1
|
Li D, Hu Q, Zhan Z, Zhang X, Zeng W, Liu L, Wu K, Yu M. Increased reactive astrocytes and NLRC4-mediated neuronal pyroptosis in advanced visual structures contralateral to the optic nerve crush eye in mice. Exp Eye Res 2025; 251:110235. [PMID: 39798846 DOI: 10.1016/j.exer.2025.110235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/21/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Currently, research on optic nerve injury predominantly focuses on the retina and optic nerve, but emerging evidence suggests that optic nerve injury also affects advanced visual structures like the superior colliculus (SC) and primary visual cortex (V1 region). However, the exact mechanisms have not been fully explored. This study aims to investigate the characteristics and mechanisms of pathology in the SC and V1 region after optic nerve crush (ONC) to deepen our understanding of the central mechanism of visual injury. After unilateral ONC, visual acuity in the injured eye declined, along with thinning of the retinal nerve fiber layer, and the latency and amplitude of FVEPs decreased. Furthermore, neuronal loss and degeneration were observed in the contralateral SC and V1 region, accompanied by astrocytic activation. Additionally, protein markers C3, and Serping1 for A1 astrocytes, which had neurotoxic effects and S100A10, and PTX3 for A2 astrocytes, which promoted tissue repair, were increased in the two regions. A1 astrocytes were mainly present in the early stages of observation, while A2 astrocytes were mainly increased later. Notably, NLRC4, GSDMD-N, cleaved caspase-1 expression, and IL-1β, IL-18 secretion increased in the contralateral SC and V1 region. Collectively, our findings reveal that A1 (neurotoxic) and A2 astrocytes (neuroprotective), NLRC4-mediated neuronal pyroptosis are enhanced in SC and V1 region contralateral to the ONC eye. The primary visual cortex responds to injury later than the superior colliculus after ONC, with less pronounced damage changes. Reactive astrocytes and NLRC4 inflammasome may act as promising targets for the prevention and treatment of optic nerve injury.
Collapse
Affiliation(s)
- Deling Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Qinyuan Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Zongyi Zhan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Xinyi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Weiting Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Liling Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Kaili Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Minbin Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
2
|
Lindborg JA, Tran NM, Chenette DM, DeLuca K, Foli Y, Kannan R, Sekine Y, Wang X, Wollan M, Kim IJ, Sanes JR, Strittmatter SM. Optic nerve regeneration screen identifies multiple genes restricting adult neural repair. Cell Rep 2021; 34:108777. [PMID: 33657370 PMCID: PMC8009559 DOI: 10.1016/j.celrep.2021.108777] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/21/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Adult mammalian central nervous system (CNS) trauma interrupts neural networks and, because axonal regeneration is minimal, neurological deficits persist. Repair via axonal growth is limited by extracellular inhibitors and cell-autonomous factors. Based on results from a screen in vitro, we evaluate nearly 400 genes through a large-scale in vivo regeneration screen. Suppression of 40 genes using viral-driven short hairpin RNAs (shRNAs) promotes retinal ganglion cell (RGC) axon regeneration after optic nerve crush (ONC), and most are validated by separate CRISPR-Cas9 editing experiments. Expression of these axon-regeneration-suppressing genes is not significantly altered by axotomy. Among regeneration-limiting genes, loss of the interleukin 22 (IL-22) cytokine allows an early, yet transient, inflammatory response in the retina after injury. Reduced IL-22 drives concurrent activation of signal transducer and activator of transcription 3 (Stat3) and dual leucine zipper kinase (DLK) pathways and upregulation of multiple neuron-intrinsic regeneration-associated genes (RAGs). Including IL-22, our screen identifies dozens of genes that limit CNS regeneration. Suppression of these genes in the context of axonal damage could support improved neural repair.
Collapse
Affiliation(s)
- Jane A Lindborg
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Devon M Chenette
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Kristin DeLuca
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yram Foli
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ramakrishnan Kannan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Yuichi Sekine
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Xingxing Wang
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Marius Wollan
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - In-Jung Kim
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA.
| |
Collapse
|
3
|
Xin X, Li Y, Liu H. Hesperidin ameliorates hypobaric hypoxia-induced retinal impairment through activation of Nrf2/HO-1 pathway and inhibition of apoptosis. Sci Rep 2020; 10:19426. [PMID: 33173100 PMCID: PMC7655840 DOI: 10.1038/s41598-020-76156-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/19/2020] [Indexed: 11/09/2022] Open
Abstract
High-altitude retinopathy is initiated by hypobaric hypoxia and characterized by retinal functional changes, but the precise cellular and molecular mechanisms that mediate this dysfunction remain unclear. The aim of our investigation is to determine the protective efficacy of hesperidin (HSD) on the hypobaric hypoxia-induced damage to the retina. Experiment rats were randomly grouped as the control, hypobaric hypoxia group and HSD intervention group. The hypobaric hypoxia and the HSD intervention groups were maintained in a low-pressure oxygen cabin. We found that hypobaric hypoxia dramatically reduced nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1(HO-1) levels, induced an elevation in immunostaining of TUNEL-positive cells. Hypobaric hypoxia exposure resulted in the increase of Bcl-2, decrease of caspase3 and caspase9 expression as well as Bax level. HSD protected the retina from hypobaric hypoxia-caused impairment by enhancing Nrf2 and HO-1 activation, attenuating apoptotic caspases levels, and reducing Bax and preserving Bcl-2 expression. Additionally, oxidative stress increased poly (ADP-ribose) polymerase 1 (PARP1) and suppressed ciliary neurotrophic factor (CNTF) level, HSD treatment reverted this effect by down-regulation of PARP1 and up-regulation of CNTF expression. Taken together, our findings implicate that HSD exerts a protective role in response to hypobaric hypoxia stress by activating Nrf2/HO-1 pathway and inhibiting apoptosis.
Collapse
Affiliation(s)
- Xiaorong Xin
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan Province, China.
| | - Yanrong Li
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan Province, China
| | - Haiping Liu
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan Province, China
| |
Collapse
|
4
|
Linnerbauer M, Rothhammer V. Protective Functions of Reactive Astrocytes Following Central Nervous System Insult. Front Immunol 2020; 11:573256. [PMID: 33117368 PMCID: PMC7561408 DOI: 10.3389/fimmu.2020.573256] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022] Open
Abstract
Astrocytes play important roles in numerous central nervous system disorders including autoimmune inflammatory, hypoxic, and degenerative diseases such as Multiple Sclerosis, ischemic stroke, and Alzheimer’s disease. Depending on the spatial and temporal context, activated astrocytes may contribute to the pathogenesis, progression, and recovery of disease. Recent progress in the dissection of transcriptional responses to varying forms of central nervous system insult has shed light on the mechanisms that govern the complexity of reactive astrocyte functions. While a large body of research focuses on the pathogenic effects of reactive astrocytes, little is known about how they limit inflammation and contribute to tissue regeneration. However, these protective astrocyte pathways might be of relevance for the understanding of the underlying pathology in disease and may lead to novel targeted approaches to treat autoimmune inflammatory and degenerative disorders of the central nervous system. In this review article, we have revisited the emerging concept of protective astrocyte functions and discuss their role in the recovery from inflammatory and ischemic disease as well as their role in degenerative disorders. Focusing on soluble astrocyte derived mediators, we aggregate the existing knowledge on astrocyte functions in the maintenance of homeostasis as well as their reparative and tissue-protective function after acute lesions and in neurodegenerative disorders. Finally, we give an outlook of how these mediators may guide future therapeutic strategies to tackle yet untreatable disorders of the central nervous system.
