51
|
Torres-Ceja B, Olsen ML. A closer look at astrocyte morphology: Development, heterogeneity, and plasticity at astrocyte leaflets. Curr Opin Neurobiol 2022; 74:102550. [PMID: 35544965 PMCID: PMC9376008 DOI: 10.1016/j.conb.2022.102550] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/20/2022] [Accepted: 04/03/2022] [Indexed: 11/20/2022]
Abstract
Astrocytes represent an abundant type of glial cell involved in nearly every aspect of central nervous system (CNS) function, including synapse formation and maturation, ion and neurotransmitter homeostasis, blood-brain barrier maintenance, as well as neuronal metabolic support. These various functions are enabled by the morphological complexity that astrocytes adopt. Recent experimental advances in genetic and viral labeling, lineage tracing, and live- and ultrastructural imaging of miniscule astrocytic sub-compartments reveal a complex morphological heterogeneity that is based on the origin, local function, and environmental context in which astrocytes reside. In this minireview, we highlight recent findings that reveal the plastic nature of astrocytes in the healthy brain, particularly at the synapse, and emerging technologies that have advanced our understanding of these morphologically complex cells.
Collapse
Affiliation(s)
- Beatriz Torres-Ceja
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA. https://twitter.com/beatriztc
| | - Michelle L Olsen
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
52
|
Lawal O, Ulloa Severino FP, Eroglu C. The role of astrocyte structural plasticity in regulating neural circuit function and behavior. Glia 2022; 70:1467-1483. [PMID: 35535566 PMCID: PMC9233050 DOI: 10.1002/glia.24191] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022]
Abstract
Brain circuits undergo substantial structural changes during development, driven by the formation, stabilization, and elimination of synapses. Synaptic connections continue to undergo experience‐dependent structural rearrangements throughout life, which are postulated to underlie learning and memory. Astrocytes, a major glial cell type in the brain, are physically in contact with synaptic circuits through their structural ensheathment of synapses. Astrocytes strongly contribute to the remodeling of synaptic structures in healthy and diseased central nervous systems by regulating synaptic connectivity and behaviors. However, whether structural plasticity of astrocytes is involved in their critical functions at the synapse is unknown. This review will discuss the emerging evidence linking astrocytic structural plasticity to synaptic circuit remodeling and regulation of behaviors. Moreover, we will survey possible molecular and cellular mechanisms regulating the structural plasticity of astrocytes and their non‐cell‐autonomous effects on neuronal plasticity. Finally, we will discuss how astrocyte morphological changes in different physiological states and disease conditions contribute to neuronal circuit function and dysfunction.
Collapse
Affiliation(s)
- Oluwadamilola Lawal
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Francesco Paolo Ulloa Severino
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neuroscience and Psychology, Duke University, Durham, North Carolina, USA.,Howard Hughes Medical Institute, Duke University, Durham, North Carolina, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA.,Howard Hughes Medical Institute, Duke University, Durham, North Carolina, USA.,Duke Institute for Brain Sciences, Durham, North Carolina, USA
| |
Collapse
|
53
|
Jin T, Zhang Y, Botchway BOA, Zhang J, Fan R, Zhang Y, Liu X. Curcumin can improve Parkinson's disease via activating BDNF/PI3k/Akt signaling pathways. Food Chem Toxicol 2022; 164:113091. [PMID: 35526734 DOI: 10.1016/j.fct.2022.113091] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023]
Abstract
Parkinson's disease is a common progressive neurodegenerative disease, and presently has no curative agent. Curcumin, as one of the natural polyphenols, has great potential in neurodegenerative diseases and other different pathological settings. The brain-derived neurotrophic factor (BDNF) and phosphatidylinositol 3 kinase (PI3k)/protein kinase B (Akt) signaling pathways are significantly involved nerve regeneration and anti-apoptotic activities. Currently, relevant studies have confirmed that curcumin has an optimistic impact on neuroprotection via regulating BDNF and PI3k/Akt signaling pathways in neurodegenerative disease. Here, we summarized the relationship between BDNF and PI3k/Akt signaling pathway, the main biological functions and neuroprotective effects of curcumin via activating BDNF and PI3k/Akt signaling pathways in Parkinson's disease. This paper illustrates that curcumin, as a neuroprotective agent, can delay the progression of Parkinson's disease by protecting nerve cells.
