1
|
Velasquez E, Savchenko E, Marmolejo-Martínez-Artesero S, Challuau D, Aebi A, Pomeshchik Y, Lamas NJ, Vihinen M, Rezeli M, Schneider B, Raoul C, Roybon L. TNFα prevents FGF4-mediated rescue of astrocyte dysfunction and reactivity in human ALS models. Neurobiol Dis 2024; 201:106687. [PMID: 39362568 DOI: 10.1016/j.nbd.2024.106687] [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: 05/29/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024] Open
Abstract
Astrocytes play a crucial role in the onset and progression of amyotrophic lateral sclerosis (ALS), a fatal disorder marked by the degeneration of motor neurons (MNs) in the central nervous system. Although astrocytes in ALS are known to be toxic to MNs, the pathological changes leading to their neurotoxic phenotype remain poorly understood. In this study, we generated human astrocytes from induced pluripotent stem cells (iPSCs) carrying the ALS-associated A4V mutation in superoxide dismutase 1 (SOD1) to examine early cellular pathways and network changes. Proteomic analysis revealed that ALS astrocytes are both dysfunctional and reactive compared to control astrocytes. We identified significant alterations in the levels of proteins linked to ALS pathology and the innate immune cGAS-STING pathway. Furthermore, we found that ALS astrocyte reactivity differs from that of control astrocytes treated with tumor necrosis factor alpha (TNFα), a key cytokine in inflammatory reactions. We then evaluated the potential of fibroblast growth factor (FGF) 2, 4, 16, and 18 to reverse ALS astrocyte phenotype. Among these, FGF4 successfully reversed ALS astrocyte dysfunction and reactivity in vitro. When delivered to the spinal cord of the SOD1G93A mouse model of ALS, FGF4 lowered astrocyte reactivity. However, this was not sufficient to protect MNs from cell death. Further analysis indicated that TNFα abrogated the reactivity reduction achieved by FGF4, suggesting that complete rescue of the ALS phenotype by FGF4 is hindered by ongoing complex neuroinflammatory processes in vivo. In summary, our data demonstrate that astrocytes generated from ALS iPSCs are inherently dysfunctional and exhibit an immune reactive phenotype. Effectively targeting astrocyte dysfunction and reactivity in vivo may help mitigate ALS and prevent MN death.
Collapse
Affiliation(s)
- Erika Velasquez
- iPSC Laboratory for CNS Disease Modelling, Department of Experimental Medical Science, BMC D10, Lund University, 22184 Lund, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden.
| | - Ekaterina Savchenko
- iPSC Laboratory for CNS Disease Modelling, Department of Experimental Medical Science, BMC D10, Lund University, 22184 Lund, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden.
| | | | | | - Aline Aebi
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland.
| | - Yuriy Pomeshchik
- iPSC Laboratory for CNS Disease Modelling, Department of Experimental Medical Science, BMC D10, Lund University, 22184 Lund, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden.
| | - Nuno Jorge Lamas
- Anatomic Pathology Service, Pathology Department, Centro Hospitalar e Universitário do Porto, Largo Professor Abel Salazar, 4099-001 Porto, Portugal; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, University of Minho, 4710-057 Braga, Portugal.
| | - Mauno Vihinen
- Department of Experimental Medical Science, BMC B13, Lund University, 22184 Lund, Sweden..
| | - Melinda Rezeli
- Department of Biomedical Engineering, Lund University, Lund, Sweden; BioMS - Swedish National Infrastructure for Biological Mass Spectrometry, Lund University, Lund, Sweden.
| | - Bernard Schneider
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland; Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Cedric Raoul
- INM, Univ Montpellier, INSERM, 34091, Montpellier, France.
| | - Laurent Roybon
- iPSC Laboratory for CNS Disease Modelling, Department of Experimental Medical Science, BMC D10, Lund University, 22184 Lund, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden; Department of Neurodegenerative Science, the MiND program, Van Andel Institute, Grand Rapids, 49503, MI, USA.
| |
Collapse
|
2
|
Fujiwara S, Nakano-Doi A, Sawano T, Kubo S, Doe N, Nakagomi T. Administration of Human-Derived Mesenchymal Stem Cells Activates Locally Stimulated Endogenous Neural Progenitors and Reduces Neurological Dysfunction in Mice after Ischemic Stroke. Cells 2024; 13:939. [PMID: 38891071 PMCID: PMC11171641 DOI: 10.3390/cells13110939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Increasing evidence shows that the administration of mesenchymal stem cells (MSCs) is a promising option for various brain diseases, including ischemic stroke. Studies have demonstrated that MSC transplantation after ischemic stroke provides beneficial effects, such as neural regeneration, partially by activating endogenous neural stem/progenitor cells (NSPCs) in conventional neurogenic zones, such as the subventricular and subgranular zones. However, whether MSC transplantation regulates the fate of injury-induced NSPCs (iNSPCs) regionally activated at injured regions after ischemic stroke remains unclear. Therefore, mice were subjected to ischemic stroke, and mCherry-labeled human MSCs (h-MSCs) were transplanted around the injured sites of nestin-GFP transgenic mice. Immunohistochemistry of brain sections revealed that many GFP+ cells were observed around the grafted sites rather than in the regions in the subventricular zone, suggesting that transplanted mCherry+ h-MSCs stimulated GFP+ locally activated endogenous iNSPCs. In support of these findings, coculture studies have shown that h-MSCs promoted the proliferation and neural differentiation of iNSPCs extracted from ischemic areas. Furthermore, pathway analysis and gene ontology analysis using microarray data showed that the expression patterns of various genes related to self-renewal, neural differentiation, and synapse formation were changed in iNSPCs cocultured with h-MSCs. We also transplanted h-MSCs (5.0 × 104 cells/µL) transcranially into post-stroke mouse brains 6 weeks after middle cerebral artery occlusion. Compared with phosphate-buffered saline-injected controls, h-MSC transplantation displayed significantly improved neurological functions. These results suggest that h-MSC transplantation improves neurological function after ischemic stroke in part by regulating the fate of iNSPCs.
Collapse
Affiliation(s)
- Shuichi Fujiwara
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
| | - Toshinori Sawano
- Department of Biomedical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu 525-8577, Japan;
| | - Shuji Kubo
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
| | - Nobutaka Doe
- Department of Rehabilitation, Hyogo Medical University (Kobe Campus), 1-3-6 Minatojima, Chuo-ku, Kobe 650-8530, Japan;
| | - Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo Medical University (Nishinomiya Campus), 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (S.F.); (A.N.-D.); (S.K.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
| |
Collapse
|
3
|
Valenza M, Facchinetti R, Torazza C, Ciarla C, Bronzuoli MR, Balbi M, Bonanno G, Popoli M, Steardo L, Milanese M, Musazzi L, Bonifacino T, Scuderi C. Molecular signatures of astrocytes and microglia maladaptive responses to acute stress are rescued by a single administration of ketamine in a rodent model of PTSD. Transl Psychiatry 2024; 14:209. [PMID: 38796504 PMCID: PMC11127980 DOI: 10.1038/s41398-024-02928-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/28/2024] Open
Abstract
Stress affects the brain and alters its neuroarchitecture and function; these changes can be severe and lead to psychiatric disorders. Recent evidence suggests that astrocytes and microglia play an essential role in the stress response by contributing to the maintenance of cerebral homeostasis. These cells respond rapidly to all stimuli that reach the brain, including stressors. Here, we used a recently validated rodent model of post-traumatic stress disorder in which rats can be categorized as resilient or vulnerable after acute inescapable footshock stress. We then investigated the functional, molecular, and morphological determinants of stress resilience and vulnerability in the prefrontal cortex, focusing on glial and neuronal cells. In addition, we examined the effects of a single subanesthetic dose of ketamine, a fast-acting antidepressant recently approved for the treatment of resistant depression and proposed for other stress-related psychiatric disorders. The present results suggest a prompt glial cell response and activation of the NF-κB pathway after acute stress, leading to an increase in specific cytokines such as IL-18 and TNF-α. This response persists in vulnerable individuals and is accompanied by a significant change in the levels of critical glial proteins such as S100B, CD11b, and CX43, brain trophic factors such as BDNF and FGF2, and proteins related to dendritic arborization and synaptic architecture such as MAP2 and PSD95. Administration of ketamine 24 h after the acute stress event rescued many of the changes observed in vulnerable rats, possibly contributing to support brain homeostasis. Overall, our results suggest that pivotal events, including reactive astrogliosis, changes in brain trophic factors, and neuronal damage are critical determinants of vulnerability to acute traumatic stress and confirm the therapeutic effect of acute ketamine against the development of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Marta Valenza
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Roberta Facchinetti
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Carola Torazza
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Claudia Ciarla
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Maria Rosanna Bronzuoli
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Matilde Balbi
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Maurizio Popoli
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Luca Steardo
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy.
| |
Collapse
|
4
|
Colón Ortiz C, Eroglu C. Astrocyte signaling and interactions in Multiple Sclerosis. Curr Opin Cell Biol 2024; 86:102307. [PMID: 38145604 PMCID: PMC10922437 DOI: 10.1016/j.ceb.2023.102307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/27/2023]
Abstract
Multiple Sclerosis (MS) is a common cause of impairment in working-aged adults. MS is characterized by neuroinflammation and infiltration of peripheral immune cells to the brain, which cause myelin loss and death of oligodendrocytes and neurons. Many studies on MS have focused on the peripheral immune sources of demyelination and repair. However, recent studies revealed that a glial cell type, the astrocytes, undergo robust morphological and transcriptomic changes that contribute significantly to demyelination and myelin repair. Here, we discuss recent findings elucidating signaling modalities that astrocytes acquire or lose in MS and how these changes alter the interactions of astrocytes with other nervous system cell types.
Collapse
Affiliation(s)
- Crystal Colón Ortiz
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC, 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC, 27710, USA; Howard Hughes Medical Institute, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
5
|
Chen J, Stork T, Kang Y, Nardone KAM, Auer F, Farrell RJ, Jay TR, Heo D, Sheehan A, Paton C, Nagel KI, Schoppik D, Monk KR, Freeman MR. Astrocyte growth is driven by the Tre1/S1pr1 phospholipid-binding G protein-coupled receptor. Neuron 2024; 112:93-112.e10. [PMID: 38096817 PMCID: PMC11073822 DOI: 10.1016/j.neuron.2023.11.008] [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: 01/10/2023] [Revised: 07/31/2023] [Accepted: 11/08/2023] [Indexed: 01/06/2024]
Abstract
Astrocytes play crucial roles in regulating neural circuit function by forming a dense network of synapse-associated membrane specializations, but signaling pathways regulating astrocyte morphogenesis remain poorly defined. Here, we show the Drosophila lipid-binding G protein-coupled receptor (GPCR) Tre1 is required for astrocytes to establish their intricate morphology in vivo. The lipid phosphate phosphatases Wunen/Wunen2 also regulate astrocyte morphology and, via Tre1, mediate astrocyte-astrocyte competition for growth-promoting lipids. Loss of s1pr1, the functional analog of Tre1 in zebrafish, disrupts astrocyte process elaboration, and live imaging and pharmacology demonstrate that S1pr1 balances proper astrocyte process extension/retraction dynamics during growth. Loss of Tre1 in flies or S1pr1 in zebrafish results in defects in simple assays of motor behavior. Tre1 and S1pr1 are thus potent evolutionarily conserved regulators of the elaboration of astrocyte morphological complexity and, ultimately, astrocyte control of behavior.
Collapse
Affiliation(s)
- Jiakun Chen
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Tobias Stork
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yunsik Kang
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Katherine A M Nardone
- Departments of Otolaryngology and Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Franziska Auer
- Departments of Otolaryngology and Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ryan J Farrell
- Neuroscience Institute, NYU Medical Center, New York, NY 10016, USA
| | - Taylor R Jay
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Dongeun Heo
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy Sheehan
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Cameron Paton
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - David Schoppik
- Departments of Otolaryngology and Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kelly R Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Marc R Freeman
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
6
|
Manu DR, Slevin M, Barcutean L, Forro T, Boghitoiu T, Balasa R. Astrocyte Involvement in Blood-Brain Barrier Function: A Critical Update Highlighting Novel, Complex, Neurovascular Interactions. Int J Mol Sci 2023; 24:17146. [PMID: 38138976 PMCID: PMC10743219 DOI: 10.3390/ijms242417146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Neurological disorders have been linked to a defective blood-brain barrier (BBB), with dysfunctions triggered by stage-specific disease mechanisms, some of these being generated through interactions in the neurovascular unit (NVU). Advanced knowledge of molecular and signaling mechanisms in the NVU and the emergence of improved experimental models allow BBB permeability prediction and the development of new brain-targeted therapies. As NVU constituents, astrocytes are the most numerous glial cells, characterized by a heterogeneity that occurs as a result of developmental and context-based gene expression profiles and the differential expression of non-coding ribonucleic acids (RNAs). Due to their heterogeneity and dynamic responses to different signals, astrocytes may have a beneficial or detrimental role in the BBB's barrier function, with deep effects on the pathophysiology of (and on the progression of) central nervous system diseases. The implication of astrocytic-derived extracellular vesicles in pathological mechanisms, due to their ability to pass the BBB, must also be considered. The molecular mechanisms of astrocytes' interaction with endothelial cells at the BBB level are considered promising therapeutic targets in different neurological conditions. Nevertheless, a personalized and well-founded approach must be addressed, due to the temporal and spatial heterogeneity of reactive astrogliosis states during disease.