Collapse
Affiliation(s)
- Mathias Linnerbauer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Veit Rothhammer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
5
|
Tao Y, Zhou X, Zheng X, Li S, Mou C. Deciphering the Forebrain Disorder in a Chicken Model of Cerebral Hernia. Genes (Basel) 2020; 11:E1008. [PMID: 32867218 PMCID: PMC7564858 DOI: 10.3390/genes11091008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/02/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
Cerebral hernia in crested chicken has been characterized as the protrusion of cerebral hemispheres into the unsealed skull for hundreds of years, since Charles Darwin. The development of deformed forebrain (telencephalon) of cerebral hernia remains largely unknown. Here, the unsealed frontal skull combined with misplaced sphenoid bone was observed and potentially associated with brain protuberance. The shifted pallidum, elongated hippocampus, expanded mesopallium and nidopallium, and reduced hyperpallium were observed in seven regions of the malformed telencephalon. The neurons were detected with nuclear pyknosis and decreased density. Astrocytes showed uneven distribution and disordered protuberances in hyperpallium and hippocampus. Transcriptome analyses of chicken telencephalon (cerebral hernia vs. control) revealed 547 differentially expressed genes (DEGs), mainly related to nervous system development, and immune system processes, including astrocyte marker gene GFAP, and neuron and astrocyte developmental gene S100A6. The upregulation of GFAP and S100A6 genes in abnormal telencephalon was correlated with reduced DNA methylation levels in the promoter regions. The morphological, cellular, and molecular variations in the shape, regional specification, and cellular states of malformed telencephalon potentially participate in brain plasticity and previously reported behavior changes. Chickens with cerebral hernia might be an interesting and valuable disease model to further explore the recognition, diagnosis, and therapy of cerebral hernia development of crested chickens and other species.
Collapse
Affiliation(s)
| | | | | | | | - Chunyan Mou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, HuaZhong Agricultural University, Wuhan 430070, China; (Y.T.); (X.Z.); (X.Z.); (S.L.)
| |
Collapse
|
6
|
Sala-Jarque J, Mesquida-Veny F, Badiola-Mateos M, Samitier J, Hervera A, del Río JA. Neuromuscular Activity Induces Paracrine Signaling and Triggers Axonal Regrowth after Injury in Microfluidic Lab-On-Chip Devices. Cells 2020; 9:cells9020302. [PMID: 32012727 PMCID: PMC7072511 DOI: 10.3390/cells9020302] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
Peripheral nerve injuries, including motor neuron axonal injury, often lead to functional impairments. Current therapies are mostly limited to surgical intervention after lesion, yet these interventions have limited success in restoring functionality. Current activity-based therapies after axonal injuries are based on trial-error approaches in which the details of the underlying cellular and molecular processes are largely unknown. Here we show the effects of the modulation of both neuronal and muscular activity with optogenetic approaches to assess the regenerative capacity of cultured motor neuron (MN) after lesion in a compartmentalized microfluidic-assisted axotomy device. With increased neuronal activity, we observed an increase in the ratio of regrowing axons after injury in our peripheral-injury model. Moreover, increasing muscular activity induces the liberation of leukemia inhibitory factor and glial cell line-derived neurotrophic factor in a paracrine fashion that in turn triggers axonal regrowth of lesioned MN in our 3D hydrogel cultures. The relevance of our findings as well as the novel approaches used in this study could be useful not only after axotomy events but also in diseases affecting MN survival.
Collapse
Affiliation(s)
- Julia Sala-Jarque
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Francina Mesquida-Veny
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
| | - Maider Badiola-Mateos
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28029 Madrid, Spain
- Department of Electronics and Biomedical Engineering, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Arnau Hervera
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: (A.H.); (J.A.d.R.)
| | - José Antonio del Río
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (J.S.-J.); (F.M.-V.); (M.B.-M.); (J.S.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: (A.H.); (J.A.d.R.)
| |
Collapse
|
7
|
Pöyhönen S, Er S, Domanskyi A, Airavaara M. Effects of Neurotrophic Factors in Glial Cells in the Central Nervous System: Expression and Properties in Neurodegeneration and Injury. Front Physiol 2019; 10:486. [PMID: 31105589 PMCID: PMC6499070 DOI: 10.3389/fphys.2019.00486] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 04/08/2019] [Indexed: 12/28/2022] Open
Abstract
Astrocytes, oligodendrocytes, and microglia are abundant cell types found in the central nervous system and have been shown to play crucial roles in regulating both normal and disease states. An increasing amount of evidence points to the critical importance of glia in mediating neurodegeneration in Alzheimer’s and Parkinson’s diseases (AD, PD), and in ischemic stroke, where microglia are involved in initial tissue clearance, and astrocytes in the subsequent formation of a glial scar. The importance of these cells for neuronal survival has previously been studied in co-culture experiments and the search for neurotrophic factors (NTFs) initiated after finding that the addition of conditioned media from astrocyte cultures could support the survival of primary neurons in vitro. This led to the discovery of the potent dopamine neurotrophic factor, glial cell line-derived neurotrophic factor (GDNF). In this review, we focus on the relationship between glia and NTFs including neurotrophins, GDNF-family ligands, CNTF family, and CDNF/MANF-family proteins. We describe their expression in astrocytes, oligodendrocytes and their precursors (NG2-positive cells, OPCs), and microglia during development and in the adult brain. Furthermore, we review existing data on the glial phenotypes of NTF knockout mice and follow NTF expression patterns and their effects on glia in disease models such as AD, PD, stroke, and retinal degeneration.