Collapse
Affiliation(s)
- Tian Jin
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Zhang
- Department of Pharmacology, Medical College, Shaoxing University, Zhejiang, China
| | - Ruihua Fan
- School of Life Science, Shaoxing University, Zhejiang, China
| | - Yufeng Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China.
| |
Collapse
|
54
|
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
|
55
|
Ameroso D, Meng A, Chen S, Felsted J, Dulla CG, Rios M. Astrocytic BDNF signaling within the ventromedial hypothalamus regulates energy homeostasis. Nat Metab 2022; 4:627-643. [PMID: 35501599 PMCID: PMC9177635 DOI: 10.1038/s42255-022-00566-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is essential for maintaining energy and glucose balance within the central nervous system. Because the study of its metabolic actions has been limited to effects in neuronal cells, its role in other cell types within the brain remains poorly understood. Here we show that astrocytic BDNF signaling within the ventromedial hypothalamus (VMH) modulates neuronal activity in response to changes in energy status. This occurs via the truncated TrkB.T1 receptor. Accordingly, either fasting or central BDNF depletion enhances astrocytic synaptic glutamate clearance, thereby decreasing neuronal activity in mice. Notably, selective depletion of TrkB.T1 in VMH astrocytes blunts the effects of energy status on excitatory transmission, as well as on responses to leptin, glucose and lipids. These effects are driven by increased astrocytic invasion of excitatory synapses, enhanced glutamate reuptake and decreased neuronal activity. We thus identify BDNF/TrkB.T1 signaling in VMH astrocytes as an essential mechanism that participates in energy and glucose homeostasis.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Stella Chen
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Jennifer Felsted
- Graduate Program in Biochemical and Molecular Nutrition, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Chris G Dulla
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
56
|
Dolotov OV, Inozemtseva LS, Myasoedov NF, Grivennikov IA. Stress-Induced Depression and Alzheimer's Disease: Focus on Astrocytes. Int J Mol Sci 2022; 23:4999. [PMID: 35563389 PMCID: PMC9104432 DOI: 10.3390/ijms23094999] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases and depression are multifactorial disorders with a complex and poorly understood physiopathology. Astrocytes play a key role in the functioning of neurons in norm and pathology. Stress is an important factor for the development of brain disorders. Here, we review data on the effects of stress on astrocyte function and evidence of the involvement of astrocyte dysfunction in depression and Alzheimer's disease (AD). Stressful life events are an important risk factor for depression; meanwhile, depression is an important risk factor for AD. Clinical data indicate atrophic changes in the same areas of the brain, the hippocampus and prefrontal cortex (PFC), in both pathologies. These brain regions play a key role in regulating the stress response and are most vulnerable to the action of glucocorticoids. PFC astrocytes are critically involved in the development of depression. Stress alters astrocyte function and can result in pyroptotic death of not only neurons, but also astrocytes. BDNF-TrkB system not only plays a key role in depression and in normalizing the stress response, but also appears to be an important factor in the functioning of astrocytes. Astrocytes, being a target for stress and glucocorticoids, are a promising target for the treatment of stress-dependent depression and AD.
Collapse
Affiliation(s)
- Oleg V. Dolotov
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (O.V.D.); (L.S.I.); (N.F.M.)