Collapse
Affiliation(s)
- Doina Ramona Manu
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania; (D.R.M.); (M.S.)
| | - Mark Slevin
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania; (D.R.M.); (M.S.)
- Department of Life Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK
| | - Laura Barcutean
- Neurology 1 Clinic, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| | - Timea Forro
- Doctoral School, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| | - Tudor Boghitoiu
- Psychiatry II Clinic, County Clinical Hospital, 540072 Targu Mures, Romania;
| | - Rodica Balasa
- Neurology 1 Clinic, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
- Department of Neurology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| |
Collapse
|
7
|
Cheng YT, Woo J, Luna-Figueroa E, Maleki E, Harmanci AS, Deneen B. Social deprivation induces astrocytic TRPA1-GABA suppression of hippocampal circuits. Neuron 2023; 111:1301-1315.e5. [PMID: 36787749 PMCID: PMC10121837 DOI: 10.1016/j.neuron.2023.01.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 12/13/2022] [Accepted: 01/18/2023] [Indexed: 02/16/2023]
Abstract
Social experience is essential for the development and maintenance of higher-order brain function. Social deprivation results in a host of cognitive deficits, and cellular studies have largely focused on associated neuronal dysregulation; how astrocyte function is impacted by social deprivation is unknown. Here, we show that hippocampal astrocytes from juvenile mice subjected to social isolation exhibit increased Ca2+ activity and global changes in gene expression. We found that the Ca2+ channel TRPA1 is upregulated in astrocytes after social deprivation and astrocyte-specific deletion of TRPA1 reverses the physiological and cognitive deficits associated with social deprivation. Mechanistically, TRPA1 inhibition of hippocampal circuits is mediated by a parallel increase of astrocytic production and release of the inhibitory neurotransmitter GABA after social deprivation. Collectively, our studies reveal how astrocyte function is tuned to social experience and identifies a social-context-specific mechanism by which astrocytic TRPA1 and GABA coordinately suppress hippocampal circuit function.
Collapse
Affiliation(s)
- Yi-Ting Cheng
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Junsung Woo
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Estefania Luna-Figueroa
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ehson Maleki
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Benjamin Deneen
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Markey KM, Saunders JC, Smuts J, von Reyn CR, Garcia ADR. Astrocyte development—More questions than answers. Front Cell Dev Biol 2023; 11:1063843. [PMID: 37051466 PMCID: PMC10083403 DOI: 10.3389/fcell.2023.1063843] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
The past 15–20 years has seen a remarkable shift in our understanding of astrocyte contributions to central nervous system (CNS) function. Astrocytes have emerged from the shadows of neuroscience and are now recognized as key elements in a broad array of CNS functions. Astrocytes comprise a substantial fraction of cells in the human CNS. Nevertheless, fundamental questions surrounding their basic biology remain poorly understood. While recent studies have revealed a diversity of essential roles in CNS function, from synapse formation and function to blood brain barrier maintenance, fundamental mechanisms of astrocyte development, including their expansion, migration, and maturation, remain to be elucidated. The coincident development of astrocytes and synapses highlights the need to better understand astrocyte development and will facilitate novel strategies for addressing neurodevelopmental and neurological dysfunction. In this review, we provide an overview of the current understanding of astrocyte development, focusing primarily on mammalian astrocytes and highlight outstanding questions that remain to be addressed. We also include an overview of Drosophila glial development, emphasizing astrocyte-like glia given their close anatomical and functional association with synapses. Drosophila offer an array of sophisticated molecular genetic tools and they remain a powerful model for elucidating fundamental cellular and molecular mechanisms governing astrocyte development. Understanding the parallels and distinctions between astrocyte development in Drosophila and vertebrates will enable investigators to leverage the strengths of each model system to gain new insights into astrocyte function.
Collapse
Affiliation(s)
- Kathryn M. Markey
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | | | - Jana Smuts
- Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, United States
| | - Catherine R. von Reyn
- Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, United States
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, United States
| | - A. Denise R. Garcia
- Department of Biology, Drexel University, Philadelphia, PA, United States
- Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, United States
- *Correspondence: A. Denise R. Garcia,
| |
Collapse
|
9
|
Smith CJ. Evolutionarily conserved concepts in glial cell biology. Curr Opin Neurobiol 2023; 78:102669. [PMID: 36577179 PMCID: PMC9845142 DOI: 10.1016/j.conb.2022.102669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 12/28/2022]
Abstract
The evolutionary conservation of glial cells has been appreciated since Ramon y Cajal and Del Rio Hortega first described the morphological features of cells in the nervous system. We now appreciate that glial cells have essential roles throughout life in most nervous systems. The field of glial cell biology has grown exponentially in the last ten years. This new wealth of knowledge has been aided by seminal findings in non-mammalian model systems. Ultimately, such concepts help us to understand glia in mammalian nervous systems. Rather than summarizing the field of glial biology, I will first briefly introduce glia in non-mammalian models systems. Then, highlight seminal findings across the glial field that utilized non-mammalian model systems to advance our understanding of the mammalian nervous system. Finally, I will call attention to some recent findings that introduce new questions about glial cell biology that will be investigated for years to come.
Collapse
Affiliation(s)
- Cody J Smith
- Department of Biological Sciences, IN, USA; The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
10
|
Distinct and Dynamic Transcriptome Adaptations of iPSC-Generated Astrocytes after Cytokine Stimulation. Cells 2022; 11:cells11172644. [PMID: 36078052 PMCID: PMC9455058 DOI: 10.3390/cells11172644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Astrocytes (ACs) do not only play a role in normal neurogenesis and brain homeostasis, but also in inflammatory and neurodevelopmental disorders. We studied here the different patterns of inflammatory activation triggered by cytokines in human induced pluripotent stem cell (iPSC)-derived ACs. An optimized differentiation protocol provided non-inflamed ACs. These cells reacted to TNFα with a rapid translocation of NFκB, while AC precursors showed little response. Transcriptome changes were quantified at seven time points (2–72 h) after stimulation with TNFα, IFNγ or TNFα plus IFNγ. TNFα triggered a strong response within 2 h. It peaked from 12–24 h and reverted towards the ground state after 72 h. Activation by IFNγ was also rapid, but the response pattern differed from that of TNFα. For instance, several chemokines up-regulated by TNFα were not affected by IFNγ. Instead, MHC-II-related antigen presentation was drastically enhanced. The combination of the two cytokines led to a stronger and more persistent response. For instance, TRIB3 up-regulation by the combination of TNFα plus IFNγ may have slowed NFκB inactivation. Additionally, highly synergistic regulation was observed for inflammation modifiers, such as CASP4, and for STAT1-controlled genes. The combination of the cytokines also increased oxidative stress markers (e.g., CHAC1), led to phenotypic changes in ACs and triggered markers related to cell death. In summary, these data demonstrate that there is a large bandwidth of pro-inflammatory AC states, and that single markers are not suitable to describe AC activation or their modulation in disease, development and therapy.
Collapse
|
11
|
Somaiya RD, Huebschman NA, Chaunsali L, Sabbagh U, Carrillo GL, Tewari BP, Fox MA. Development of astrocyte morphology and function in mouse visual thalamus. J Comp Neurol 2022; 530:945-962. [PMID: 34636034 PMCID: PMC8957486 DOI: 10.1002/cne.25261] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 11/10/2022]
Abstract
The rodent visual thalamus has served as a powerful model to elucidate the cellular and molecular mechanisms that underlie sensory circuit formation and function. Despite significant advances in our understanding of the role of axon-target interactions and neural activity in orchestrating circuit formation in visual thalamus, the role of non-neuronal cells, such as astrocytes, is less clear. In fact, we know little about the transcriptional identity and development of astrocytes in mouse visual thalamus. To address this gap in knowledge, we studied the expression of canonical astrocyte molecules in visual thalamus using immunostaining, in situ hybridization, and reporter lines. While our data suggests some level of heterogeneity of astrocytes in different nuclei of the visual thalamus, the majority of thalamic astrocytes appeared to be labeled in Aldh1l1-EGFP mice. This led us to use this transgenic line to characterize the neonatal and postnatal development of these cells in visual thalamus. Our data show that not only have the entire cohort of astrocytes migrated into visual thalamus by eye-opening but they also have acquired their adult-like morphology, even while retinogeniculate synapses are still maturing. Furthermore, ultrastructural, immunohistochemical, and functional approaches revealed that by eye-opening, thalamic astrocytes ensheathe retinogeniculate synapses and are capable of efficient uptake of glutamate. Taken together, our results reveal that the morphological, anatomical, and functional development of astrocytes in visual thalamus occurs prior to eye-opening and the emergence of experience-dependent visual activity.
Collapse
Affiliation(s)
- Rachana D. Somaiya
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24016
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016
| | - Natalie A. Huebschman
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016
- Neuroscience Department, Ohio Wesleyan University, Delaware, OH 43015
| | - Lata Chaunsali
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016
- School of Neuroscience Graduate Program, Virginia Tech, Blacksburg, VA 24061
| | - Ubadah Sabbagh
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24016
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016
| | - Gabriela L. Carrillo
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24016
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016
| | - Bhanu P. Tewari
- Neuroscience Department, School of Medicine, University of Virginia, Charlottesville, VA 22903
| | - Michael A. Fox
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24061
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061
- Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016
| |
Collapse
|
12
|
Wang X, Zhang Z, Zhu Z, Liang Z, Zuo X, Ju C, Song Z, Li X, Hu X, Wang Z. Photobiomodulation Promotes Repair Following Spinal Cord Injury by Regulating the Transformation of A1/A2 Reactive Astrocytes. Front Neurosci 2021; 15:768262. [PMID: 34795557 PMCID: PMC8593167 DOI: 10.3389/fnins.2021.768262] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/04/2021] [Indexed: 01/11/2023] Open
Abstract
After spinal cord injury (SCI), reactive astrocytes can be classified into two distinctive phenotypes according to their different functions: neurotoxic (A1) astrocytes and neuroprotective (A2) astrocytes. Our previous studies proved that photobiomodulation (PBM) can promote motor function recovery and improve tissue repair after SCI, but little is known about the underlying mechanism. Therefore, we aimed to investigate whether PBM contributes to repair after SCI by regulating the activation of astrocytes. Male rats subjected to clip-compression SCI were treated with PBM for two consecutive weeks, and the results showed that recovery of motor function was improved, the lesion cavity size was reduced, and the number of neurons retained was increased. We determined the time course of A1/A2 astrocyte activation after SCI by RNA sequencing (RNA-Seq) and verified that PBM inhibited A1 astrocyte activation and promoted A2 astrocyte activation at 7 days postinjury (dpi) and 14 dpi. Subsequently, potential signaling pathways related to A1/A2 astrocyte activation were identified by GO function analysis and KEGG pathway analysis and then studied in animal experiments and preliminarily analyzed in cultured astrocytes. Next, we observed that the expression of basic fibroblast growth factor (bFGF) and transforming growth factor-β (TGF-β) was upregulated by PBM and that both factors contributed to the transformation of A1/A2 astrocytes in a dose-dependent manner. Finally, we found that PBM reduced the neurotoxicity of A1 astrocytes to dorsal root ganglion (DRG) neurons. In conclusion, PBM can promote better recovery after SCI, which may be related to the transformation of A1/A2 reactive astrocytes.
Collapse
Affiliation(s)
- Xuankang Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhihao Zhang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhijie Zhu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhuowen Liang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaoshuang Zuo
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Cheng Ju
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhiwen Song
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Hospital of People's Liberation Army Joint Logistic Support Force, Dalian, China
| | - Xueyu Hu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhe Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
13
|
Amphetamine sensitization alters hippocampal neuronal morphology and memory and learning behaviors. Mol Psychiatry 2021; 26:4784-4794. [PMID: 32555421 DOI: 10.1038/s41380-020-0809-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/22/2022]
Abstract
It is known that continuous abuse of amphetamine (AMPH) results in alterations in neuronal structure and cognitive behaviors related to the reward system. However, the impact of AMPH abuse on the hippocampus remains unknown. The aim of this study was to determine the damage caused by AMPH in the hippocampus in an addiction model. We reproduced the AMPH sensitization model proposed by Robinson et al. in 1997 and performed the novel object recognition test (NORt) to evaluate learning and memory behaviors. After the NORt, we performed Golgi-Cox staining, a stereological cell count, immunohistochemistry to determine the presence of GFAP, CASP3, and MT-III, and evaluated oxidative stress in the hippocampus. We found that AMPH treatment generates impairment in short- and long-term memories and a decrease in neuronal density in the CA1 region of the hippocampus. The morphological test showed an increase in the total dendritic length, but a decrease in the number of mature spines in the CA1 region. GFAP labeling increased in the CA1 region and MT-III increased in the CA1 and CA3 regions. Finally, we found a decrease in Zn concentration in the hippocampus after AMPH treatment. An increase in the dopaminergic tone caused by AMPH sensitization generates oxidative stress, neuronal death, and morphological changes in the hippocampus that affect cognitive behaviors like short- and long-term memories.
