Glaucoma is an age-related optic neuropathy involving sensitivity to ocular pressure. The disease is now seen increasingly as one of the central nervous system, as powerful new approaches highlight an increasing number of similarities with other age-related neurodegenerations such as Alzheimer's and Parkinson's. While the etiologies of these diseases are diverse, they involve many important common elements including compartmentalized programs of degeneration targeting axons, dendrites and finally cell bodies. Most age-related degenerations display early functional deficits that precede actual loss of neuronal substrate. These are linked to several specific neurochemical cascades that can be linked back to dysregulation of Ca(2+)-dependent processes. We are now in the midst of identifying similar cascades in glaucoma. Here we review recent evidence on the pathological progression of neurodegeneration in glaucoma and some of the Ca(2+)-dependent mechanisms that could underlie these changes. These mechanisms present clear implications for efforts to develop interventions targeting neuronal loss directly and make glaucoma an attractive model for both interrogating and informing other neurodegenerative diseases.
Collapse
Affiliation(s)
- S D Crish
- Department of Pharmaceutical Sciences, Northeastern Ohio Universities Colleges of Medicine and Pharmacy, Rootstown, OH 44272, USA
| | | |
Collapse
|
44
|
Calandrella N, De Seta C, Scarsella G, Risuleo G. Carnitine reduces the lipoperoxidative damage of the membrane and apoptosis after induction of cell stress in experimental glaucoma. Cell Death Dis 2010; 1:e62. [PMID: 21364667 PMCID: PMC3032525 DOI: 10.1038/cddis.2010.40] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The pathological damage caused by glaucoma is associated to a high intraocular pressure. The ocular hypertone is most likely due to a defective efflux of aqueous humor from the anterior chamber of the eye. Ocular hypertension causes apoptotic death of retinal ganglion cells and overexpression of molecular markers typical of cell stress response and apoptosis. In this work, we report on the neuroprotective, antiapoptotic and antioxidant action of a natural substance, -carnitine. This compound is known for its ability to improve the mitochondrial performance. We analyze a number of cellular and molecular markers, typical of ocular hypertension and, in general, of the cell stress response. In particular, -carnitine reduces the expression of glial fibrillary acidic protein, inducible nitric oxide synthase, ubiquitin and caspase 3 typical markers of cell stress. In addition, the morphological analysis of the optic nerve evidenced a reduction of the pathological excavation of the optic disk. This experimental hypertone protocol induces a severe lipoperoxidation, which is significantly reduced by -carnitine. The overall interpretation is that mortality of the retinal cells is due to membrane damage.
Collapse
Affiliation(s)
- N Calandrella
- Dipartimento di Biologia Cellulare e dello Sviluppo, Università di Roma La Sapienza, Roma, Italy
| | | | | | | |
Collapse
|
45
|
Miao H, Crabb AW, Hernandez MR, Lukas TJ. Modulation of factors affecting optic nerve head astrocyte migration. Invest Ophthalmol Vis Sci 2010; 51:4096-103. [PMID: 20375339 DOI: 10.1167/iovs.10-5177] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
PURPOSE The authors investigated the role of myosin light chain kinase (MYLK) and transforming growth factor beta (TGFbeta) receptor pathways in optic nerve head (ONH) astrocyte migration. They further investigated how the expression of these genes is altered by elevated hydrostatic pressure (HP). METHODS PCR was used to determine the isoforms of MYLK expressed in ONH astrocytes. siRNAs against MYLK (all isoforms) and TGFbeta receptor 2 (TGFBR2) were prepared and tested for effects on the migration of cultured ONH astrocytes. Finally, the effects of elevated HP (24-96 hours) on the expression of MYLK isoforms and selected TGFbeta pathway components were measured. RESULTS Multiple isoforms of MYLK are present in ONH astrocytes from Caucasian (CA) and African American (AA) donors. Both populations express the short form (MYLK-130) and the long form (MYLK-210) of MYLK and a splicing variant within MYLK-210. MYLK-directed siRNA decreased MYLK expression and cell migration compared with control siRNA. siRNA directed against TGFbeta receptor 2 also decreased cell migration compared with control and decreased extracellular matrix genes regulated by TGFbeta signaling. Elevated HP increased the expression of MYLK-130 and MYLK-210 in both populations of astrocytes. However, TGFbeta2 was uniquely upregulated by exposure to elevated HP in CA compared with AA astrocytes. CONCLUSIONS Differential expression of TGFbeta pathway genes and MYLK isoforms observed in populations of glaucomatous astrocytes applies to the elevated HP model system. MYLK may be a new target for intervention in glaucoma to alter reactive astrocyte migration in the ONH.