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 119234 Moscow, Russia
| | - Ludmila S. Inozemtseva
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (O.V.D.); (L.S.I.); (N.F.M.)
| | - Nikolay F. Myasoedov
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (O.V.D.); (L.S.I.); (N.F.M.)
| | - Igor A. Grivennikov
- Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, 123182 Moscow, Russia; (O.V.D.); (L.S.I.); (N.F.M.)
| |
Collapse
|
57
|
Cheon S, Culver AM, Bagnell AM, Ritchie FD, Vacharasin JM, McCord MM, Papendorp CM, Chukwurah E, Smith AJ, Cowen MH, Moreland TA, Ghate PS, Davis SW, Liu JS, Lizarraga SB. Counteracting epigenetic mechanisms regulate the structural development of neuronal circuitry in human neurons. Mol Psychiatry 2022; 27:2291-2303. [PMID: 35210569 PMCID: PMC9133078 DOI: 10.1038/s41380-022-01474-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 02/02/2022] [Indexed: 01/23/2023]
Abstract
Autism spectrum disorders (ASD) are associated with defects in neuronal connectivity and are highly heritable. Genetic findings suggest that there is an overrepresentation of chromatin regulatory genes among the genes associated with ASD. ASH1 like histone lysine methyltransferase (ASH1L) was identified as a major risk factor for ASD. ASH1L methylates Histone H3 on Lysine 36, which is proposed to result primarily in transcriptional activation. However, how mutations in ASH1L lead to deficits in neuronal connectivity associated with ASD pathogenesis is not known. We report that ASH1L regulates neuronal morphogenesis by counteracting the catalytic activity of Polycomb Repressive complex 2 group (PRC2) in stem cell-derived human neurons. Depletion of ASH1L decreases neurite outgrowth and decreases expression of the gene encoding the neurotrophin receptor TrkB whose signaling pathway is linked to neuronal morphogenesis. The neuronal morphogenesis defect is overcome by inhibition of PRC2 activity, indicating that a balance between the Trithorax group protein ASH1L and PRC2 activity determines neuronal morphology. Thus, our work suggests that ASH1L may epigenetically regulate neuronal morphogenesis by modulating pathways like the BDNF-TrkB signaling pathway. Defects in neuronal morphogenesis could potentially impair the establishment of neuronal connections which could contribute to the neurodevelopmental pathogenesis associated with ASD in patients with ASH1L mutations.
Collapse
Affiliation(s)
- Seonhye Cheon
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Allison M Culver
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Anna M Bagnell
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Foster D Ritchie
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Janay M Vacharasin
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Mikayla M McCord
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Carin M Papendorp
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Evelyn Chukwurah
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Austin J Smith
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Mara H Cowen
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Trevor A Moreland
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Pankaj S Ghate
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Shannon W Davis
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA
| | - Judy S Liu
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, USA
- Department of Neurology, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Sofia B Lizarraga
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA.
- Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
58
|
Tessarollo L, Yanpallewar S. TrkB Truncated Isoform Receptors as Transducers and Determinants of BDNF Functions. Front Neurosci 2022; 16:847572. [PMID: 35321093 PMCID: PMC8934854 DOI: 10.3389/fnins.2022.847572] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) belongs to the neurotrophin family of secreted growth factors and binds with high affinity to the TrkB tyrosine kinase receptors. BDNF is a critical player in the development of the central (CNS) and peripheral (PNS) nervous system of vertebrates and its strong pro-survival function on neurons has attracted great interest as a potential therapeutic target for the management of neurodegenerative disorders such as Amyotrophic Lateral Sclerosis (ALS), Huntington, Parkinson's and Alzheimer's disease. The TrkB gene, in addition to the full-length receptor, encodes a number of isoforms, including some lacking the catalytic tyrosine kinase domain. Importantly, one of these truncated isoforms, namely TrkB.T1, is the most widely expressed TrkB receptor in the adult suggesting an important role in the regulation of BDNF signaling. Although some progress has been made, the mechanism of TrkB.T1 function is still largely unknown. Here we critically review the current knowledge on TrkB.T1 distribution and functions that may be helpful to our understanding of how it regulates and participates in BDNF signaling in normal physiological and pathological conditions.