Collapse
|
14
|
Klimaschewski L, Claus P. Fibroblast Growth Factor Signalling in the Diseased Nervous System. Mol Neurobiol 2021; 58:3884-3902. [PMID: 33860438 PMCID: PMC8280051 DOI: 10.1007/s12035-021-02367-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factors (FGFs) act as key signalling molecules in brain development, maintenance, and repair. They influence the intricate relationship between myelinating cells and axons as well as the association of astrocytic and microglial processes with neuronal perikarya and synapses. Advances in molecular genetics and imaging techniques have allowed novel insights into FGF signalling in recent years. Conditional mouse mutants have revealed the functional significance of neuronal and glial FGF receptors, not only in tissue protection, axon regeneration, and glial proliferation but also in instant behavioural changes. This review provides a summary of recent findings regarding the role of FGFs and their receptors in the nervous system and in the pathogenesis of major neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Lars Klimaschewski
- Department of Anatomy, Histology and Embryology, Institute of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria.
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
15
|
SRF Is Required for Maintenance of Astrocytes in Non-Reactive State in the Mammalian Brain. eNeuro 2021; 8:ENEURO.0447-19.2020. [PMID: 33441399 PMCID: PMC7877468 DOI: 10.1523/eneuro.0447-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 11/21/2022] Open
Abstract
Astrocytes play several critical roles in the normal functioning of the mammalian brain, including ion homeostasis, synapse formation, and synaptic plasticity. Following injury and infection or in the setting of neurodegeneration, astrocytes become hypertrophic and reactive, a process termed astrogliosis. Although acute reactive gliosis is beneficial in limiting further tissue damage, chronic gliosis becomes detrimental for neuronal recovery and regeneration. Several extracellular factors have been identified that generate reactive astrocytes; however, very little is known about the cell-autonomous transcriptional mechanisms that regulate the maintenance of astrocytes in the normal non-reactive state. Here, we show that conditional deletion of the stimulus-dependent transcription factor, serum response factor (SRF) in astrocytes (SrfGFAPCKO) results in astrogliosis marked by hypertrophic morphology and increased expression of GFAP, vimentin, and nestin. These reactive astrocytes were not restricted to any specific brain region and were seen in both white and gray matter in the entire brain. This astrogliosis persisted throughout adulthood concomitant with microglial activation. Importantly, the Srf mutant mouse brain did not exhibit any cell death or blood brain barrier (BBB) deficits suggesting that apoptosis and leaky BBB are not the causes for the reactive phenotype. The mutant astrocytes expressed more A2 reactive astrocyte marker genes and the SrfGFAPCKO mice exhibited normal neuronal numbers indicating that SRF-deficient gliosis astrocytes are not neurotoxic. Together, our findings suggest that SRF plays a critical role in astrocytes to maintain them in a non-reactive state.
Collapse
|
16
|
Tatomir A, Beltrand A, Nguyen V, Boodhoo D, Mekala A, Cudrici C, Badea TC, Muresanu DF, Rus V, Rus H. RGC-32 Regulates Generation of Reactive Astrocytes in Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 11:608294. [PMID: 33569054 PMCID: PMC7868332 DOI: 10.3389/fimmu.2020.608294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022] Open
Abstract
Astrocytes are increasingly recognized as critical contributors to multiple sclerosis pathogenesis. We have previously shown that lack of Response Gene to Complement 32 (RGC-32) alters astrocyte morphology in the spinal cord at the peak of experimental autoimmune encephalomyelitis (EAE), suggesting a role for RGC-32 in astrocyte differentiation. In this study, we analyzed the expression and distribution of astrocytes and astrocyte progenitors by immunohistochemistry in spinal cords of wild-type (WT) and RGC-32-knockout (KO) mice with EAE and of normal adult mice. Our analysis showed that during acute EAE, WT astrocytes had a reactive morphology and increased GFAP expression, whereas RGC-32 KO astrocytes had a morphology similar to that of radial glia and an increased expression of progenitor markers such as vimentin and fatty acid binding protein 7 (FABP7). In control mice, GFAP expression and astrocyte density were also significantly higher in the WT group, whereas the number of vimentin and FABP7-positive radial glia was significantly higher in the RGC-32 KO group. In vitro studies on cultured neonatal astrocytes from WT and RGC-32 KO mice showed that RGC-32 regulates a complex array of molecular networks pertaining to signal transduction, growth factor expression and secretion, and extracellular matrix (ECM) remodeling. Among the most differentially expressed factors were insulin-like growth factor 1 (IGF1), insulin-like growth factor binding proteins (IGFBPs), and connective tissue growth factor (CTGF); their expression was downregulated in RGC-32-depleted astrocytes. The nuclear translocation of STAT3, a transcription factor critical for astrogliogenesis and driving glial scar formation, was also impaired after RGC-32 silencing. Taken together, these data suggest that RGC-32 is an important regulator of astrocyte differentiation during EAE and that in the absence of RGC-32, astrocytes are unable to fully mature and become reactive astrocytes.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Cell Differentiation
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fatty Acid-Binding Protein 7/metabolism
- Female
- Glial Fibrillary Acidic Protein/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Rats, Sprague-Dawley
- Signal Transduction
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Vimentin/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Austin Beltrand
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Armugam Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Cornelia Cudrici
- Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tudor C. Badea
- Retinal Circuit Development and Genetics Unit, Neurobiology Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, Bethesda, MD, United States
| | - Dafin F. Muresanu
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, United States
| |
Collapse
|
17
|
Gottipati MK, D'Amato AR, Ziemba AM, Popovich PG, Gilbert RJ. TGFβ3 is neuroprotective and alleviates the neurotoxic response induced by aligned poly-l-lactic acid fibers on naïve and activated primary astrocytes. Acta Biomater 2020; 117:273-282. [PMID: 33035696 DOI: 10.1016/j.actbio.2020.09.057] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022]
Abstract
Following spinal cord injury, astrocytes at the site of injury become reactive and exhibit a neurotoxic (A1) phenotype, which leads to neuronal death. In addition, the glial scar, which is composed of reactive astrocytes, acts as a chemical and physical barrier to subsequent axonal regeneration. Biomaterials, specifically electrospun fibers, induce a migratory phenotype of astrocytes and promote regeneration of axons following acute spinal cord injury in preclinical models. However, no study has examined the potential of electrospun fibers or biomaterials in general to modulate neurotoxic (A1) or neuroprotective (A2) astrocytic phenotypes. To assess astrocyte reactivity in response to aligned poly-l-lactic acid microfibers, naïve spinal cord astrocytes or spinal cord astrocytes primed towards the neurotoxic phenotype (A1) were cultured on fibrous scaffolds. Gene expression analysis of the pan-reactive astrocyte makers (GFAP, Lcn2, SerpinA3), A1 specific markers (H2-D1, SerpinG1), and A2 specific makers (Emp1, S100a10) was done using quantitative polymerase chain reaction (qPCR). Electrospun fibers mildly increased the expression of the pan-reactive and A1-specific markers, showing the ability of fibrous materials to induce a more reactive, A1 phenotype. However, when naïve or activated astrocytes were cultured on fibers in the presence of transforming growth factor β3 (TGFβ3), the expression of A1-specific markers was greatly reduced, which in turn improved neuronal survival in culture.
Collapse
|
18
|
Abbink MR, Kotah JM, Hoeijmakers L, Mak A, Yvon-Durocher G, van der Gaag B, Lucassen PJ, Korosi A. Characterization of astrocytes throughout life in wildtype and APP/PS1 mice after early-life stress exposure. J Neuroinflammation 2020; 17:91. [PMID: 32197653 PMCID: PMC7083036 DOI: 10.1186/s12974-020-01762-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Early-life stress (ES) is an emerging risk factor for later life development of Alzheimer’s disease (AD). We have previously shown that ES modulates amyloid-beta pathology and the microglial response to it in the APPswe/PS1dE9 mouse model. Because astrocytes are key players in the pathogenesis of AD, we studied here if and how ES affects astrocytes in wildtype (WT) and APP/PS1 mice and how these relate to the previously reported amyloid pathology and microglial profile. Methods We induced ES by limiting nesting and bedding material from postnatal days (P) 2–9. We studied in WT mice (at P9, P30, and 6 months) and in APP/PS1 mice (at 4 and 10 months) (i) GFAP coverage, cell density, and complexity in hippocampus (HPC) and entorhinal cortex (EC); (ii) hippocampal gene expression of astrocyte markers; and (iii) the relationship between astrocyte, microglia, and amyloid markers. Results In WT mice, ES increased GFAP coverage in HPC subregions at P9 and decreased it at 10 months. APP/PS1 mice at 10 months exhibited both individual cell as well as clustered GFAP signals. APP/PS1 mice when compared to WT exhibited reduced total GFAP coverage in HPC, which is increased in the EC, while coverage of the clustered GFAP signal in the HPC was increased and accompanied by increased expression of several astrocytic genes. While measured astrocytic parameters in APP/PS1 mice appear not be further modulated by ES, analyzing these in the context of ES-induced alterations to amyloid pathology and microglial shows alterations at both 4 and 10 months of age. Conclusions Our data suggest that ES leads to alterations to the astrocytic response to amyloid-β pathology.
Collapse
Affiliation(s)
- Maralinde R Abbink
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Janssen M Kotah
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Aline Mak
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Genevieve Yvon-Durocher
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Bram van der Gaag
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Malik VA, Zajicek F, Mittmann LA, Klaus J, Unterseer S, Rajkumar S, Pütz B, Deussing JM, Neumann ID, Rupprecht R, Di Benedetto B. GDF15 promotes simultaneous astrocyte remodeling and tight junction strengthening at the blood-brain barrier. J Neurosci Res 2020; 98:1433-1456. [PMID: 32170776 DOI: 10.1002/jnr.24611] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/24/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022]
Abstract
Perivascular astrocyte processes (PAP) surround cerebral endothelial cells (ECs) and modulate the strengthening of tight junctions to influence blood-brain barrier (BBB) permeability. Morphologically altered astrocytes may affect barrier properties and trigger the onset of brain pathologies. However, astrocyte-dependent mediators of these events remain poorly studied. Here, we show a pharmacologically driven elevated expression and release of growth/differentiation factor 15 (GDF15) in rat primary astrocytes and cerebral PAP. GDF15 has been shown to possess trophic properties for motor neurons, prompting us to hypothesize similar effects on astrocytes. Indeed, its increased expression and release occurred simultaneously to morphological changes of astrocytes in vitro and PAP, suggesting modulatory effects of GDF15 on these cells, but also neighboring EC. Administration of recombinant GDF15 was sufficient to promote astrocyte remodeling and enhance barrier properties between ECs in vitro, whereas its pharmacogenetic abrogation prevented these effects. We validated our findings in male high anxiety-related behavior rats, an animal model of depressive-like behavior, with shrunk PAP associated with reduced expression of the junctional protein claudin-5, which were both restored by a pharmacologically induced increase in GDF15 expression. Thus, we identified GDF15 as an astrocyte-derived trigger of astrocyte process remodeling linked to enhanced tight junction strengthening at the BBB.