Collapse
Affiliation(s)
- Suvi Pöyhönen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Safak Er
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.,Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
8
|
Inhibition of Microglia-Derived Oxidative Stress by Ciliary Neurotrophic Factor Protects Dopamine Neurons In Vivo from MPP⁺ Neurotoxicity. Int J Mol Sci 2018; 19:ijms19113543. [PMID: 30423807 PMCID: PMC6274815 DOI: 10.3390/ijms19113543] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 01/03/2023] Open
Abstract
We demonstrated that capsaicin (CAP), an agonist of transient receptor potential vanilloid subtype 1 (TRPV1), inhibits microglia activation and microglia-derived oxidative stress in the substantia nigra (SN) of MPP⁺-lesioned rat. However, the detailed mechanisms how microglia-derived oxidative stress is regulated by CAP remain to be determined. Here we report that ciliary neurotrophic factor (CNTF) endogenously produced by CAP-activated astrocytes through TRPV1, but not microglia, inhibits microglial activation and microglia-derived oxidative stress, as assessed by OX-6 and OX-42 immunostaining and hydroethidine staining, respectively, resulting in neuroprotection. The significant increase in levels of CNTF receptor alpha (CNTFRα) expression was evident on microglia in the MPP⁺-lesioned rat SN and the observed beneficial effects of CNTF was abolished by treatment with CNTF receptor neutralizing antibody. It is therefore likely that CNTF can exert its effect via CNTFRα on microglia, which rescues dopamine neurons in the SN of MPP⁺-lesioned rats and ameliorates amphetamine-induced rotations. Immunohistochemical analysis revealed also a significantly increased expression of CNTFRα on microglia in the SN from human Parkinson's disease patients compared with age-matched controls, indicating that these findings may have relevance to the disease. These data suggest that CNTF originated from TRPV1 activated astrocytes may be beneficial to treat neurodegenerative disease associated with neuro-inflammation such as Parkinson's disease.
Collapse
|
9
|
Echevarria FD, Formichella CR, Sappington RM. Interleukin-6 Deficiency Attenuates Retinal Ganglion Cell Axonopathy and Glaucoma-Related Vision Loss. Front Neurosci 2017; 11:318. [PMID: 28620279 PMCID: PMC5450377 DOI: 10.3389/fnins.2017.00318] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/19/2017] [Indexed: 11/13/2022] Open
Abstract
The pleotropic cytokine interleukin-6 (IL-6) is implicated in retinal ganglion cell (RGC) survival and degeneration, including that associated with glaucoma. IL-6 protects RGCs from pressure-induced apoptosis in vitro. However, it is unknown how IL-6 impacts glaucomatous degeneration in vivo. To study how IL-6 influences glaucomatous RGC axonopathy, accompanying glial reactivity, and resultant deficits in visual function, we performed neural tracing, histological, and neurobehavioral assessments in wildtype (B6;129SF2/J; WT) and IL-6 knock-out mice (B6;129S2-IL6tm1kopf/J; IL-6-/-) after 8 weeks of unilateral or bilateral microbead-induced glaucoma (microbead occlusion model). IOP increased by 20% following microbead injection in both genotypes (p < 0.05). However, deficits in wound healing at the site of corneal injection were noted. In WT mice, elevated IOP produced degenerating axon profiles and decreased axon density in the optic nerve by 15% (p < 0.01). In IL-6-/- mice, axon density in the optic nerve did not differ between microbead- and saline-injected mice (p > 0.05) and degenerating axon profiles were minimal. Preservation of RGC axons was reflected in visual function, where visual acuity decreased significantly in a time-dependent manner with microbead-induced IOP elevation in WT (p < 0.001), but not IL-6-/- mice (p > 0.05). Despite this preservation of RGC axons and visual acuity, both microbead-injected WT and IL-6-/- mice exhibited a 50% decrease in anterograde CTB transport to the superior colliculus, as compared to saline-injected controls (p < 0.01). Assessment of glial reactivity revealed no genotype- or IOP-dependent changes in retinal astrocytes. IOP elevation decreased microglia density and percent retinal area covered in WT mice (p < 0.05), while IL-6-/- mice exhibited only a decrease in density (p < 0.05). Together, our findings indicate that two defining features of RGC axonopathy—axon transport deficits and structural degeneration of axons—likely occur via independent mechanisms. Our data suggest that IL-6 is part of a mechanism that specifically leads to structural degeneration of axons. Furthermore, its absence is sufficient to prevent both structural degeneration of the optic nerve and vision loss. Overall, our work supports the proposition that functional deficits in axon transport represent a therapeutic window for RGC axonopathy and identify IL-6 signaling as a strong target for such a therapeutic.
Collapse
Affiliation(s)
| | - Cathryn R Formichella
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of MedicineNashville, TN, United States.,Vanderbilt Eye Institute, Vanderbilt University Medical CenterNashville, TN, United States
| | - Rebecca M Sappington
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of MedicineNashville, TN, United States.,Vanderbilt Eye Institute, Vanderbilt University Medical CenterNashville, TN, United States.,Department of Pharmacology, Vanderbilt University School of MedicineNashville, TN, United States
| |
Collapse
|
10
|
Sub-Chronic Neuropathological and Biochemical Changes in Mouse Visual System after Repetitive Mild Traumatic Brain Injury. PLoS One 2016; 11:e0153608. [PMID: 27088355 PMCID: PMC4835061 DOI: 10.1371/journal.pone.0153608] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 03/31/2016] [Indexed: 11/29/2022] Open
Abstract
Repetitive mild traumatic brain injury (r-mTBI) results in neuropathological and biochemical consequences in the human visual system. Using a recently developed mouse model of r-mTBI, with control mice receiving repetitive anesthesia alone (r-sham) we assessed the effects on the retina and optic nerve using histology, immunohistochemistry, proteomic and lipidomic analyses at 3 weeks post injury. Retina tissue was used to determine retinal ganglion cell (RGC) number, while optic nerve tissue was examined for cellularity, myelin content, protein and lipid changes. Increased cellularity and areas of demyelination were clearly detectable in optic nerves in r-mTBI, but not in r-sham. These changes were accompanied by a ~25% decrease in the total number of Brn3a-positive RGCs. Proteomic analysis of the optic nerves demonstrated various changes consistent with a negative effect of r-mTBI on major cellular processes like depolymerization of microtubules, disassembly of filaments and loss of neurons, manifested by decrease of several proteins, including neurofilaments (NEFH, NEFM, NEFL), tubulin (TUBB2A, TUBA4A), microtubule-associated proteins (MAP1A, MAP1B), collagen (COL6A1, COL6A3) and increased expression of other proteins, including heat shock proteins (HSP90B1, HSPB1), APOE and cathepsin D. Lipidomic analysis showed quantitative changes in a number of phospholipid species, including a significant increase in the total amount of lysophosphatidylcholine (LPC), including the molecular species 16:0, a known demyelinating agent. The overall amount of some ether phospholipids, like ether LPC, ether phosphatidylcholine and ether lysophosphatidylethanolamine were also increased, while the majority of individual molecular species of ester phospholipids, like phosphatidylcholine and phosphatidylethanolamine, were decreased. Results from the biochemical analysis correlate well with changes detected by histological and immunohistochemical methods and indicate the involvement of several important molecular pathways. This will allow future identification of therapeutic targets for improving the visual consequences of r-mTBI.