Collapse
Affiliation(s)
- Lino Tessarollo
- Neural Development Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | | |
Collapse
|
59
|
Albizzati E, Florio E, Miramondi F, Sormonta I, Landsberger N, Frasca A. Identification of Region-Specific Cytoskeletal and Molecular Alterations in Astrocytes of Mecp2 Deficient Animals. Front Neurosci 2022; 16:823060. [PMID: 35242007 PMCID: PMC8886113 DOI: 10.3389/fnins.2022.823060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that represents the most common genetic cause of severe intellectual disability in females. Most patients carry mutations in the X-linked MECP2 gene, coding for the methyl-CpG-binding protein 2 (MeCP2), originally isolated as an epigenetic transcriptional factor able to bind methylated DNA and repress transcription. Recent data implicated a role for glia in RTT, showing that astrocytes express Mecp2 and that its deficiency affects their ability to support neuronal maturation by non-cell autonomous mechanisms. To date, some molecular, structural and functional alterations have been attributed to Mecp2 null astrocytes, but how they evolve over time and whether they follow a spatial heterogeneity are two aspects which deserve further investigations. In this study, we assessed cytoskeletal features of astrocytes in Mecp2 deficient brains by analyzing their arbor complexity and processes in reconstructed GFAP+ cells at different ages, corresponding to peculiar stages of the disorder, and in different cerebral regions (motor and somatosensory cortices and CA1 layer of hippocampus). Our findings demonstrate the presence of defects in Mecp2 null astrocytes that worsen along disease progression and strictly depend on the brain area, highlighting motor and somatosensory cortices as the most affected regions. Of relevance, astrocyte cytoskeleton is impaired also in the somatosensory cortex of symptomatic heterozygous animals, with Mecp2 + astrocytes showing slightly more pronounced defects with respect to the Mecp2 null cells, emphasizing the importance of non-cell autonomous effects. We reported a temporal correlation between the progressive thinning of layer I and the atrophy of astrocytes, suggesting that their cytoskeletal dysfunctions might contribute to cortical defects. Considering the reciprocal link between morphology and function in astrocytes, we analyzed the effect of Mecp2 deficiency on the expression of selected astrocyte-enriched genes, which describe typical astrocytic features. qRT-PCR data corroborated our results, reporting an overall decrement of gene expression, which is area and age-dependent. In conclusion, our data show that Mecp2 deficiency causes structural and molecular alterations in astrocytes, which progress along with the severity of symptoms and diversely occur in the different cerebral regions, highlighting the importance of considering heterogeneity when studying astrocytes in RTT.
Collapse
Affiliation(s)
- Elena Albizzati
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Florio
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Federica Miramondi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Irene Sormonta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicoletta Landsberger
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelisa Frasca
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
60
|
Nelissen E, Possemis N, Van Goethem NP, Schepers M, Mulder-Jongen DAJ, Dietz L, Janssen W, Gerisch M, Hüser J, Sandner P, Vanmierlo T, Prickaerts J. The sGC stimulator BAY-747 and activator runcaciguat can enhance memory in vivo via differential hippocampal plasticity mechanisms. Sci Rep 2022; 12:3589. [PMID: 35246566 PMCID: PMC8897390 DOI: 10.1038/s41598-022-07391-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/10/2022] [Indexed: 12/22/2022] Open
Abstract
Soluble guanylate cyclase (sGC) requires a heme-group bound in order to produce cGMP, a second messenger involved in memory formation, while heme-free sGC is inactive. Two compound classes can increase sGC activity: sGC stimulators acting on heme-bound sGC, and sGC activators acting on heme-free sGC. In this rodent study, we investigated the potential of the novel brain-penetrant sGC stimulator BAY-747 and sGC activator runcaciguat to enhance long-term memory and attenuate short-term memory deficits induced by the NOS-inhibitor L-NAME. Furthermore, hippocampal plasticity mechanisms were investigated. In vivo, oral administration of BAY-747 and runcaciguat to male Wistar rats enhanced memory acquisition in the object location task (OLT), while only BAY-747 reversed L-NAME induced memory impairments in the OLT. Ex vivo, both BAY-747 and runcaciguat enhanced hippocampal GluA1-containing AMPA receptor (AMPAR) trafficking in a chemical LTP model for memory acquisition using acute mouse hippocampal slices. In vivo only runcaciguat acted on the glutamatergic AMPAR system in hippocampal memory acquisition processes, while for BAY-747 the effects on the neurotrophic system were more pronounced as measured in male mice using western blot. Altogether this study shows that sGC stimulators and activators have potential as cognition enhancers, while the underlying plasticity mechanisms may determine disease-specific effectiveness.