Collapse
Affiliation(s)
- Victoria A Malik
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Franziska Zajicek
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Laura A Mittmann
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | | | | | - Sandeep Rajkumar
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Inga D Neumann
- Department of Neurobiology and Animal Physiology, University of Regensburg, Regensburg, Germany.,Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.,Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Barbara Di Benedetto
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.,Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| |
Collapse
|
20
|
Trindade P, Loiola EC, Gasparotto J, Ribeiro CT, Cardozo PL, Devalle S, Salerno JA, Ornelas IM, Ledur PF, Ribeiro FM, Ventura ALM, Moreira JCF, Gelain DP, Porciúncula LO, Rehen SK. Short and long TNF‐alpha exposure recapitulates canonical astrogliosis events in human‐induced pluripotent stem cells‐derived astrocytes. Glia 2020; 68:1396-1409. [DOI: 10.1002/glia.23786] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 12/17/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Pablo Trindade
- Instituto D'Or de Pesquisa e Ensino (IDOR) Rio de Janeiro Brazil
- Pós‐Graduação em Biologia Molecular e CelularUniversidade Federal do Estado do Rio de Janeiro Rio de Janeiro Brazil
| | | | - Juciano Gasparotto
- Departamento de BioquímicaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Camila Tiefensee Ribeiro
- Departamento de BioquímicaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Pablo Leal Cardozo
- Departamento de Bioquímica e ImunologiaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Sylvie Devalle
- Instituto D'Or de Pesquisa e Ensino (IDOR) Rio de Janeiro Brazil
| | | | | | | | - Fabiola Mara Ribeiro
- Departamento de Bioquímica e ImunologiaUniversidade Federal de Minas Gerais Belo Horizonte Brazil
| | | | - José Claudio Fonseca Moreira
- Departamento de BioquímicaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Daniel Pens Gelain
- Departamento de BioquímicaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Lisiane Oliveira Porciúncula
- Departamento de BioquímicaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Stevens Kastrup Rehen
- Instituto D'Or de Pesquisa e Ensino (IDOR) Rio de Janeiro Brazil
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| |
Collapse
|
21
|
Hensel N, Raker V, Förthmann B, Detering NT, Kubinski S, Buch A, Katzilieris-Petras G, Spanier J, Gudi V, Wagenknecht S, Kopfnagel V, Werfel TA, Stangel M, Beineke A, Kalinke U, Paludan SR, Sodeik B, Claus P. HSV-1 triggers paracrine fibroblast growth factor response from cortical brain cells via immediate-early protein ICP0. J Neuroinflammation 2019; 16:248. [PMID: 31791351 PMCID: PMC6889453 DOI: 10.1186/s12974-019-1647-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/19/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Herpes simplex virus-1 (HSV-1) infections of the central nervous system (CNS) can result in HSV-1 encephalitis (HSE) which is characterized by severe brain damage and long-term disabilities. Different cell types including neurons and astrocytes become infected in the course of an HSE which leads to an activation of glial cells. Activated glial cells change their neurotrophic factor profile and modulate inflammation and repair. The superfamily of fibroblast growth factors (FGFs) is one of the largest family of neurotrophic factors comprising 22 ligands. FGFs induce pro-survival signaling in neurons and an anti-inflammatory answer in glial cells thereby providing a coordinated tissue response which favors repair over inflammation. Here, we hypothesize that FGF expression is altered in HSV-1-infected CNS cells. METHOD We employed primary murine cortical cultures comprising a mixed cell population of astrocytes, neurons, microglia, and oligodendrocytes. Astrocyte reactivity was morphometrically monitored by an automated image analysis algorithm as well as by analyses of A1/A2 marker expression. Altered FGF expression was detected by quantitative real-time PCR and its paracrine FGF activity. In addition, HSV-1 mutants were employed to characterize viral factors important for FGF responses of infected host cells. RESULTS Astrocytes in HSV-1-infected cortical cultures were transiently activated and became hypertrophic and expressed both A1- and A2-markers. Consistently, a number of FGFs were transiently upregulated inducing paracrine neurotrophic signaling in neighboring cells. Most prominently, FGF-4, FGF-8, FGF-9, and FGF-15 became upregulated in a switch-on like mechanism. This effect was specific for CNS cells and for a fully functional HSV-1. Moreover, the viral protein ICP0 critically mediated the FGF switch-on mechanism. CONCLUSIONS HSV-1 uses the viral protein ICP0 for the induction of FGF-expression in CNS cells. Thus, we propose that HSV-1 triggers FGF activity in the CNS for a modulation of tissue response upon infection.
Collapse
Affiliation(s)
- Niko Hensel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Verena Raker
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Benjamin Förthmann
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Nora Tula Detering
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Sabrina Kubinski
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Anna Buch
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig, Germany
| | | | - Julia Spanier
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Sylvia Wagenknecht
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hanover, Germany
| | - Verena Kopfnagel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hanover, Germany
| | - Thomas Andreas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hanover, Germany
| | - Martin Stangel
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Andreas Beineke
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ulrich Kalinke
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Søren Riis Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Beate Sodeik
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig, Germany
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
22
|
Ali AAH, Schwarz-Herzke B, Rollenhagen A, Anstötz M, Holub M, Lübke J, Rose CR, Schnittler HJ, von Gall C. Bmal1-deficiency affects glial synaptic coverage of the hippocampal mossy fiber synapse and the actin cytoskeleton in astrocytes. Glia 2019; 68:947-962. [PMID: 31743496 DOI: 10.1002/glia.23754] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 01/13/2023]
Abstract
Bmal1 is an essential component of the molecular clockwork, which drives circadian rhythms in cell function. In Bmal1-deficient (Bmal1-/-) mice, chronodisruption is associated with cognitive deficits and progressive brain pathology including astrocytosis indicated by increased expression of glial fibrillary acidic protein (GFAP). However, relatively little is known about the impact of Bmal1-deficiency on astrocyte morphology prior to astrocytosis. Therefore, in this study we analysed astrocyte morphology in young (6-8 weeks old) adult Bmal1-/- mice. At this age, overall GFAP immunoreactivity was not increased in Bmal1-deficient mice. At the ultrastructural level, we found a decrease in the volume fraction of the fine astrocytic processes that cover the hippocampal mossy fiber synapse, suggesting an impairment of perisynaptic processes and their contribution to neurotransmission. For further analyses of actin cytoskeleton, which is essential for distal process formation, we used cultured Bmal1-/- astrocytes. Bmal1-/- astrocytes showed an impaired formation of actin stress fibers. Moreover, Bmal1-/- astrocytes showed reduced levels of the actin-binding protein cortactin (CTTN). Cttn promoter region contains an E-Box like element and chromatin immunoprecipitation revealed that Cttn is a potential Bmal1 target gene. In addition, the level of GTP-bound (active) Rho-GTPase (Rho-GTP) was reduced in Bmal1-/- astrocytes. In summary, our data demonstrate that Bmal1-deficiency affects morphology of the fine astrocyte processes prior to strong upregulation of GFAP, presumably because of impaired Cttn expression and reduced Rho-GTP activation. These morphological changes might result in altered synaptic function and, thereby, relate to cognitive deficits in chronodisruption.
Collapse
Affiliation(s)
- Amira A H Ali
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Beryl Schwarz-Herzke
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Max Anstötz
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Martin Holub
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Joachim Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty/RWTH University Hospital Aachen, Aachen, Germany.,Institute of Neuroscience and Medicine INM-10, JARA Translational Brain Medicine, Aachen, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University, Düsseldorf, Germany
| | - Hans-Joachim Schnittler
- Institute of Anatomy and Vascular Biology, Medical Faculty, Westfälische Wilhelms University, Münster, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
23
|
Hultman K, Scarlett JM, Baquero AF, Cornea A, Zhang Y, Salinas CBG, Brown J, Morton GJ, Whalen EJ, Grove KL, Koegler FH, Schwartz MW, Mercer AJ. The central fibroblast growth factor receptor/beta klotho system: Comprehensive mapping in Mus musculus and comparisons to nonhuman primate and human samples using an automated in situ hybridization platform. J Comp Neurol 2019; 527:2069-2085. [PMID: 30809795 DOI: 10.1002/cne.24668] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/25/2022]
Abstract
Central activation of fibroblast growth factor (FGF) receptors regulates peripheral glucose homeostasis and reduces food intake in preclinical models of obesity and diabetes. The current work was undertaken to advance our understanding of the receptor expression, as sites of ligand action by FGF19, FGF21, and FGF1 in the mammalian brain remains unresolved. Recent advances in automated RNAscope in situ hybridization and droplet digital PCR (ddPCR) technology allowed us to interrogate central FGFR/beta klotho (Klb) system at the cellular level in the mouse, with relevant comparisons to nonhuman primate and human brain. FGFR1-3 gene expression was broadly distributed throughout the CNS in Mus musculus, with FGFR1 exhibiting the greatest heterogeneity. FGFR4 expression localized only in the medial habenula and subcommissural organ of mice. Likewise, Klb mRNA was restricted to the suprachiasmatic nucleus (SCh) and select midbrain and hindbrain nuclei. ddPCR in the rodent hypothalamus confirmed that, although expression levels are indeed low for Klb, there is nonetheless a bonafide subpopulation of Klb+ cells in the hypothalamus. In NHP and human midbrain and hindbrain, Klb + cells are quite rare, as is expression of FGFR4. Collectively, these data provide the most robust central map of the FGFR/Klb system to date and highlight central regions that may be of critical importance to assess central ligand effects with pharmacological dosing, such as the putative interactions between the endocrine FGFs and FGFR1/Klb, or FGF19 with FGFR4.
Collapse
Affiliation(s)
| | - Jarrad M Scarlett
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington.,Department of Pediatric Gastroenterology & Hepatology, Seattle Children's Hospital, Seattle, Washington
| | - Arian F Baquero
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Anda Cornea
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Yu Zhang
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | | | - Jenny Brown
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Gregory J Morton
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Erin J Whalen
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Kevin L Grove
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Frank H Koegler
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Michael W Schwartz
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Aaron J Mercer
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| |
Collapse
|
24
|
Arroyo-García LE, Vázquez-Roque RA, Díaz A, Treviño S, De La Cruz F, Flores G, Rodríguez-Moreno A. The Effects of Non-selective Dopamine Receptor Activation by Apomorphine in the Mouse Hippocampus. Mol Neurobiol 2018; 55:8625-8636. [PMID: 29582396 DOI: 10.1007/s12035-018-0991-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
Abstract
Apomorphine is a dopamine receptor agonist that activates D1-D5 dopamine receptors and that is used to treat Parkinson's disease (PD). However, the effect of apomorphine on non-motor activity has been poorly studied, and likewise, the effects of dopaminergic activation in brain areas that do not fulfill motor functions are unclear. The aim of this study was to determine how dopamine receptor activation affects behavior, as well as plasticity, morphology, and oxidative stress in the hippocampus. Adult mice were chronically administered apomorphine (1 mg/kg for 15 days), and the effects on memory and learning, synaptic plasticity, dendritic length, inflammatory responses, and oxidative stress were evaluated. Apomorphine impaired learning and long-term memory in mice, as evaluated in the Morris water maze test. In addition, electrophysiological recording of field excitatory postsynaptic potentials (fEPSP) indicated that the long-term potentiation (LTP) of synaptic transmission in the CA1 region of the hippocampus was fully impaired by apomorphine. In addition, a Sholl analysis of Golgi-Cox stained neurons showed that apomorphine reduced the total length of dendrites in the CA1 region of the hippocampus. Finally, there were more reactive astrocytes and oxidative stress biomarkers in mice administered apomorphine, as measured by GFAP immunohistochemistry and markers of redox balance, respectively. Hence, the non-selective activation of dopaminergic receptors in the hippocampus by apomorphine triggers deficiencies in learning and memory, it prevents LTP, reduces dendritic length, and provokes neuronal damage.
Collapse
Affiliation(s)
- Luis Enrique Arroyo-García
- Laboratorio de Neurociencia Celular y Plasticidad, Universidad Pablo de Olavide, Sevilla, Spain
- Laboratorio de Fisiología de la Conducta, Instituto Politécnico Nacional, Mexico City, Mexico
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, 72570, Puebla, CP, Mexico
| | - Rubén Antonio Vázquez-Roque
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, 72570, Puebla, CP, Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Fidel De La Cruz
- Laboratorio de Fisiología de la Conducta, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, 72570, Puebla, CP, Mexico.
| | - Antonio Rodríguez-Moreno
- Laboratorio de Neurociencia Celular y Plasticidad, Universidad Pablo de Olavide, Sevilla, Spain.