Collapse
|
11
|
Role of leukemia inhibitory factor in the nervous system and its pathology. Rev Neurosci 2015; 26:443-59. [DOI: 10.1515/revneuro-2014-0086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/22/2015] [Indexed: 11/15/2022]
Abstract
AbstractLeukemia inhibitory factor (LIF) is a multifunction cytokine that has various effects on different tissues and cell types in rodents and humans; however, its insufficiency has a relatively mild impact. This could explain why only some aspects of LIF activity are in the limelight, whereas other aspects are not well known. In this review, the LIF structure, signaling pathway, and primary roles in the development and function of an organism are reviewed, and the effects of LIF on stem cell growth and differentiation, which are important for its use in cell culturing, are described. The focus is on the roles of LIF in central nervous system development and on the modulation of its physiological functions as well as the involvement of LIF in the pathogenesis of brain diseases and injuries. Finally, LIF and its signaling pathway are discussed as potential targets of therapeutic interventions to influence both negative phenomena and regenerative processes following brain injury.
Collapse
|
12
|
Yang XT, Huang GH, Feng DF, Chen K. Insight into astrocyte activation after optic nerve injury. J Neurosci Res 2014; 93:539-48. [PMID: 25257183 DOI: 10.1002/jnr.23487] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/25/2014] [Accepted: 08/29/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Xi-Tao Yang
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Guo-Hui Huang
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Dong-Fu Feng
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
- Institute of Traumatic Medicine; Shanghai Jiaotong University School of Medicine; Shanghai China
| | - Kui Chen
- Department of Neurosurgery, No. 3 People's Hospital; Shanghai Jiaotong University School of Medicine; Shanghai China
| |
Collapse
|
13
|
The emerging use of in vivo optical imaging in the study of neurodegenerative diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:401306. [PMID: 25147799 PMCID: PMC4132409 DOI: 10.1155/2014/401306] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/25/2014] [Accepted: 06/26/2014] [Indexed: 12/12/2022]
Abstract
The detection and subsequent quantification of photons emitted from living tissues, using highly sensitive charged-couple device (CCD) cameras, have enabled investigators to noninvasively examine the intricate dynamics of molecular reactions in wide assortment of experimental animals under basal and pathophysiological conditions. Nevertheless, extrapolation of this in vivo optical imaging technology to the study of the mammalian brain and related neurodegenerative conditions is still in its infancy. In this review, we introduce the reader to the emerging use of in vivo optical imaging in the study of neurodegenerative diseases. We highlight the current instrumentation that is available and reporter molecules (fluorescent and bioluminescent) that are commonly used. Moreover, we examine how in vivo optical imaging using transgenic reporter mice has provided new insights into Alzheimer's disease, amyotrophic lateral sclerosis (ALS), Prion disease, and neuronal damage arising from excitotoxicity and inflammation. Furthermore, we also touch upon studies that have utilized these technologies for the development of therapeutic strategies for neurodegenerative conditions that afflict humans.
Collapse
|
14
|
Beatus P, Jhaveri DJ, Walker TL, Lucas PG, Rietze RL, Cooper HM, Morikawa Y, Bartlett PF. Oncostatin M regulates neural precursor activity in the adult brain. Dev Neurobiol 2011; 71:619-33. [PMID: 21671408 DOI: 10.1002/dneu.20871] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The regulation of neural precursor cell (NPC) activity is the major determinant of the rate of neuronal production in neurogenic regions of the adult brain. Here, we show that Oncostatin M (Osm) and its receptor, OsmRβ, are both expressed in the subventricular zone (SVZ) and that in contradistinction to leukemia inhibitory factor and ciliary neutrophic factor, Osm directly inhibits the proliferation of adult NPCs as measured by a decreased level of neurosphere formation in vitro. Similarly, intraventricular infusion of Osm dramatically decreases the pool of NPCs in both the SVZ and the hippocampus. In keeping with the inhibitory action of Osm, we reveal that mice lacking OsmRβ have substantially more NPCs in the SVZ, the hippocampus and the olfactory bulb, demonstrating that endogenous Osm signaling is important for NPC homeostasis. Finally, we show that Osm can also inhibit clonal growth of glioblastoma-derived neurospheres, further supporting the close link between NPCs and tumor stem cells.
Collapse
Affiliation(s)
- Paul Beatus
- The Queensland Brain Institute, The University of Queensland, Brisbane QLD 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Tolosa L, Caraballo-Miralles V, Olmos G, Lladó J. TNF-α potentiates glutamate-induced spinal cord motoneuron death via NF-κB. Mol Cell Neurosci 2010; 46:176-86. [PMID: 20849956 DOI: 10.1016/j.mcn.2010.09.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 08/31/2010] [Accepted: 09/01/2010] [Indexed: 12/14/2022] Open
Abstract
Besides glutamate excitotoxicity, the neuroinflammatory response is emerging as a relevant contributor to motoneuron loss in amyotrophic lateral sclerosis (ALS). In this regard, high levels of circulating proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) have been shown both in human patients and in animal models of ALS. The aim of this work was to study the effects of TNF-α on glutamate-induced excitotoxicity in spinal cord motoneurons. In rat spinal cord organotypic cultures chronic glutamate excitotoxicity, induced by the glutamate-uptake inhibitor threohydroxyaspartate (THA), resulted in motoneuron loss that was associated with a neuroinflammatory response. In the presence of TNF-α, THA-induced excitotoxic motoneuron death was potentiated. Co-exposure to TNF-α and THA also resulted in down-regulation of the astroglial glutamate transporter 1 (GLT-1) and in increased extracellular glutamate levels, which were prevented by nuclear factor-kappaB (NF-κB) inhibition. Furthermore, TNF-α and THA also cooperated in the induction of oxidative stress in a mechanism involving the NF-κB signalling pathway as well. The inhibition of this pathway abrogated the exacerbation of glutamate-mediated motoneuron death induced by TNF-α. These data link two important pathogenic mechanisms, excitotoxicity and neuroinflammation, suggested to play a role in ALS and, to our knowledge, this is the first time that TNF-α-induced NF-κB activation has been reported to potentiate glutamate excitotoxicity on motononeurons.