Collapse
Affiliation(s)
- Haixi Miao
- Department of Ophthalmology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
46
|
Vidal L, Díaz F, Villena A, Moreno M, Campos JG, Pérez de Vargas I. Reaction of Müller cells in an experimental rat model of increased intraocular pressure following timolol, latanoprost and brimonidine. Brain Res Bull 2010; 82:18-24. [PMID: 20206241 DOI: 10.1016/j.brainresbull.2010.02.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 02/19/2010] [Accepted: 02/23/2010] [Indexed: 11/30/2022]
Abstract
The aim of this study was to evaluate the reaction of Müller cells in an experimental rat model of intraocular pressure (IOP) and their response to treatment with ocular hypotensive drugs. Episcleral vein cauterization in unilateral eyes of Wistar rats was performed to produce elevated IOP. The animals were divided into five groups: control, experimental, and experimental treated with timolol, latanoprost or brimonidine. Histological sections of retina were studied by immunochemistry with antibodies to glial fibrillary acidic protein (GFAP), and the percentage of labeled area was measured to evaluate the degree of reactive gliosis. In the experimental group, the Müller cells showed hypertrophy and a significant increase in GFAP (4.39+/-0.32%) in relation to retinas of the control group (2.05+/-0.14%). Gliosis was detected in all three treated groups, with a varying increase in GFAP intensity. The timolol-treated group showed the most intense and persistent glial reactivity after 3 months of treatment (13.89+/-0.63%). Treatment with brimonidine, however, resulted in a decrease in the level of GFAP immunoreactivity (8.37+/-0.4%). The group treated with latanoprost showed the lowest glial reactivity (4.8+/-0.36%). Given that all three drugs are effective hypotensive agents, their neuroprotective effect could be related with other factors, such as gliosis, which, over long periods may have noxious effects on the neurons. Thus, hypotensives like brimonidine, and specially latanoprost, may afford greater neuroprotection to the ganglion cells by attenuating the retinal glial reaction.
Collapse
Affiliation(s)
- Lourdes Vidal
- Department of Histology and Histopathology, School of Medicine, University of Malaga, Boulevard Louis Pasteur 32, 29071 Malaga, Spain.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Glaucomatous vision loss results from the progressive degeneration of optic nerve axons and the death of retinal ganglion cells. This process is accompanied by dramatic alterations in the functional properties and distribution of glial cells in both the retina and the optic nerve head in a reaction commonly referred to as glial activation. The recent availability of rodent and cell culture glaucoma models has substantially contributed to our knowledge of glial activation under glaucomatous conditions. Conclusions drawn from these studies have led to the refinement of existing hypotheses and the generation of new ones. Because these hypotheses encompass both protective and injurious roles for glia, the impact of specific aspects of glial activation are current topics of intensive research, speculation, and debate in the field. With these unresolved issues in mind, this review will summarize recent progress in our understanding of the process of glial activation in the glaucomatous optic nerve head and retina.