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, 41013, Sevilla, Spain.
| |
Collapse
|
25
|
Neal M, Luo J, Harischandra DS, Gordon R, Sarkar S, Jin H, Anantharam V, Désaubry L, Kanthasamy A, Kanthasamy A. Prokineticin-2 promotes chemotaxis and alternative A2 reactivity of astrocytes. Glia 2018; 66:2137-2157. [PMID: 30277602 DOI: 10.1002/glia.23467] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/27/2022]
Abstract
Astrocyte reactivity is disease- and stimulus-dependent, adopting either a proinflammatory A1 phenotype or a protective, anti-inflammatory A2 phenotype. Recently, we demonstrated, using cell culture, animal models and human brain samples, that dopaminergic neurons produce and secrete higher levels of the chemokine-like signaling protein Prokineticin-2 (PK2) as a compensatory protective response against neurotoxic stress. As astrocytes express a high level of PK2 receptors, herein, we systematically characterize the role of PK2 in astrocyte structural and functional properties. PK2 treatment greatly induced astrocyte migration, which was accompanied by a shift in mitochondrial energy metabolism, a reduction in proinflammatory factors, and an increase in the antioxidant genes Arginase-1 and Nrf2. Overexpression of PK2 in primary astrocytes or in the in vivo mouse brain induced the A2 astrocytic phenotype with upregulation of key protective genes and A2 reactivity markers including Arginase-1 and Nrf2, PTX3, SPHK1, and TM4SF1. A small-molecule PK2 agonist, IS20, not only mimicked the protective effect of PK2 in primary cultures, but also increased glutamate uptake by upregulating GLAST. Notably, IS20 blocked not only MPTP-induced reductions in the A2 phenotypic markers SPHK1 and SCL10a6 but also elevation of the of A1 marker GBP2. Collectively, our results reveal that PK2 regulates a novel neuron-astrocyte signaling mechanism by promoting an alternative A2 protective phenotype in astrocytes, which could be exploited for development of novel therapeutic strategies for PD and other related chronic neurodegenerative diseases. PK2 signals through its receptors on astrocytes and promotes directed chemotaxis. PK2-induced astrocyte reactivity leads to an increase in antioxidant and anti-inflammatory proteins while increasing glutamate uptake, along with decreased inflammatory factors. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Matthew Neal
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Jie Luo
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Dilshan S Harischandra
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Richard Gordon
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Souvarish Sarkar
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Laurent Désaubry
- Therapeutic Innovation Laboratory (UMR7200), CNRS-University of Strasbourg, Illkirch, France
| | - Anumantha Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011
| |
Collapse
|
26
|
A deep convolutional neural network approach for astrocyte detection. Sci Rep 2018; 8:12878. [PMID: 30150631 PMCID: PMC6110828 DOI: 10.1038/s41598-018-31284-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 08/10/2018] [Indexed: 12/26/2022] Open
Abstract
Astrocytes are involved in various brain pathologies including trauma, stroke, neurodegenerative disorders such as Alzheimer’s and Parkinson’s diseases, or chronic pain. Determining cell density in a complex tissue environment in microscopy images and elucidating the temporal characteristics of morphological and biochemical changes is essential to understand the role of astrocytes in physiological and pathological conditions. Nowadays, manual stereological cell counting or semi-automatic segmentation techniques are widely used for the quantitative analysis of microscopy images. Detecting astrocytes automatically is a highly challenging computational task, for which we currently lack efficient image analysis tools. We have developed a fast and fully automated software that assesses the number of astrocytes using Deep Convolutional Neural Networks (DCNN). The method highly outperforms state-of-the-art image analysis and machine learning methods and provides precision comparable to those of human experts. Additionally, the runtime of cell detection is significantly less than that of other three computational methods analysed, and it is faster than human observers by orders of magnitude. We applied our DCNN-based method to examine the number of astrocytes in different brain regions of rats with opioid-induced hyperalgesia/tolerance (OIH/OIT), as morphine tolerance is believed to activate glia. We have demonstrated a strong positive correlation between manual and DCNN-based quantification of astrocytes in rat brain.
Collapse
|
27
|
Liu CY, Yang Y, Ju WN, Wang X, Zhang HL. Emerging Roles of Astrocytes in Neuro-Vascular Unit and the Tripartite Synapse With Emphasis on Reactive Gliosis in the Context of Alzheimer's Disease. Front Cell Neurosci 2018; 12:193. [PMID: 30042661 PMCID: PMC6048287 DOI: 10.3389/fncel.2018.00193] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 06/14/2018] [Indexed: 01/09/2023] Open
Abstract
Astrocytes, which are five-fold more numerous than neurons in the central nervous system (CNS), are traditionally viewed to provide simple structural and nutritional supports for neurons and to participate in the composition of the blood brain barrier (BBB). In recent years, the active roles of astrocytes in regulating cerebral blood flow (CBF) and in maintaining the homeostasis of the tripartite synapse have attracted increasing attention. More importantly, astrocytes have been associated with the pathogenesis of Alzheimer's disease (AD), a major cause of dementia in the elderly. Although microglia-induced inflammation is considered important in the development and progression of AD, inflammation attributable to astrogliosis may also play crucial roles. A1 reactive astrocytes induced by inflammatory stimuli might be harmful by up-regulating several classical complement cascade genes thereby leading to chronic inflammation, while A2 induced by ischemia might be protective by up-regulating several neurotrophic factors. Here we provide a concise review of the emerging roles of astrocytes in the homeostasis maintenance of the neuro-vascular unit (NVU) and the tripartite synapse with emphasis on reactive astrogliosis in the context of AD, so as to pave the way for further research in this area, and to search for potential therapeutic targets of AD.
Collapse
Affiliation(s)
- Cai-Yun Liu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Wei-Na Ju
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hong-Liang Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
- Department of Life Sciences, The National Natural Science Foundation of China, Beijing, China
| |
Collapse
|
28
|
Inhibition of miR-21 ameliorates excessive astrocyte activation and promotes axon regeneration following optic nerve crush. Neuropharmacology 2018; 137:33-49. [PMID: 29709341 DOI: 10.1016/j.neuropharm.2018.04.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/18/2018] [Accepted: 04/26/2018] [Indexed: 12/15/2022]
Abstract
Optic nerve injury is a leading cause of irreversible visual impairment worldwide and can even cause blindness. Excessive activation of astrocytes has negative effects on the repair and recovery of retinal ganglion cells following optic nerve injury. However, the molecular and cellular mechanisms underlying astrocyte activation after optic nerve injury remain largely unknown. In the present study, we explored the effects of microRNA-21 (miR-21) on axon regeneration and flash visual evoked potential (F-VEP) and the underlying mechanisms of these effects based on astrocyte activation in the rat model of optic nerve crush (ONC). To the best of our knowledge, this article is the first to report that inhibition of miR-21 enhances axonal regeneration and promotes functional recovery in F-VEP in the rat model of ONC. Furthermore, inhibition of miR-21 attenuates excessive astrocyte activation and glial scar formation, thereby promoting axonal regeneration by regulating the epidermal growth factor receptor (EGFR) pathway. In addition, we observed that the expression of tissue inhibitor of metalloproteinase-3, a target gene of miR-21, was inhibited during this process. Taken together, these findings demonstrate that inhibition of miR-21 regulates the EGFR pathway, ameliorating excessive astrocyte activation and glial scar progression and promoting axonal regeneration and alleviating impairment in F-VEP function in a model of ONC. This study's results suggest that miR-21 may represent a therapeutic target for optic nerve injury.
Collapse
|
29
|
Fex Svenningsen Å, Löring S, Sørensen AL, Huynh HUB, Hjæresen S, Martin N, Moeller JB, Elkjær ML, Holmskov U, Illes Z, Andersson M, Nielsen SB, Benedikz E. Macrophage migration inhibitory factor (MIF) modulates trophic signaling through interaction with serine protease HTRA1. Cell Mol Life Sci 2017; 74:4561-4572. [PMID: 28726057 PMCID: PMC5663815 DOI: 10.1007/s00018-017-2592-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/05/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor (MIF), a small conserved protein, is abundant in the immune- and central nervous system (CNS). MIF has several receptors and binding partners that can modulate its action on a cellular level. It is upregulated in neurodegenerative diseases and cancer although its function is far from clear. Here, we report the finding of a new binding partner to MIF, the serine protease HTRA1. This enzyme cleaves several growth factors, extracellular matrix molecules and is implicated in some of the same diseases as MIF. We show that the function of the binding between MIF and HTRA1 is to inhibit the proteolytic activity of HTRA1, modulating the availability of molecules that can change cell growth and differentiation. MIF is therefore the first endogenous inhibitor ever found for HTRA1. It was found that both molecules were present in astrocytes and that the functional binding has the ability to modulate astrocytic activities important in development and disease of the CNS.
Collapse
Affiliation(s)
- Åsa Fex Svenningsen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark.
| | - Svenja Löring
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands
| | - Anna Lahn Sørensen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Ha Uyen Buu Huynh
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Simone Hjæresen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Nellie Martin
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Jesper Bonnet Moeller
- Department of Molecular Medicine-Cancer and Inflammation, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Weill Cornell Medicine, Cornell University, 413 East 69th Street, New York, 10021, USA
| | - Maria Louise Elkjær
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Uffe Holmskov
- Department of Molecular Medicine-Cancer and Inflammation, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Malin Andersson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 59, 751 24, Uppsala, Sweden
| | - Solveig Beck Nielsen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Eirikur Benedikz
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Faculty of Health, University College Zealand, Parkvej 190, 4700, Næstved, Denmark
| |
Collapse
|
30
|
Bielle F, Di Stefano AL, Meyronet D, Picca A, Villa C, Bernier M, Schmitt Y, Giry M, Rousseau A, Figarella-Branger D, Maurage CA, Uro-Coste E, Lasorella A, Iavarone A, Sanson M, Mokhtari K. Diffuse gliomas with FGFR3-TACC3 fusion have characteristic histopathological and molecular features. Brain Pathol 2017; 28:674-683. [PMID: 28976058 DOI: 10.1111/bpa.12563] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/31/2017] [Accepted: 09/24/2017] [Indexed: 12/30/2022] Open
Abstract
Adult glioblastomas, IDH-wildtype represent a heterogeneous group of diseases. They are resistant to conventional treatment by concomitant radiochemotherapy and carry a dismal prognosis. The discovery of oncogenic gene fusions in these tumors has led to prospective targeted treatments, but identification of these rare alterations in practice is challenging. Here, we report a series of 30 adult diffuse gliomas with an in frame FGFR3-TACC3 oncogenic fusion (n = 27 WHO grade IV and n = 3 WHO grade II) as well as their histological and molecular features. We observed recurrent morphological features (monomorphous ovoid nuclei, nuclear palisading and thin parallel cytoplasmic processes, endocrinoid network of thin capillaries) associated with frequent microcalcifications and desmoplasia. We report a constant immunoreactivity for FGFR3, which is a valuable method for screening for the FGFR3-TACC3 fusion with 100% sensitivity and 92% specificity. We confirmed the associated molecular features (typical genetic alterations of glioblastoma, except the absence of EGFR amplification, and an increased frequency of CDK4 and MDM2 amplifications). FGFR3 immunopositivity is a valuable tool to identify gliomas that are likely to harbor the FGFR3-TACC3 fusion for inclusion in targeted therapeutic trials.
Collapse
Affiliation(s)
- Franck Bielle
- AP-HP, Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, Service de Neuropathologie Raymond Escourolle, Paris, France.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Paris, France.,Brain and Spine Institute, Paris, France
| | - Anna-Luisa Di Stefano
- Brain and Spine Institute, Paris, France.,AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France.,Foch Hospital, Service de Neurologie, Suresnes, France
| | - David Meyronet
- Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France
| | - Alberto Picca
- AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - Chiara Villa
- Department of Pathological Cytology and Anatomy, Foch Hospital, Suresnes, France.,INSERM U1016, CNRS UMR8104, Paris Descartes University, Cochin Institute, Paris, France
| | - Michèle Bernier
- Department of Pathological Cytology and Anatomy, Foch Hospital, Suresnes, France
| | | | | | - Audrey Rousseau
- Département de Pathologie Cellulaire et Tissulaire, CHU d'Angers, Angers, France.,CRCINA, INSERM, Université d'Angers, Angers, France
| | - Dominique Figarella-Branger
- AP-HM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France.,Aix-Marseille Univ, INSERM, CRO2 UMR_S 911, Marseille, France
| | | | - Emmanuelle Uro-Coste
- CHU Toulouse, Service d'Anatomie et Cytologie Pathologiques, Institut Universitaire du Cancer-Oncopole, Toulouse, France.,INSERM UMR 1037 (INSERM/Université Toulouse III-Paul Sabatier/ERL CNRS 5294), Centre de Recherche en Cancérologie de Toulouse, Toulouse, France
| | - Anna Lasorella
- Departments of Neurology and Pathology, Institute for Cancer Genetics, Irving Comprehensive Research Center, New York
| | - Antonio Iavarone
- Departments of Neurology and Pathology, Institute for Cancer Genetics, Irving Comprehensive Research Center, New York
| | - Marc Sanson
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Paris, France.,Brain and Spine Institute, Paris, France.,AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France.,OncoNeuroTek, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Karima Mokhtari
- AP-HP, Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, Service de Neuropathologie Raymond Escourolle, Paris, France.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Paris, France.,Brain and Spine Institute, Paris, France.,OncoNeuroTek, Institut du Cerveau et de la Moelle épinière, Paris, France
| |
Collapse
|
31
|
Farmer WT, Murai K. Resolving Astrocyte Heterogeneity in the CNS. Front Cell Neurosci 2017; 11:300. [PMID: 29021743 PMCID: PMC5623685 DOI: 10.3389/fncel.2017.00300] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/11/2017] [Indexed: 01/30/2023] Open
Abstract
Astrocytes play essential roles in nearly all aspects of brain function from modulating synapses and neurovasculature to preserving appropriate extracellular solute concentrations. To meet the complex needs of the central nervous system (CNS), astrocytes possess highly specialized properties that are optimized for their surrounding neural circuitry. Precisely how these diverse astrocytes types are generated in vivo, however, remains poorly understood. Key to this process is a critical balance of intrinsic developmental patterning and context-dependent environmental signaling events that configures astrocyte phenotype. Indeed, emerging lines of evidence indicate that persistent cues from neighboring cells in the mature CNS cooperate with early patterning events to promote astrocyte diversity. Consistent with this, manipulating Sonic hedgehog (Shh), Notch and fibroblast growth factor (FGF) signaling in the adult brain, have profound effects on the structural, morphological and physiological state of mature astrocytes. These pathways may become disrupted in various neurological diseases and contribute to CNS pathology. This mini-review article focuses on how context-dependent environmental cues cooperate with intrinsic developmental patterning events to control astrocyte diversity in vivo in order to promote healthy brain function.