Collapse
Affiliation(s)
- Ellis Nelissen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Nina Possemis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Nick P Van Goethem
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Melissa Schepers
- Neuro-Immune Connect and Repair Lab, Biomedical Research Institute, Hasselt University, 3500, Hasselt, Belgium
| | - Danielle A J Mulder-Jongen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Lisa Dietz
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113, Wuppertal, Germany
| | - Wiebke Janssen
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113, Wuppertal, Germany
| | - Michael Gerisch
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113, Wuppertal, Germany
| | - Jörg Hüser
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113, Wuppertal, Germany
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113, Wuppertal, Germany
- Hannover Medical School, 30625, Hannover, Germany
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- Neuro-Immune Connect and Repair Lab, Biomedical Research Institute, Hasselt University, 3500, Hasselt, Belgium
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
61
|
Ultrastructural view of astrocyte arborization, astrocyte-astrocyte and astrocyte-synapse contacts, intracellular vesicle-like structures, and mitochondrial network. Prog Neurobiol 2022; 213:102264. [DOI: 10.1016/j.pneurobio.2022.102264] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 12/15/2022]
|
62
|
Wang J, Gao F, Cui S, Yang S, Gao F, Wang X, Zhu G. Utility of 7,8-dihydroxyflavone in preventing astrocytic and synaptic deficits in the hippocampus elicited by PTSD. Pharmacol Res 2022; 176:106079. [PMID: 35026406 DOI: 10.1016/j.phrs.2022.106079] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 02/07/2023]
Abstract
Astrocytic functions and brain-derived neurotrophic factor (BDNF)-tyrosine kinase receptor B (TrkB) signaling pathways are impaired in stress-related neuropsychiatric diseases. Previous studies have reported neuroprotective effects of 7,8-dihydroxyflavone (7,8-DHF), a TrkB activator. Here, we investigated the molecular mechanisms underlying pathogenesis of post-traumatic stress disorder (PTSD) using a modified single-prolonged stress (SPS&S) model and the potential beneficial effects of 7,8-DHF. SPS&S reduced the hippocampal expression of glial fibrillary acidic protein (GFAP), a marker of astrocytes, and induced morphological changes in astrocytes. From the perspective of synaptic function, the SPS&S model displayed reduced expression of BDNF, p-TrkB, postsynaptic density protein 95 (PSD95), AMPA receptor subunit GluR1 (GluA1), NMDA receptor subunit N2A/N2B ratio, calpain-1, phosphorylated protein kinase B (Akt) and phosphorylated mammalian target of rapamycin (mTOR) and conversely, higher phosphatase and tension homolog (PTEN) expression in the hippocampus. Acute or continuous intraperitoneal administration of 7,8-DHF (5 mg/kg) after SPS&S procedures prevented SPS&S-induced fear memory generalization and anxiety-like behaviors as well as abnormalities of hippocampal oscillations. Most importantly, 7,8-DHF attenuated SPS&S-induced abnormal BDNF-TrkB signaling and calpain-1-dependent cascade of synaptic deficits. Furthermore, treatment with a TrkB inhibitor completely blocked while an mTOR inhibitor partially blocked the effects of 7,8-DHF on behavioral changes of SPS&S model mice. Our collective findings suggest that 7,8-DHF effectively alleviates PTSD-like symptoms, including fear generalization and anxiety-like behavior, potentially by preventing astrocytic and synaptic deficits in the hippocampus through targeting of TrkB.