Collapse
Affiliation(s)
- Laia Tolosa
- Grup de Neurobiologia Cel·lular, Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS)/Departament de Biologia, Universitat de les Illes Balears, Cra. de Valldemossa km 7.5,E-07122 Palma de Mallorca, Spain
| | | | | | | |
Collapse
|
16
|
Kirsch M, Trautmann N, Ernst M, Hofmann HD. Involvement of gp130-associated cytokine signaling in Müller cell activation following optic nerve lesion. Glia 2010; 58:768-79. [PMID: 20091786 DOI: 10.1002/glia.20961] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ciliary neurotrophic factor (CNTF) and the related cytokine leukemia inhibitory factor (LIF) have been implicated in regulating astrogliosis following CNS lesions. Application of the factors activates astrocytes in vivo and in vitro, and their expression as well as their receptors is upregulated after brain injury. Here, we investigated their function by studying Müller cell activation induced by optic nerve crush in CNTF- and LIF-deficient mice, and in animals with deficiencies in cytokine signaling pathways. In the retina of CNTF(-/-) mice, basal GFAP expression was reduced, but unexpectedly, injury-induced upregulation in activated Müller cells was increased during the first 3 days after lesion as compared to wild-type animals and this corresponded with higher phosphorylation level of STAT3, an indicator of cytokine signaling. The observation that LIF expression was strongly upregulated in CNTF(-/-) mice but not in wild-type animals following optic nerve lesion provided a possible explanation. In fact, additional ablation of the LIF gene in CNTF/LIF double knockout mice almost completely abolished early lesion-induced GFAP upregulation in Müller cells and STAT3 phosphorylation. Early Müller cell activation was also eliminated in LIF(-/-) mice, despite normal CNTF levels, as well as in mutants deficient in gp130/JAK/STAT signaling and in conditional STAT3 knockout mice. Our results demonstrate that LIF signaling via the gp130/JAK/STAT3 pathway is required for the initiation of the astrogliosis-like reaction of retinal Müller cells after optic nerve injury. A potential role of CNTF was possibly masked by a compensatory increase in LIF signaling in the absence of CNTF.
Collapse
Affiliation(s)
- Matthias Kirsch
- Institute of Anatomy and Cell Biology, University of Freiburg, Albertstr. 23, Freiburg, Germany
| | | | | | | |
Collapse
|
17
|
Postnov DE, Koreshkov RN, Brazhe NA, Brazhe AR, Sosnovtseva OV. Dynamical patterns of calcium signaling in a functional model of neuron-astrocyte networks. J Biol Phys 2009; 35:425-45. [PMID: 19669421 PMCID: PMC2750744 DOI: 10.1007/s10867-009-9156-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 04/15/2009] [Indexed: 11/26/2022] Open
Abstract
We propose a functional mathematical model for neuron-astrocyte networks. The model incorporates elements of the tripartite synapse and the spatial branching structure of coupled astrocytes. We consider glutamate-induced calcium signaling as a specific mode of excitability and transmission in astrocytic-neuronal networks. We reproduce local and global dynamical patterns observed experimentally.
Collapse
Affiliation(s)
- D. E. Postnov
- Physics Department, Saratov State University, Astrakhanskaya Street 83, Saratov, 410012 Russia
| | - R. N. Koreshkov
- Physics Department, Saratov State University, Astrakhanskaya Street 83, Saratov, 410012 Russia
| | - N. A. Brazhe
- Biophysics Department, Biological Faculty, Moscow State University, Leninskie gory 1, Building 12, 119991 Moscow, Russia
| | - A. R. Brazhe
- Biophysics Department, Biological Faculty, Moscow State University, Leninskie gory 1, Building 12, 119991 Moscow, Russia
| | - O. V. Sosnovtseva
- Department of Physics, Technical University of Denmark, Building 309, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
18
|
Astrocytes in the damaged brain: molecular and cellular insights into their reactive response and healing potential. Biochem Pharmacol 2009; 79:77-89. [PMID: 19765548 DOI: 10.1016/j.bcp.2009.09.014] [Citation(s) in RCA: 249] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 09/08/2009] [Accepted: 09/10/2009] [Indexed: 12/29/2022]
Abstract
Long considered merely a trophic and mechanical support to neurons, astrocytes have progressively taken the center stage as their ability to react to acute and chronic neurodegenerative situations became increasingly clear. Reactive astrogliosis starts when trigger molecules produced at the injury site drive astrocytes to leave their quiescent state and become activated. Distinctive morphological and biochemical features characterize this process (cell hypertrophy, upregulation of intermediate filaments, and increased cell proliferation). Moreover, reactive astrocytes migrate towards the injured area to constitute the glial scar, and release factors mediating the tissue inflammatory response and remodeling after lesion. A novel view of astrogliosis derives from the finding that subsets of reactive astrocytes can recapitulate stem cell/progenitor features after damage, fostering the concept of astroglia as a promising target for reparative therapies. But which biochemical/signaling pathways modulate astrogliosis with respect to both the time after injury and the type of damage? Are reactive astrocytes overall beneficial or detrimental for neuroprotection and tissue regeneration? This debate has been animating this research field for several years now, and an integrated view on the results obtained and the possible future perspectives is needed. With this Commentary article we have attempted to answer the above-mentioned questions by reviewing the current knowledge on the molecular mechanisms controlling and sustaining the reaction of astroglia to injury and its stem cell-like properties. Moreover, the cellular/molecular mechanisms supporting the detrimental or beneficial features of astrogliosis have been scrutinized to gain insights on possible pharmacological approaches to enhance astrocyte neuroprotective activities.
Collapse
|
19
|
Wallerian degeneration and axonal regeneration after sciatic nerve crush are altered in ICAM-1-deficient mice. Cell Tissue Res 2009; 338:19-28. [DOI: 10.1007/s00441-009-0837-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 06/26/2009] [Indexed: 01/09/2023]
|
20
|
Oku H, Fukuhara M, Kurimoto T, Okuno T, Sugiyama T, Ikeda T. Endothelin-1 (ET-1) is Increased in Rat Retina After Crushing Optic Nerve. Curr Eye Res 2009; 33:611-20. [DOI: 10.1080/02713680802213614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
21
|
Ciliary neurotrophic factor (CNTF) plus soluble CNTF receptor alpha increases cyclooxygenase-2 expression, PGE2 release and interferon-gamma-induced CD40 in murine microglia. J Neuroinflammation 2009; 6:7. [PMID: 19267906 PMCID: PMC2660310 DOI: 10.1186/1742-2094-6-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Accepted: 03/06/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ciliary neurotrophic factor (CNTF) has been regarded as a potent trophic factor for motor neurons. However, recent studies have shown that CNTF exerts effects on glial cells as well as neurons. For instance, CNTF stimulates astrocytes to secrete FGF-2 and rat microglia to secrete glial cell line-derived neurotrophic factor (GDNF), which suggest that CNTF exerts effects on astrocytes and microglia to promote motor neuron survival indirectly. As CNTF is structurally related to IL-6, which can stimulate immune functions of microglia, we hypothesized that CNTF might exert similar effects. METHODS We performed 2-D and 1-D proteomic experiments with western blotting and flow cytometry to examine effects of CNTF on primary microglia derived from neonatal mouse brains. RESULTS We show that murine microglia express CNTF receptor alpha (CNTFRalpha), which can be induced by interferon-gamma (IFNgamma). Whereas IL-6 activated STAT-3 and ERK phosphorylation, CNTF did not activate these pathways, nor did CNTF increase p38 MAP kinase phosphorylation. Using 2-D western blot analysis, we demonstrate that CNTF induced the dephosphorylation of a set of proteins and phosphorylation of a different set. Two proteins that were phosphorylated upon CNTF treatment were the LYN substrate-1 and beta-tubulin 5. CNTF weakly stimulated microglia, whereas a stronger response was obtained by adding exogenous soluble CNTFRalpha (sCNTFRalpha) as has been observed for IL-6. When used in combination, CNTF and sCNTFRalpha collaborated with IFNgamma to increase microglial surface expression of CD40 and this effect was quite pronounced when the microglia were differentiated towards dendritic-like cells. CNTF/sCNTFRalpha complex, however, failed to increase MHC class II expression beyond that induced by IFNgamma. The combination of CNTF and sCNTFRalpha, but not CNTF alone, enhanced microglial Cox-2 protein expression and PGE2 secretion (although CNTF was 30 times less potent than LPS). Surprisingly, Cox-2 production was enhanced 2-fold, rather than being inhibited, upon addition of a gp130 blocking antibody. CONCLUSION Our studies indicate that CNTF can activate microglia and dendritic-like microglia similar to IL-6; however, unlike IL-6, CNTF does not stimulate the expected signaling pathways in microglia, nor does it appear to require gp130.