Collapse
Affiliation(s)
- W Todd Farmer
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Keith Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| |
Collapse
|
32
|
Li H, Yang Q, Han X, Tan X, Qin J, Jin G. Low-dose DHA-induced astrocyte proliferation can be attenuated by insufficient expression of BLBP in vitro. Prostaglandins Other Lipid Mediat 2017; 134:114-122. [PMID: 28917610 DOI: 10.1016/j.prostaglandins.2017.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/02/2017] [Accepted: 09/12/2017] [Indexed: 11/24/2022]
Abstract
Docosahexaenoic acid (DHA) is an n-3 long chain polyunsaturated fatty acid (PUFA) that is involved in a wide range of cellular processes in human cells. Brain lipid binding protein (BLBP) exhibits a high affinity for n-3 PUFAs, especially DHA, but the precise functional contributions of DHA and BLBP in astrocytes are not clear. We analyzed cell viability and the ratio of Ki67 positive cells after manipulating DHA and/or BLBP levels in cultured astrocytes, and found that low-dose DHA stimulated proliferation of astrocytes, whereas this proliferative effect could be attenuated by downregulation of BLBP. Moreover, we found that astrocyte proliferation was directly regulated by BLBP independently of DHA. Taken together, low-dose DHA-induced astrocyte proliferation was disturbed by insufficient BLBP; and besides acting as a fatty acid transporter, BLBP was also involved in the proliferation of astrocytes directly.
Collapse
Affiliation(s)
- Haoming Li
- Department of Anatomy, Institute of Neurobiology, Collaborative Innovation Center of Inflammatory Microenviroment, Medical School, Nantong University, Nantong 226001, China
| | - Qingqing Yang
- Xinglin College, Department of Medicine, Nantong University, Nantong 226001, China
| | - Xiao Han
- Department of Anatomy, Institute of Neurobiology, Collaborative Innovation Center of Inflammatory Microenviroment, Medical School, Nantong University, Nantong 226001, China
| | - Xuefeng Tan
- Department of Anatomy, Institute of Neurobiology, Collaborative Innovation Center of Inflammatory Microenviroment, Medical School, Nantong University, Nantong 226001, China
| | - Jianbing Qin
- Department of Anatomy, Institute of Neurobiology, Collaborative Innovation Center of Inflammatory Microenviroment, Medical School, Nantong University, Nantong 226001, China.
| | - Guohua Jin
- Department of Anatomy, Institute of Neurobiology, Collaborative Innovation Center of Inflammatory Microenviroment, Medical School, Nantong University, Nantong 226001, China.
| |
Collapse
|
33
|
Richier B, Vijandi CDM, Mackensen S, Salecker I. Lapsyn controls branch extension and positioning of astrocyte-like glia in the Drosophila optic lobe. Nat Commun 2017; 8:317. [PMID: 28827667 PMCID: PMC5567088 DOI: 10.1038/s41467-017-00384-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 06/21/2017] [Indexed: 11/09/2022] Open
Abstract
Astrocytes have diverse, remarkably complex shapes in different brain regions. Their branches closely associate with neurons. Despite the importance of this heterogeneous glial cell type for brain development and function, the molecular cues controlling astrocyte branch morphogenesis and positioning during neural circuit assembly remain largely unknown. We found that in the Drosophila visual system, astrocyte-like medulla neuropil glia (mng) variants acquire stereotypic morphologies with columnar and layered branching patterns in a stepwise fashion from mid-metamorphosis onwards. Using knockdown and loss-of-function analyses, we uncovered a previously unrecognized role for the transmembrane leucine-rich repeat protein Lapsyn in regulating mng development. lapsyn is expressed in mng and cell-autonomously required for branch extension into the synaptic neuropil and anchoring of cell bodies at the neuropil border. Lapsyn works in concert with the fibroblast growth factor (FGF) pathway to promote branch morphogenesis, while correct positioning is essential for mng survival mediated by gliotrophic FGF signaling. How glial cells, such as astrocytes, acquire their characteristic morphology during development is poorly understood. Here the authors describe the morphogenesis of astrocyte-like glia in the Drosophila optic lobe, and through a RNAi screen, they identify a transmembrane LRR protein–Lapsyn–that plays a critical role in this process.
Collapse
Affiliation(s)
- Benjamin Richier
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.,The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | | | - Stefanie Mackensen
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.,University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, 48149, Muenster, Germany
| | - Iris Salecker
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
34
|
Fibroblast growth factor signaling instructs ensheathing glia wrapping of Drosophila olfactory glomeruli. Proc Natl Acad Sci U S A 2017; 114:7505-7512. [PMID: 28674010 PMCID: PMC5530699 DOI: 10.1073/pnas.1706533114] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
This research reports that reciprocal interactions between Drosophila olfactory neurons and ensheathing glia mediate the formation of neuronal compartments, groups of synapses that are packed into discrete structures called “glomeruli” that carry specific olfactory information. Ensheathing glia respond to a neuronal cue, the FGF Thisbe, to pattern the boundaries of the nascent compartments. Neural compartments, in turn, require such glial barriers to separate themselves from neighboring compartments and thus ensure the correct organization of the olfactory circuit. These findings highlight the importance of glia in the assembly and maintenance of neural circuits and the functions of FGF signaling in these processes. The formation of complex but highly organized neural circuits requires interactions between neurons and glia. During the assembly of the Drosophila olfactory circuit, 50 olfactory receptor neuron (ORN) classes and 50 projection neuron (PN) classes form synaptic connections in 50 glomerular compartments in the antennal lobe, each of which represents a discrete olfactory information-processing channel. Each compartment is separated from the adjacent compartments by membranous processes from ensheathing glia. Here we show that Thisbe, an FGF released from olfactory neurons, particularly from local interneurons, instructs ensheathing glia to wrap each glomerulus. The Heartless FGF receptor acts cell-autonomously in ensheathing glia to regulate process extension so as to insulate each neuropil compartment. Overexpressing Thisbe in ORNs or PNs causes overwrapping of the glomeruli their axons or dendrites target. Failure to establish the FGF-dependent glia structure disrupts precise ORN axon targeting and discrete glomerular formation.
Collapse
|
35
|
Gzielo K, Kielbinski M, Ploszaj J, Janeczko K, Gazdzinski SP, Setkowicz Z. Long-Term Consumption of High-Fat Diet in Rats: Effects on Microglial and Astrocytic Morphology and Neuronal Nitric Oxide Synthase Expression. Cell Mol Neurobiol 2017; 37:783-789. [PMID: 27541371 PMCID: PMC5435787 DOI: 10.1007/s10571-016-0417-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/10/2016] [Indexed: 12/30/2022]
Abstract
Obesity in humans is associated with cognitive decline and elevated risk of neurodegenerative diseases of old age. Variations of high-fat diet are often used to model these effects in animal studies. However, we previously reported improvements in markers of memory and learning, as well as larger hippocampi and higher metabolite concentrations in Wistar rats fed high-fat, high-carbohydrate diet (HFCD, 60 % energy from fat, 28 % from carbohydrates) for 1 year; this diet leads to mild ketonemia (Setkowicz et al. in PLoS One 10:e0139987, 2015). In the present study, we follow up on this cohort to assess glial morphology and expression of markers related to gliosis. Twenty-five male Wistar rats were kept on HFCD and twenty-five on normal chow. At 12 months of age, the animals were sacrificed and processed for immunohistochemical staining for astrocytic (glial fibrillary acidic protein), microglial (Iba1), and neuronal (neuronal nitric oxide synthetase, nNOS) markers in the hippocampus. We have found changes in immunopositive area fraction and cellular complexity, as studied by a simplified Sholl procedure. To our knowledge, this study is the first to apply this methodology to the study of glial cells in HFCD animals. GFAP and Iba1 immunoreactive area fraction in the hippocampi of HFCD-fed rats were decreased, while the mean number of intersections (an indirect measure of cell complexity) was decreased in GFAP-positive astrocytes, but not in Iba1-expressing microglia. At the same time, nNOS expression was lowered after HFCD in both the cortex and the hippocampus.
Collapse
Affiliation(s)
- Kinga Gzielo
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland.
| | - Michal Kielbinski
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Jakub Ploszaj
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Krzysztof Janeczko
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Stefan P Gazdzinski
- Military Institute of Aviation Medicine, Krasinskiego 54, 01-755, Warsaw, Poland
| | - Zuzanna Setkowicz
- Department of Neuroanatomy, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| |
Collapse
|
36
|
Cabezas R, Vega-Vela NE, González-Sanmiguel J, González J, Esquinas P, Echeverria V, Barreto GE. PDGF-BB Preserves Mitochondrial Morphology, Attenuates ROS Production, and Upregulates Neuroglobin in an Astrocytic Model Under Rotenone Insult. Mol Neurobiol 2017; 55:3085-3095. [DOI: 10.1007/s12035-017-0567-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/19/2017] [Indexed: 12/21/2022]
|
37
|
Ruzafa N, Rey-Santano C, Mielgo V, Pereiro X, Vecino E. Effect of hypoxia on the retina and superior colliculus of neonatal pigs. PLoS One 2017; 12:e0175301. [PMID: 28407001 PMCID: PMC5391064 DOI: 10.1371/journal.pone.0175301] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 03/23/2017] [Indexed: 11/20/2022] Open
Abstract
Purpose To evaluate the effect of hypoxia on the neonatal pig retina and brain, we analysed the retinal ganglion cells (RGCs) and neurons in the superior colliculus, as well as the response of astrocytes in both these central nervous system (CNS) structures. Methods Newborn pigs were exposed to 120 minutes of hypoxia, induced by decreasing the inspiratory oxygen fraction (FiO2: 10–15%), followed by a reoxygenation period of 240 minutes (FiO2: 21–35%). RGCs were quantified using Brn3a, a specific nuclear marker for these cells, and apoptosis was assessed through the appearance of active caspase-3. A morphometric analysis of the cytoskeleton of astrocytes (identified with GFAP) was performed in both the retina and superior colliculus. Results Hypoxia produced no significant change in the RGCs, although, it did induce a 37.63% increase in the number of active caspase-3 positive cells in the superior colliculus. This increase was particularly evident in the superficial layers of the superior colliculus, where 56.93% of the cells were positive for active caspase-3. In addition, hypoxia induced changes in the morphology of the astrocytes in the superior colliculus but not in the retina. Conclusions Hypoxia in the neonatal pig does not affect the retina but it does affect more central structures in the brain, increasing the number of apoptotic cells in the superior colliculus and inducing changes in astrocyte morphology. This distinct sensibility to hypoxia may pave the way to design specific approaches to combat the effects of hypoxia in specific areas of the CNS.
Collapse
Affiliation(s)
- Noelia Ruzafa
- Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| | - Carmen Rey-Santano
- Research Unit for Experimental Neonatal Respiratory Physiology, Cruces University Hospital, Barakaldo, Vizcaya, Spain
| | - Victoria Mielgo
- Research Unit for Experimental Neonatal Respiratory Physiology, Cruces University Hospital, Barakaldo, Vizcaya, Spain
| | - Xandra Pereiro
- Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| | - Elena Vecino
- Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Vizcaya, Spain
- * E-mail:
| |
Collapse
|
38
|
Zhang Y, Xu S, Liang KY, Li K, Zou ZP, Yang CL, Tan K, Cao X, Jiang Y, Gao TM, Bai XC. Neuronal mTORC1 Is Required for Maintaining the Nonreactive State of Astrocytes. J Biol Chem 2016; 292:100-111. [PMID: 27895121 DOI: 10.1074/jbc.m116.744482] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/10/2016] [Indexed: 11/06/2022] Open
Abstract
Astrocytes respond to CNS insults through reactive astrogliosis, but the underlying mechanisms are unclear. In this study, we show that inactivation of mechanistic target of rapamycin complex (mTORC1) signaling in postnatal neurons induces reactive astrogliosis in mice. Ablation of Raptor (an mTORC1-specific component) in postmitotic neurons abolished mTORC1 activity and produced neurons with smaller soma and fewer dendrites, resulting in microcephaly and aberrant behavior in adult mice. Interestingly, extensive astrogliosis without significant astrocyte proliferation and glial scar formation was observed in these mice. The inhibition of neuronal mTORC1 may activate astrogliosis by reducing neuron-derived fibroblast growth factor 2 (FGF-2), which might trigger FGF receptor signaling in astrocytes to maintain their nonreactive state, and FGF-2 injection successfully prevented astrogliosis in Raptor knock-out mice. This study demonstrates that neuronal mTORC1 inhibits reactive astrogliosis and plays an important role in CNS pathologies.