Collapse
|
48
|
Fuller JA, Brun-Zinkernagel AM, Clark AF, Wordinger RJ. Subtilisin-like proprotein convertase expression, localization, and activity in the human retina and optic nerve head. Invest Ophthalmol Vis Sci 2009; 50:5759-68. [PMID: 19339735 DOI: 10.1167/iovs.08-2616] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Subtilisin-like proprotein convertases (SPCs) are a family of calcium-dependent cleavage enzymes that act on dibasic sites of various peptide/protein substrates. The purpose of this study was to investigate the expression, localization, and activity of SPCs in the human retina and optic nerve head. METHODS mRNA expression of the SPC family in the human retina and optic nerve head tissues was evaluated by quantitative reverse transcription polymerase chain reaction (QRT-PCR). Double immunofluorescence staining was performed on paraffin-embedded human posterior sections to localize SPC family members. Western blot analysis was used to identify PACE4 isoform expression within the optic nerve head and retina. In addition, a fluorogenic SPC substrate-based assay was used to elucidate SPC enzyme activity within human retina and optic nerve head (ONH) tissues. RESULTS QPCR results indicated that PC1 and PC2 were expressed 4.1- and 5.7-fold higher in retina compared to optic nerve head, whereas PACE4 was expressed 4.1-fold higher in the ONH. PC1 and PC2 were localized primarily in neuronal cells, whereas PACE4 and PC5 were limited to the glia of the retina and optic nerve head. SPC activity in ONH lysate was significantly higher than that of retinal lysate; however, when an SPC inhibitor was added, activity in ONH decreased more than that in retina. CONCLUSIONS These results indicate that the SPCs are expressed in distinct patterns throughout the human retina and ONH. PC1 and PC2 were primarily expressed in neurons, whereas PACE4 appeared to be largely restricted to glia. Thus, elevated PACE4 may modulate the bioactivity of proteins secreted in the ONH and retina.
Collapse
Affiliation(s)
- John A Fuller
- Department of Cell Biology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA.
| | | | | | | |
Collapse
|
49
|
Lukas TJ, Miao H, Chen L, Riordan SM, Li W, Crabb AM, Wise A, Du P, Lin SM, Hernandez MR. Susceptibility to glaucoma: differential comparison of the astrocyte transcriptome from glaucomatous African American and Caucasian American donors. Genome Biol 2008; 9:R111. [PMID: 18613964 PMCID: PMC2530868 DOI: 10.1186/gb-2008-9-7-r111] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 06/18/2008] [Accepted: 07/09/2008] [Indexed: 12/23/2022] Open
Abstract
Comparison of gene expression in normal and glaucomatous eyes from Caucasian American and African American donors reveals differences that might reflect different susceptibility to glaucoma. Background Epidemiological and genetic studies indicate that ethnic/genetic background plays an important role in susceptibility to primary open angle glaucoma (POAG). POAG is more prevalent among the African-descent population compared to the Caucasian population. Damage in POAG occurs at the level of the optic nerve head (ONH) and is mediated by astrocytes. Here we investigated differences in gene expression in primary cultures of ONH astrocytes obtained from age-matched normal and glaucomatous donors of Caucasian American (CA) and African American (AA) populations using oligonucleotide microarrays. Results Gene expression data were obtained from cultured astrocytes representing 12 normal CA and 12 normal AA eyes, 6 AA eyes with POAG and 8 CA eyes with POAG. Data were normalized and significant differential gene expression levels detected by using empirical Bayesian shrinkage moderated t-statistics. Gene Ontology analysis and networks of interacting proteins were constructed using the BioGRID database. Network maps included regulation of myosin, actin, and protein trafficking. Real-time RT-PCR, western blots, ELISA, and functional assays validated genes in the networks. Conclusion Cultured AA and CA glaucomatous astrocytes retain differential expression of genes that promote cell motility and migration, regulate cell adhesion, and are associated with structural tissue changes that collectively contribute to neural degeneration. Key upregulated genes include those encoding myosin light chain kinase (MYLK), transforming growth factor-β receptor 2 (TGFBR2), rho-family GTPase-2 (RAC2), and versican (VCAN). These genes along with other differentially expressed components of integrated networks may reflect functional susceptibility to chronic elevated intraocular pressure that is enhanced in the optic nerve head of African Americans.
Collapse
Affiliation(s)
- Thomas J Lukas
- Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, E Chicago Ave, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hernandez MR, Miao H, Lukas T. Astrocytes in glaucomatous optic neuropathy. PROGRESS IN BRAIN RESEARCH 2008; 173:353-73. [DOI: 10.1016/s0079-6123(08)01125-4] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|