Collapse
|
22
|
Vacher CM, Crépin D, Aubourg A, Couvreur O, Bailleux V, Nicolas V, Férézou J, Gripois D, Gertler A, Taouis M. A putative physiological role of hypothalamic CNTF in the control of energy homeostasis. FEBS Lett 2008; 582:3832-8. [PMID: 18950628 DOI: 10.1016/j.febslet.2008.10.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Revised: 08/26/2008] [Accepted: 10/10/2008] [Indexed: 10/21/2022]
Abstract
Administration of CNTF durably reduces food intake and body weight in obese humans and rodent models. However, the involvement of endogenous CNTF in the central regulation of energy homeostasis needs to be elucidated. Here, we demonstrate that CNTF and its receptor are expressed in the arcuate nucleus, a key hypothalamic region controlling food intake, and that CNTF levels are inversely correlated to body weight in rats fed a high-sucrose diet. Thus endogenous CNTF may act, in some individuals, as a protective factor against weight gain during hypercaloric diet and could account for individual differences in the susceptibility to obesity.
Collapse
Affiliation(s)
- C-M Vacher
- Laboratoire de Neuroendocrinologie Moléculaire de la Prise Alimentaire, UMR 1197 INRA, Université Paris 11, Bâtiment 447, 91405 Orsay, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Krady JK, Lin HW, Liberto CM, Basu A, Kremlev SG, Levison SW. Ciliary neurotrophic factor and interleukin-6 differentially activate microglia. J Neurosci Res 2008; 86:1538-47. [DOI: 10.1002/jnr.21620] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
24
|
GFAPpromoter elements required for region-specific and astrocyte-specific expression. Glia 2008; 56:481-93. [DOI: 10.1002/glia.20622] [Citation(s) in RCA: 241] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
Bauer S, Kerr BJ, Patterson PH. The neuropoietic cytokine family in development, plasticity, disease and injury. Nat Rev Neurosci 2007; 8:221-32. [PMID: 17311007 DOI: 10.1038/nrn2054] [Citation(s) in RCA: 290] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neuropoietic cytokines are well known for their role in the control of neuronal, glial and immune responses to injury or disease. Since this discovery, it has emerged that several of these proteins are also involved in nervous system development, in particular in the regulation of neurogenesis and stem cell fate. Recent data indicate that these proteins have yet more functions, as key modulators of synaptic plasticity and of various behaviours. In addition, neuropoietic cytokines might be a factor in the aetiology of psychiatric disorders.
Collapse
Affiliation(s)
- Sylvian Bauer
- Physiologie Neurovégétative, UMR 6153 CNRS, 1147 INRA, Université Paul Cézanne-Aix-Marseille-3, Ave. Escadrille Normandie-Niemen, BP 351-352, 13397 Marseille Cedex 20, France
| | | | | |
Collapse
|
26
|
Naumann T, Steup A, Schnell O, Schubert KO, Zhi Q, Guijarro C, Kirsch M, Hofmann HD. Altered neuronal responses and regulation of neurotrophic proteins in the medial septum following fimbria-fornix transection in CNTF- and leukaemia inhibitory factor-deficient mice. Eur J Neurosci 2006; 24:2223-32. [PMID: 17074046 DOI: 10.1111/j.1460-9568.2006.05104.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Degeneration of axotomized GABAergic septohippocampal neurones has been shown to be enhanced in ciliary neurotrophic factor (CNTF)-deficient mice following fimbria-fornix transection (FFT), indicating a neuroprotective function of endogenous CNTF. Paradoxically, however, the cholinergic population of septohippocampal neurones was more resistant to axotomy in these mutants. As leukaemia inhibitory factor (LIF) has been identified as a potential neuroprotective factor for the cholinergic medial septum (MS) neurones, FFT-induced responses were compared in CNTF(-/-), LIF(-/-) and CNTF/LIF double knockout mice. In CNTF(-/-) mice, FFT-induced cholinergic degeneration was confirmed to be attenuated as compared with wildtype mice. The expression of both LIF and LIF receptor beta was increased in the MS providing a possible explanation for the enhanced neuronal resistance to FFT in these animals. However, ablation of the LIF gene also produced paradoxical effects; following FFT in LIF(-/-) mice no loss of GABAergic or cholinergic MS neurones was detectable during the first postlesional week, suggesting that other efficient neuroprotective mechanisms are activated in these animals. In fact, enhanced activation of astrocytes, a source of neurotrophic proteins, was indicated by increased up-regulation of glial fibrillary acidic protein and vimentin expression. In addition, mRNA levels for neurotrophin signalling components (e.g. nerve growth factor, p75(NTR)) were differentially regulated. The positive effect on axotomized cholinergic neurones seen in CNTF(-/-) and LIF(-/-) mice as well as the increased up-regulation of astrogliose markers was abolished in CNTF/LIF double knockout animals. Our results indicate that endogenous CNTF and LIF are involved in the regulation of neuronal survival following central nervous system lesion and are integrated into a network of neurotrophic signals that mutually influence their expression and function.