Collapse
Affiliation(s)
- Yue Zhang
- From the Academy of Orthopedics, Guangdong Province, the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China.,the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Song Xu
- From the Academy of Orthopedics, Guangdong Province, the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China.,the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Kang-Yan Liang
- the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Kai Li
- the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Zhi-Peng Zou
- the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Cui-Lan Yang
- the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Kang Tan
- the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Xiong Cao
- the Department of Neurobiology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China, and
| | - Yu Jiang
- the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Tian-Ming Gao
- the Department of Neurobiology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China, and
| | - Xiao-Chun Bai
- From the Academy of Orthopedics, Guangdong Province, the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China, .,the Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
39
|
Mishra A. Binaural blood flow control by astrocytes: listening to synapses and the vasculature. J Physiol 2016; 595:1885-1902. [PMID: 27619153 DOI: 10.1113/jp270979] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/15/2016] [Indexed: 12/28/2022] Open
Abstract
Astrocytes are the most common glial cells in the brain with fine processes and endfeet that intimately contact both neuronal synapses and the cerebral vasculature. They play an important role in mediating neurovascular coupling (NVC) via several astrocytic Ca2+ -dependent signalling pathways such as K+ release through BK channels, and the production and release of arachidonic acid metabolites. They are also involved in maintaining the resting tone of the cerebral vessels by releasing ATP and COX-1 derivatives. Evidence also supports a role for astrocytes in maintaining blood pressure-dependent change in cerebrovascular tone, and perhaps also in blood vessel-to-neuron signalling as posited by the 'hemo-neural hypothesis'. Thus, astrocytes are emerging as new stars in preserving the intricate balance between the high energy demand of active neurons and the supply of oxygen and nutrients from the blood by maintaining both resting blood flow and activity-evoked changes therein. Following neuropathology, astrocytes become reactive and many of their key signalling mechanisms are altered, including those involved in NVC. Furthermore, as they can respond to changes in vascular pressure, cardiovascular diseases might exert previously unknown effects on the central nervous system by altering astrocyte function. This review discusses the role of astrocytes in neurovascular signalling in both physiology and pathology, and the impact of these findings on understanding BOLD-fMRI signals.
Collapse
Affiliation(s)
- Anusha Mishra
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
40
|
O'Neill P, Lindsay SL, Pantiru A, Guimond SE, Fagoe N, Verhaagen J, Turnbull JE, Riddell JS, Barnett SC. Sulfatase-mediated manipulation of the astrocyte-Schwann cell interface. Glia 2016; 65:19-33. [PMID: 27535874 PMCID: PMC5244676 DOI: 10.1002/glia.23047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 12/29/2022]
Abstract
Schwann cell (SC) transplantation following spinal cord injury (SCI) may have therapeutic potential. Functional recovery is limited however, due to poor SC interactions with host astrocytes and the induction of astrogliosis. Olfactory ensheathing cells (OECs) are closely related to SCs, but intermix more readily with astrocytes in culture and induce less astrogliosis. We previously demonstrated that OECs express higher levels of sulfatases, enzymes that remove 6-O-sulfate groups from heparan sulphate proteoglycans, than SCs and that RNAi knockdown of sulfatase prevented OEC-astrocyte mixing in vitro. As human OECs are difficult to culture in large numbers we have genetically engineered SCs using lentiviral vectors to express sulfatase 1 and 2 (SC-S1S2) and assessed their ability to interact with astrocytes. We demonstrate that SC-S1S2s have increased integrin-dependent motility in the presence of astrocytes via modulation of NRG and FGF receptor-linked PI3K/AKT intracellular signaling and do not form boundaries with astrocytes in culture. SC-astrocyte mixing is dependent on local NRG concentration and we propose that sulfatase enzymes influence the bioavailability of NRG ligand and thus influence SC behavior. We further demonstrate that injection of sulfatase expressing SCs into spinal cord white matter results in less glial reactivity than control SC injections comparable to that of OEC injections. Our data indicate that sulfatase-mediated modification of the extracellular matrix can influence glial interactions with astrocytes, and that SCs engineered to express sulfatase may be more OEC-like in character. This approach may be beneficial for cell transplant-mediated spinal cord repair. GLIA 2016 GLIA 2017;65:19-33.
Collapse
Affiliation(s)
- Paul O'Neill
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Susan L Lindsay
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Andreea Pantiru
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Scott E Guimond
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Nitish Fagoe
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, Amsterdam, BA, 1105, the Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, Amsterdam, BA, 1105, the Netherlands
| | - Jeremy E Turnbull
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - John S Riddell
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Susan C Barnett
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| |
Collapse
|
41
|
Stewart CE, Corella KM, Samberg BD, Jones PT, Linscott ML, Chung WCJ. Perinatal midline astrocyte development is impaired in fibroblast growth factor 8 hypomorphic mice. Brain Res 2016; 1646:287-296. [PMID: 27291295 DOI: 10.1016/j.brainres.2016.06.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 05/27/2016] [Accepted: 06/09/2016] [Indexed: 12/31/2022]
Abstract
Our previous studies showed that Fgf8 mutations can cause Kallmann syndrome (KS), a form of congenital hypogonadotropic hypogonadism, in which patients do not undergo puberty and are infertile. Interestingly, some KS patients also have agenesis of the corpus callosum (ACC) suggesting that KS pathology is not limited to reproductive function. Here, we asked whether FGF8 dysfunction is the underlying cause of ACC in some KS patients. Indeed, early studies in transgenic mice with Fgf8 mutations reported the presence of failed or incomplete corpus callosum formation. Additional studies in transgenic mice showed that FGF8 function most likely prevents the prenatal elimination of glial fibrillary acidic protein (GFAP)-immunoreactive (IR) glial cells in the indusium griseum (IG) and midline zipper (MZ), two anterior-dorsal midline regions required for corpus callosum formation (i.e., between embryonic days (E) 15.5-18.5). Here, we tested the hypothesis that FGF8 function is critical for the survival of the GFAP-IR midline glial cells. First, we measured the incidence of apoptosis in the anterior-dorsal midline region in Fgf8 hypomorphic mice during embryonic corpus callosum formation. Second, we quantified the GFAP expression in the anterior-dorsal midbrain region during pre- and postnatal development, in order to study: 1) how Fgf8 hypomorphy disrupts prenatal GFAP-IR midline glial cell development, and 2) whether Fgf8 hypomorphy continues to disrupt postnatal GFAP-IR midline glial cell development. Our results indicate that perinatal FGF8 signaling is important for the timing of the onset of anterior-dorsal Gfap expression in midline glial cells suggesting that FGF8 function regulates midline GFAP-IR glial cell development, which when disrupted by Fgf8 deficiency prevents the formation of the corpus callosum. These studies provide an experimentally-based mechanistic explanation as to why corpus callosum formation may fail in KS patients with deficits in FGF signaling.
Collapse
Affiliation(s)
- Courtney E Stewart
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Kristina M Corella
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | - Brittany D Samberg
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Paula T Jones
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Megan L Linscott
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Wilson C J Chung
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA; Department of Biological Sciences, Kent State University, Kent, OH 44242, USA.
| |
Collapse
|
42
|
Mechanisms of Reduced Astrocyte Surface Coverage in Cortical Neuron-Glia Co-cultures on Nanoporous Gold Surfaces. Cell Mol Bioeng 2016; 9:433-442. [PMID: 27795742 DOI: 10.1007/s12195-016-0449-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nanoporous gold (np-Au) is a promising multifunctional material for neural electrodes. We have previously shown that np-Au nanotopography reduces astrocyte surface coverage (linked to undesirable gliosis) while maintaining high neuronal coverage in a cortical primary neuron-glia co-culture model as long as two weeks in vitro. Here, we investigate the potential influence of secreted soluble factors from cells grown on np-Au on the cell type-specific surface coverage of cells grown on conventional tissue culture plastic and test the hypothesis that secretion of factors is responsible for inhibiting astrocyte coverage on np-Au. In order to assess whether factors secreted from cells grown on np-Au surfaces reduced surface coverage by astrocytes, we seeded fresh primary rat neuron-glia co-cultures on conventional polystyrene culture dishes, but maintained the cells in conditioned media from co-cultures grown on np-Au surfaces. After one week in vitro, a preferential reduction in astrocyte surface coverage was not observed, suggesting that soluble factors are not playing a role. In contrast, four hours after cell seeding there were a significant number of non-adhered, yet still viable, cells for the cultures on np-Au surfaces. We hypothesize that the non-adherent cells are mainly astrocytes, because: (i) there was no difference in neuronal cell coverage between np-Au and pl-Au for long culture durations and (ii) neurons are post-mitotic and not expected to increase in number upon attaching to the surface. Overall, the results suggest that the np-Au topography leads to preferential neuronal attachment shortly after cell seeding and limits astrocyte-specific np-Au surface coverage at longer culture durations.
Collapse
|
43
|
Hizay A, Seitz M, Grosheva M, Sinis N, Kaya Y, Bendella H, Sarikcioglu L, Dunlop SA, Angelov DN. FGF-2 is required to prevent astrogliosis in the facial nucleus after facial nerve injury and mechanical stimulation of denervated vibrissal muscles. J Biomed Res 2016; 30:142-148. [PMID: 28276669 PMCID: PMC4820891 DOI: 10.7555/jbr.30.20140042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 11/28/2014] [Accepted: 04/10/2015] [Indexed: 01/20/2023] Open
Abstract
Recently, we have shown that manual stimulation of paralyzed vibrissal muscles after facial-facial anastomosis reduced the poly-innervation of neuromuscular junctions and restored vibrissal whisking. Using gene knock outs, we found a differential dependence of manual stimulation effects on growth factors. Thus, insulin-like growth factor-1 and brain-derived neurotrophic factor are required to underpin manual stimulation-mediated improvements, whereas FGF-2 is not. The lack of dependence on FGF-2 in mediating these peripheral effects prompted us to look centrally, i.e. within the facial nucleus where increased astrogliosis after facial-facial anastomosis follows "synaptic stripping". We measured the intensity of Cy3-fluorescence after immunostaining for glial fibrillary acidic protein (GFAP) as an indirect indicator of synaptic coverage of axotomized neurons in the facial nucleus of mice lacking FGF-2 (FGF-2-/- mice). There was no difference in GFAP-Cy3-fluorescence (pixel number, gray value range 17–103) between intact wildtype mice (2.12± 0.37×107) and their intact FGF-2-/- counterparts (2.12± 0.27×107) nor after facial-facial anastomosis +handling (wildtype: 4.06± 0.32×107; FGF-2-/-: 4.39±0.17×107). However, after facial-facial anastomosis, GFAP-Cy3-fluorescence remained elevated in FGF-2-/--animals (4.54±0.12×107), whereas manual stimulation reduced the intensity of GFAP-immunofluorescence in wild type mice to values that were not significantly different from intact mice (2.63± 0.39×10 ). We conclude that FGF-2 is not required to underpin the beneficial effects of manual stimulation at the neuro-muscular junction, but it is required to minimize astrogliosis in the brainstem and, by implication, restore synaptic coverage of recovering facial motoneurons.