Collapse
Affiliation(s)
- Thomas Naumann
- Institute of Anatomy and Cell Biology, Center of Neuroscience, Albertstrasse 21, D-79104 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Deleyrolle L, Marchal-Victorion S, Dromard C, Fritz V, Saunier M, Sabourin JC, Tran Van Ba C, Privat A, Hugnot JP. Exogenous and Fibroblast Growth Factor 2/Epidermal Growth Factor-Regulated Endogenous Cytokines Regulate Neural Precursor Cell Growth and Differentiation. Stem Cells 2006; 24:748-62. [PMID: 16166253 DOI: 10.1634/stemcells.2005-0138] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neurospheres (NSs) are clonal cellular aggregates composed of neural stem cells and progenitors. A comprehensive description of their proliferation and differentiation regulation is an essential prerequisite for their use in biotherapies. Cytokines are essential molecules regulating cell precursor fate. Using a gene-array strategy, we conducted a descriptive and functional analysis of endogenous cytokines and receptors expressed by spinal cord-derived NSs during their growth or their differentiation into neuronal and glial cells. NSs were found to express approximately 100 receptor subunits and cytokine/secreted developmental factors. Several angiogenic factors and receptors that could mediate neural precursor cell-endothelial cell relationships were detected. Among them, receptor B for endothelins was highly expressed, and endothelins were found to increase NS growth. In contrast, NSs express receptors for ciliary neurotrophic factor (CNTF), bone morphogenetic protein (BMP), interferon (IFN)-gamma, or tumor necrosis factor (TNF)-alpha, which, when added in the growth phase, led to a dramatic growth reduction followed by a reduction or a loss of oligodendrocyte formation on differentiation. In addition, NSs synthesize fibroblast growth factor 2/epidermal growth factor (FGF2/EGF)-regulated endogenous cytokines that participate in their growth and differentiation. Notably, BMP-7 and CNTF were expressed during expansion, but upon differentiation there was a remarkable switch from BMP-7 to BMP-4 and -6 and a sharp increase of CNTF. Reintroduction of growth factors reverses the BMP expression profile, indicating growth factor-BMP cross-regulations. The role of endogenous CNTF was investigated by deriving NSs from CNTF knockout mice. These NSs have an increased growth rate associated with reduction of apoptosis and generate astrocytes with a reduced glial fibulary acidic protein (GFAP) content. These results demonstrate the combined role of endogenous and exogenous cytokines in neural precursor cell growth and differentiation.
Collapse
Affiliation(s)
- Loïc Deleyrolle
- INSERM U583, INM-Hôpital Saint Eloi, 80 rue Augustin Fliche, 34295 Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Conley L, Geurs TL, Levin LA. Transcriptional regulation of ceruloplasmin by an IL-6 response element pathway. ACTA ACUST UNITED AC 2006; 139:235-41. [PMID: 15979198 DOI: 10.1016/j.molbrainres.2005.05.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2004] [Revised: 05/10/2005] [Accepted: 05/15/2005] [Indexed: 11/25/2022]
Abstract
Cp is an acute phase reactant protein that also acts as a ferroxidase, and thus indirectly decreases the production of the reactive oxygen species hydroxyl radical. Ceruloplasmin (Cp) expression is induced by a variety of central nervous system injuries, but the mechanism by which this occurs is unclear. Based on the fact that peripheral nerve injury induces interleukin-6 (IL-6) expression and that there are three IL-6 response elements in the upstream region of the Cp gene, we studied their role in transcriptional regulation of Cp in astrocytic C6 glioma cells, using transfection of a rat Cp-luciferase construct, followed by sequential and simultaneous mutation of the IL-6 response elements. We found that 0.8 kb of sequence upstream to the rat ceruloplasmin start site was sufficient to drive luciferase expression in C6 glioma cells. Cells transfected with Cp-luc and treated with 100 ng/ml rat IL-6 induced 216.8% +/- 4.6% of control activity. Mutagenesis of the IL-6 response elements decreased luciferase activity, with the maximal decline (9.7 +/- 0.7% of wild-type) after mutation of the second site. Mutagenesis of multiple sites decreased activity beyond mutagenesis of single sites with mutation of all three sites decreasing activity to 5.3 +/- 0.4% of wild-type. Gel shift and supershift assays indicated that activation of Cp in these cells was not via STAT-3. These results are consistent with a signaling process via IL-6 response elements for Cp upregulation.
Collapse
Affiliation(s)
- Laurie Conley
- Department of Ophthalmology and Visual Sciences, University of Wisconsin Medical School, 600 Highland Avenue, Madison, WI 53792, USA
| | | | | |
Collapse
|
29
|
Barnabé-Heider F, Wasylnka JA, Fernandes KJL, Porsche C, Sendtner M, Kaplan DR, Miller FD. Evidence that Embryonic Neurons Regulate the Onset of Cortical Gliogenesis via Cardiotrophin-1. Neuron 2005; 48:253-65. [PMID: 16242406 DOI: 10.1016/j.neuron.2005.08.037] [Citation(s) in RCA: 271] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Revised: 05/17/2005] [Accepted: 08/25/2005] [Indexed: 11/30/2022]
Abstract
Precursor cells of the embryonic cortex sequentially generate neurons and then glial cells, but the mechanisms regulating this neurogenic-to-gliogenic transition are unclear. Using cortical precursor cultures, which temporally mimic this in vivo differentiation pattern, we demonstrate that cortical neurons synthesize and secrete the neurotrophic cytokine cardiotrophin-1, which activates the gp130-JAK-STAT pathway and is essential for the timed genesis of astrocytes in vitro. Our data indicate that a similar phenomenon also occurs in vivo. In utero electroporation of neurotrophic cytokines in the environment of embryonic cortical precursors causes premature gliogenesis, while acute perturbation of gp130 in cortical precursors delays the normal timed appearance of astrocytes. Moreover, the neonatal cardiotrophin-1-/- cortex contains fewer astrocytes. Together, these results describe a neural feedback mechanism; newly born neurons produce cardiotrophin-1, which instructs multipotent cortical precursors to generate astrocytes, thereby ensuring that gliogenesis does not occur until neurogenesis is largely complete.
Collapse
|
30
|
Schubert KO, Naumann T, Schnell O, Zhi Q, Steup A, Hofmann HD, Kirsch M. Activation of STAT3 signaling in axotomized neurons and reactive astrocytes after fimbria-fornix transection. Exp Brain Res 2005; 165:520-31. [PMID: 15991029 DOI: 10.1007/s00221-005-2330-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Accepted: 03/01/2005] [Indexed: 10/25/2022]
Abstract
It is an open question to what extent neuroprotective mechanisms involving neurotrophic proteins are activated after central nervous system (CNS) lesions. Results from previous studies have indicated that ciliary neurotrophic factor (CNTF) and other members of the family of gp130-associated cytokines have neuroprotective effects on septohippocampal projection neurons axotomized by fimbria-fornix transection (FFT). Here we demonstrate that the transcription factor STAT3, a component of the primary cytokine signal transduction pathway, is transiently activated after FFT in the medial septum and in the lateral septum deafferented by the lesion. Immunocytochemical double-labeling showed nuclear signals for phosphorylated STAT3 in both types (GABAergic and cholinergic) of axotomized medial septal neurons around day 4 postlesion. This response temporally coincided with the cell-type-specific upregulation of the cytokine receptor components CNTF receptor alpha and LIF receptor beta in the same neurons. However, neuronal STAT3-activation was not abolished in CNTF- or LIF-deficient mouse mutants. Furthermore, lesion-induced STAT3 signaling was also found in reactive GFAP-positive astrocytes of the medial and lateral septum. Interestingly, basal GFAP expression was reduced but postlesional upregulation was markedly enhanced in CNTF(-/-) animals. These results demonstrate transient activation of cytokine signaling after CNS lesion and suggest that gp130-associated cytokines have multiple functions in the lesioned CNS by directly acting on axotomized neurons and on reactive astrocytes.