Collapse
Affiliation(s)
- Arzu Hizay
- Department of Anatomy, Akdeniz University, Faculty of Medicine, Dumlupinar Bulvari 07058 Kampus, Antalya, Turkey
| | - Mark Seitz
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Strasse 9, D-50924 Cologne, FR Germany
| | - Maria Grosheva
- Department of Oto-Rhino-Laryngology, University Hospital Cologne, Joseph-Stelzmann-Strasse 9, D-50924 Cologne, FR Germany
| | - Nektarios Sinis
- Depeartment of Plastic-, Hand- and Reconstructive Microsurgery, Berlin, FR 12249, Germany
| | - Yasemin Kaya
- Department of Anatomy, Akdeniz University, Faculty of Medicine, Dumlupinar Bulvari 07058 Kampus, Antalya, Turkey
| | - Habib Bendella
- Department of Neurosurgery, Hospital Merheim, University of Witten-Herdecke, Ostmerheimer Straße 200, 51109 Cologne, Germany
| | - Levent Sarikcioglu
- Department of Anatomy, Akdeniz University, Faculty of Medicine, Dumlupinar Bulvari 07058 Kampus, Antalya, Turkey
| | - Sarah A Dunlop
- Experimental and Regenerative Neurosciences, School of Animal Biology, University of Western Australia (M092) 35 Stirling Highway, CRAWLEY WA 6009, Australia
| | - Doychin N Angelov
- Department of Anatomy I, University of Cologne, Joseph-Stelzmann-Strasse 9, D-50924 Cologne, FR Germany;
| |
Collapse
|
44
|
Gilmour AD, Woolley AJ, Poole-Warren LA, Thomson CE, Green RA. A critical review of cell culture strategies for modelling intracortical brain implant material reactions. Biomaterials 2016; 91:23-43. [PMID: 26994876 DOI: 10.1016/j.biomaterials.2016.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/29/2016] [Accepted: 03/06/2016] [Indexed: 02/07/2023]
Abstract
The capacity to predict in vivo responses to medical devices in humans currently relies greatly on implantation in animal models. Researchers have been striving to develop in vitro techniques that can overcome the limitations associated with in vivo approaches. This review focuses on a critical analysis of the major in vitro strategies being utilized in laboratories around the world to improve understanding of the biological performance of intracortical, brain-implanted microdevices. Of particular interest to the current review are in vitro models for studying cell responses to penetrating intracortical devices and their materials, such as electrode arrays used for brain computer interface (BCI) and deep brain stimulation electrode probes implanted through the cortex. A background on the neural interface challenge is presented, followed by discussion of relevant in vitro culture strategies and their advantages and disadvantages. Future development of 2D culture models that exhibit developmental changes capable of mimicking normal, postnatal development will form the basis for more complex accurate predictive models in the future. Although not within the scope of this review, innovations in 3D scaffold technologies and microfluidic constructs will further improve the utility of in vitro approaches.
Collapse
Affiliation(s)
- A D Gilmour
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - A J Woolley
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia; Western Sydney University, Sydney, NSW, Australia
| | - L A Poole-Warren
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - C E Thomson
- Department of Veterinary Medicine, University of Alaska, Fairbanks, AK 99775, USA
| | - R A Green
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
45
|
Pineda JR, Encinas JM. The Contradictory Effects of Neuronal Hyperexcitation on Adult Hippocampal Neurogenesis. Front Neurosci 2016; 10:74. [PMID: 26973452 PMCID: PMC4776215 DOI: 10.3389/fnins.2016.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/17/2016] [Indexed: 01/19/2023] Open
Abstract
Adult hippocampal neurogenesis is a highly plastic process that responds swiftly to neuronal activity. Adult hippocampal neurogenesis can be regulated at the level of neural stem cell recruitment and activation, progenitor proliferation, as well as newborn cell survival and differentiation. An "excitation-neurogenesis" rule was proposed after the demonstration of the capability of cultured neural stem and progenitor cells to intrinsically sense neuronal excitatory activity. In vivo, this property has remained elusive although recently the direct response of neural stem cells to GABA in the hippocampus via GABAA receptors has evidenced a mechanism for a direct talk between neurons and neural stem cells. As it is pro-neurogenic, the effect of excitatory neuronal activity has been generally considered beneficial. But what happens in situations of neuronal hyperactivity in which neurogenesis can be dramatically boosted? In animal models, electroconvulsive shock markedly increases neurogenesis. On the contrary, in epilepsy rodent models, seizures induce the generation of misplaced neurons with abnormal morphological and electrophysiological properties, namely aberrant neurogenesis. We will herein discuss what is known about the mechanisms of influence of neurons on neural stem cells, as well as the severe effects of neuronal hyperexcitation on hippocampal neurogenesis.
Collapse
Affiliation(s)
- José R Pineda
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for Neuroscience Zamudio, Spain
| | - Juan M Encinas
- Laboratory of Neural Stem Cells and Neurogenesis, Achucarro Basque Center for NeuroscienceZamudio, Spain; IKERBASQUE, The Basque Foundation for ScienceBilbao, Spain
| |
Collapse
|
46
|
Krencik R, Hokanson KC, Narayan AR, Dvornik J, Rooney GE, Rauen KA, Weiss LA, Rowitch DH, Ullian EM. Dysregulation of astrocyte extracellular signaling in Costello syndrome. Sci Transl Med 2016; 7:286ra66. [PMID: 25947161 DOI: 10.1126/scitranslmed.aaa5645] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astrocytes produce an assortment of signals that promote neuronal maturation according to a precise developmental timeline. Is this orchestrated timing and signaling altered in human neurodevelopmental disorders? To address this question, the astroglial lineage was investigated in two model systems of a developmental disorder with intellectual disability caused by mutant Harvey rat sarcoma viral oncogene homolog (HRAS) termed Costello syndrome: mutant HRAS human induced pluripotent stem cells (iPSCs) and transgenic mice. Human iPSCs derived from patients with Costello syndrome differentiated to astroglia more rapidly in vitro than those derived from wild-type cell lines with normal HRAS, exhibited hyperplasia, and also generated an abundance of extracellular matrix remodeling factors and proteoglycans. Acute treatment with a farnesyl transferase inhibitor and knockdown of the transcription factor SNAI2 reduced expression of several proteoglycans in Costello syndrome iPSC-derived astrocytes. Similarly, mice in which mutant HRAS was expressed selectively in astrocytes exhibited experience-independent increased accumulation of perineuronal net proteoglycans in cortex, as well as increased parvalbumin expression in interneurons, when compared to wild-type mice. Our data indicate that astrocytes expressing mutant HRAS dysregulate cortical maturation during development as shown by abnormal extracellular matrix remodeling and implicate excessive astrocyte-to-neuron signaling as a possible drug target for treating mental impairment and enhancing neuroplasticity.
Collapse
Affiliation(s)
- Robert Krencik
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kenton C Hokanson
- Neuroscience Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Aditi R Narayan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jill Dvornik
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gemma E Rooney
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Katherine A Rauen
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
| | - Lauren A Weiss
- Department of Psychiatry and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Institute for Regenerative Medicine and Stem Cell Research, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Erik M Ullian
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA. Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
47
|
Lee HR, Farhanullah, Lee J, Jajoo R, Kong SY, Shin JY, Kim JO, Lee J, Lee J, Kim HJ. Discovery of a Small Molecule that Enhances Astrocytogenesis by Activation of STAT3, SMAD1/5/8, and ERK1/2 via Induction of Cytokines in Neural Stem Cells. ACS Chem Neurosci 2016; 7:90-9. [PMID: 26505647 DOI: 10.1021/acschemneuro.5b00243] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Identification of small molecules that direct neural stem cells (NSCs) into specific cell types would be helpful to understand the molecular mechanisms involved in regulation of NSC fate, and facilitate the development of therapeutic applications. In the current study, we developed and screened small molecules that can modulate the fate of NSCs that are derived from rat fetal cortex. Among these compounds, compounds 5 and 6 successfully differentiated NSCs into astrocytes and neurons, respectively. Compound 5 induced astrocytogenesis by increasing expression of interleukin-6, bone morphogenetic protein 2 and leukemia inhibitory factor and through consequent phosphorylation of signal transducer and activator of transcription 3 and Sma- and Mad-related protein 1/5/8 in NSCs. In addition, compound 5 increased the expression of fibroblast growth factor (FGF) 2 and FGF8 which may regulate the branching and morphology of astrocytes. Taken together, our results suggest that these small molecules can serve as a useful tool to study cell fate determination in NSCs and be used as an inexpensive alternative to cytokines to study mechanisms of astrocytogenesis.
Collapse
Affiliation(s)
- Ha-Rim Lee
- College
of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Farhanullah
- GVK Bioscience Pvt
Ltd, Hyderabad-500076, India
| | - JiSoo Lee
- College
of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Rahul Jajoo
- GVK Bioscience Pvt
Ltd, Hyderabad-500076, India
| | - Sun-Young Kong
- College
of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Jae-Yeon Shin
- College
of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Jae-Ouk Kim
- Laboratory
Science Division, International Vaccine Institute, Seoul 151-919, Korea
| | - Jiyoun Lee
- Department
of Global Medical Science, Sungshin University, Seoul 142-732, Korea
| | - Jeewoo Lee
- College
of Pharmacy, Seoul National University, Seoul 151-742, Korea
| | - Hyun-Jung Kim
- College
of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| |
Collapse
|
48
|
Pearson-Leary J, Osborne DM, McNay EC. Role of Glia in Stress-Induced Enhancement and Impairment of Memory. Front Integr Neurosci 2016; 9:63. [PMID: 26793072 PMCID: PMC4707238 DOI: 10.3389/fnint.2015.00063] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/05/2015] [Indexed: 12/20/2022] Open
Abstract
Both acute and chronic stress profoundly affect hippocampally-dependent learning and memory: moderate stress generally enhances, while chronic or extreme stress can impair, neural and cognitive processes. Within the brain, stress elevates both norepinephrine and glucocorticoids, and both affect several genomic and signaling cascades responsible for modulating memory strength. Memories formed at times of stress can be extremely strong, yet stress can also impair memory to the point of amnesia. Often overlooked in consideration of the impact of stress on cognitive processes, and specifically memory, is the important contribution of glia as a target for stress-induced changes. Astrocytes, microglia, and oligodendrocytes all have unique contributions to learning and memory. Furthermore, these three types of glia express receptors for both norepinephrine and glucocorticoids and are hence immediate targets of stress hormone actions. It is becoming increasingly clear that inflammatory cytokines and immunomodulatory molecules released by glia during stress may promote many of the behavioral effects of acute and chronic stress. In this review, the role of traditional genomic and rapid hormonal mechanisms working in concert with glia to affect stress-induced learning and memory will be emphasized.
Collapse
Affiliation(s)
- Jiah Pearson-Leary
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia Philadelphia, PA, USA
| | | | - Ewan C McNay
- Behavioral Neuroscience and Biology, University at Albany Albany, NY, USA
| |
Collapse
|
49
|
Heller JP, Rusakov DA. Morphological plasticity of astroglia: Understanding synaptic microenvironment. Glia 2015; 63:2133-51. [PMID: 25782611 PMCID: PMC4737250 DOI: 10.1002/glia.22821] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 03/02/2015] [Indexed: 12/27/2022]
Abstract
Memory formation in the brain is thought to rely on the remodeling of synaptic connections which eventually results in neural network rewiring. This remodeling is likely to involve ultrathin astroglial protrusions which often occur in the immediate vicinity of excitatory synapses. The phenomenology, cellular mechanisms, and causal relationships of such astroglial restructuring remain, however, poorly understood. This is in large part because monitoring and probing of the underpinning molecular machinery on the scale of nanoscopic astroglial compartments remains a challenge. Here we briefly summarize the current knowledge regarding the cellular organisation of astroglia in the synaptic microenvironment and discuss molecular mechanisms potentially involved in use-dependent astroglial morphogenesis. We also discuss recent observations concerning morphological astroglial plasticity, the respective monitoring methods, and some of the newly emerging techniques that might help with conceptual advances in the area.
Collapse
Affiliation(s)
- Janosch P Heller
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, Queen Square, London, United Kingdom
| | - Dmitri A Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, Queen Square, London, United Kingdom
| |
Collapse
|
50
|
Feng D, Guo B, Liu G, Wang B, Wang W, Gao G, Qin H, Wu S. FGF2 alleviates PTSD symptoms in rats by restoring GLAST function in astrocytes via the JAK/STAT pathway. Eur Neuropsychopharmacol 2015; 25:1287-99. [PMID: 25979764 DOI: 10.1016/j.euroneuro.2015.04.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/06/2015] [Accepted: 04/24/2015] [Indexed: 12/18/2022]
Abstract
In our previous study, we demonstrated that fibroblast growth factor 2 (FGF2) administration alleviated posttraumatic stress disorder (PTSD) symptoms via an "astrocyte-related" mechanism. We further investigated the changes in the astrocytic glutamate transporters GLAST and GLT-1 and in JAK/STAT3 signaling (which is involved in astrocyte activation and GLAST/GLT-1 function) in single prolonged stress (SPS) model rats. High-performance liquid chromatography (HPLC), Western blot and immunohistochemistry analyses revealed a significant SPS-induced increase in the concentration of glutamate in the cerebrospinal fluid and decrease in GLAST/GLT-1 expression and JAK/STAT3 signaling. Treatment with FGF2 significantly alleviated GLAST/GLT-1 dysfunction, JAK/STAT3 signaling inhibition, and the behavioral abnormalities. The administration of the JAK/STAT pathway inhibitor AG490 blocked the effects of FGF2 on PTSD symptoms, astrocyte activation, and GLAST, but not GLT-1, expression in vivo and in vitro. Our findings suggest that astrocytic JAK/STAT signaling is associated with SPS-induced GLAST dysfunction and that FGF2 protects against PTSD symptoms by restoring astrocytic glutamate uptake via the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Dayun Feng
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, PR China; Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Baolin Guo
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, PR China
| | - Gaohua Liu
- Department of Psychosomatics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Bao Wang
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Wen Wang
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, PR China
| | - Guodong Gao
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, PR China
| | - Huaizhou Qin
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, PR China.
| | - Shengxi Wu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|