Collapse
Affiliation(s)
- Klaus Oliver Schubert
- Institute of Anatomy and Cell Biology I, University of Freiburg, P.O. Box 111, 79001, Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
van Adel BA, Arnold JM, Phipps J, Doering LC, Ball AK. Ciliary neurotrophic factor protects retinal ganglion cells from axotomy-induced apoptosis via modulation of retinal gliain vivo. ACTA ACUST UNITED AC 2005; 63:215-34. [PMID: 15719421 DOI: 10.1002/neu.20117] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Adenoviral-mediated transfer of ciliary neurotrophic factor (CNTF) to the retina rescued retinal ganglion cells (RGCs) from axotomy-induced apoptosis, presumably via activation of the high affinity CNTF receptor alpha (CNTFRalpha) expressed on RGCs. CNTF can also activate astrocytes, via its low affinity leukemia inhibitory receptor beta expressed on mature astrocytes, suggesting that CNTF may also protect injured neurons indirectly by modulating glia. Adenoviral-mediated overexpression of CNTF in normal and axotomized rat retinas was examined to determine if it could increase the expression of several glial markers previously demonstrated to have a neuroprotective function in the injured brain and retina. Using Western blotting, the expression of glial fibrillary acid protein (GFAP), glutamate/aspartate transporter-1 (GLAST-1), glutamine synthetase (GS), and connexin 43 (Cx43) was examined 7 days after intravitreal injections of Ad.CNTF or control Ad.LacZ. Compared to controls, intravitreal injection of Ad.CNTF led to significant changes in the expression of CNTFRalpha, pSTAT(3), GFAP, GLAST, GS, and Cx43 in normal and axotomized retinas. Taken together, these results suggest that the neuroprotective effects of CNTF may result from a shift of retinal glia cells to a more neuroprotective phenotype. Moreover, the modulation of astrocytes may buffer high concentrations of glutamate that have been shown to contribute to the death of RGCs after optic nerve transection.
Collapse
Affiliation(s)
- B A van Adel
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, HSC-1R1, McMaster University, Hamilton, Canada
| | | | | | | | | |
Collapse
|
33
|
Zhao Z, Alam S, Oppenheim RW, Prevette DM, Evenson A, Parsadanian A. Overexpression of glial cell line-derived neurotrophic factor in the CNS rescues motoneurons from programmed cell death and promotes their long-term survival following axotomy. Exp Neurol 2004; 190:356-72. [PMID: 15530875 DOI: 10.1016/j.expneurol.2004.06.015] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2004] [Revised: 06/02/2004] [Accepted: 06/07/2004] [Indexed: 10/26/2022]
Abstract
To study the role of one of the most potent motoneuron (MN) survival factors, glial cell line-derived neurotrophic factor (GDNF) derived from the CNS, we generated transgenic animals overexpressing GDNF under the control of an astrocyte-specific GFAP promoter. In situ hybridization revealed that GDNF was expressed at high levels in astrocytes throughout the brain and spinal cord. We analyzed the effects of CNS-derived GDNF on MN survival during the period of programmed cell death (PCD) and after nerve axotomy. In GFAP-GDNF mice at E15, E18, and P1, the survival of brachial MNs was increased on average by 30%, lumbar MNs by 20%, and thoracic MNs at P1 by 33%. GDNF also prevented MN PCD in several cranial motor nuclei. We demonstrated for the first time that the number of MNs in the mouse abducens nucleus was also increased by 40%, thus extending known MN populations that are responsive to GDNF. Next, we tested if GDNF could support complete and relatively long-term survival of MNs following neonatal facial nerve axotomy. We found that virtually all MNs (91%) in GFAP-GDNF mice survived for up to 18 weeks post-axotomy. This is the longest GDNF-mediated survival of neonatal MNs reported following axotomy. Most of surviving MNs were not atrophic, and MN-specific ChAT and neurofilament immunoreactivity (IR) were preserved. Furthermore, GDNF attenuated axotomy-induced astroglial activation. These data demonstrate that overexpression of GDNF in the CNS has very profound effects on MN survival both during the PCD period and after neuronal injury. GFAP-GDNF mice will be valuable to study the effects of CNS-derived GDNF in mouse models of MN degenerative diseases and axonal regeneration in vivo.
Collapse
Affiliation(s)
- Zhongqiu Zhao
- Center for the Study of Nervous System Injury, Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
34
|
Ostrow LW, Sachs F. Mechanosensation and endothelin in astrocytes--hypothetical roles in CNS pathophysiology. ACTA ACUST UNITED AC 2004; 48:488-508. [PMID: 15914254 DOI: 10.1016/j.brainresrev.2004.09.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2003] [Revised: 08/19/2004] [Accepted: 09/09/2004] [Indexed: 01/23/2023]
Abstract
Endothelin (ET) is a potent autocrine mitogen produced by reactive and neoplastic astrocytes. ET has been implicated in the induction of astrocyte proliferation and other transformations engendered by brain pathology, and in promoting the malignant behavior of astrocytomas. Reactive astrocytes containing ET are found in the periphery/penumbra of a wide array of CNS pathologies. Virtually all brain pathology deforms the surrounding parenchyma, either by direct mass effect or edema. Mechanical stress is a well established stimulus for ET production and release by other cell types, but has not been well studied in the brain. However, numerous studies have illustrated that astrocytes can sense mechanical stress and translate it into chemical messages. Furthermore, the ubiquitous reticular meshwork formed by interconnected astrocytes provides an ideal morphology for sensing and responding to mechanical disturbances. We have recently demonstrated stretch-induced ET production by astrocytes in vitro. Inspired by this finding, the purpose of this article is to review the literature on (1) astrocyte mechanosensation, and (2) the endothelin system in astrocytes, and to consider the hypothesis that mechanical induction of the ET system may influence astrocyte functioning in CNS pathophysiology. We conclude by discussing evidence supporting future investigations to determine whether specific inhibition of stretch-activated ion channels may represent a novel strategy for treating or preventing CNS disturbances, as well as the relevance to astrocyte-derived tumors.
Collapse
Affiliation(s)
- Lyle W Ostrow
- Department of Physiology and Biophysics, S.U.N.Y. at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY 14214, USA
| | | |
Collapse
|