Collapse
Affiliation(s)
- Juan Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Feng Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Shuai Cui
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Shaojie Yang
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Fang Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Xuncui Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China; Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.
| |
Collapse
|
63
|
Bupivacaine reduces GlyT1 expression by potentiating the p-AMPKα/BDNF signalling pathway in spinal astrocytes of rats. Sci Rep 2022; 12:1378. [PMID: 35082359 PMCID: PMC8792009 DOI: 10.1038/s41598-022-05478-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022] Open
Abstract
Bupivacaine, a local anaesthetic, is widely applied in the epidural or subarachnoid space to clinically manage acute and chronic pain. However, the underlying mechanisms are complex and unclear. Glycine transporter 1 (GlyT1) in the spinal cord plays a critical role in various pathologic pain conditions. Therefore, we sought to determine whether bupivacaine exerts its analgesic effect by regulating GlyT1 expression and to determine the underlying mechanisms of regulation. Primary astrocytes prepared from the spinal cord of rats were treated with bupivacaine. The protein levels of GlyT1, brain-derived neurotrophic factor (BDNF) and phosphorylated adenosine 5′-monophosphate (AMP)-activated protein kinase α (p-AMPKα) were measured by western blotting or immunofluorescence. In addition, 7,8-dihydroxyflavone (7,8-DHF, BDNF receptor agonist) and AMPK shRNA were applied to verify the relationship between the regulation of GlyT1 by bupivacaine and the p-AMPKα/BDNF signalling pathway. After treatment with bupivacaine, GlyT1 expression was diminished in a concentration-dependent manner, while the expression of BDNF and p-AMPK was increased. Moreover, 7,8-DHF decreased GlyT1 expression, and AMPK knockdown suppressed the upregulation of BDNF expression by bupivacaine. Finally, we concluded that bupivacaine reduced GlyT1 expression in spinal astrocytes by activating the p-AMPKα/BDNF signalling pathway. These results provide a new mechanism for the analgesic effect of intrathecal bupivacaine in the treatment of acute and chronic pain.
Collapse
|
64
|
George KK, Heithoff BP, Shandra O, Robel S. Mild Traumatic Brain Injury/Concussion Initiates an Atypical Astrocyte Response Caused by Blood-Brain Barrier Dysfunction. J Neurotrauma 2022; 39:211-226. [PMID: 34806422 PMCID: PMC8785769 DOI: 10.1089/neu.2021.0204] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Mild traumatic brain injury/concussion (mTBI) accounts for 70-90% of all reported TBI cases and causes long-lasting neurological consequences in 10-40% of patients. Recent clinical studies revealed increased blood-brain barrier (BBB) permeability in mTBI patients, which correlated with secondary damage after mTBI. However, the cascade of cellular events initiated by exposure to blood-borne factors resulting in sustained damage is not fully understood. We previously reported that astrocytes respond atypically to mTBI, rapidly losing many proteins essential to their homeostatic function, while classic scar formation does not occur. Here, we tested the hypothesis that mTBI-induced BBB damage causes atypical astrocytes through exposure to blood-borne factors. Using an mTBI mouse model, two-photon imaging, an endothelial cell-specific genetic ablation approach, and serum-free primary astrocyte cultures, we demonstrated that areas with atypical astrocytes coincide with BBB damage and that exposure of astrocytes to plasma proteins is sufficient to initiate loss of astrocyte homeostatic proteins. Although mTBI resulted in frequent impairment of both physical and metabolic BBB properties and leakage of small-sized blood-borne factors, deposition of the coagulation factor fibrinogen or vessel rupture were rare. Surprisingly, even months after mTBI, BBB repair did not occur in areas with atypical astrocytes. Together, these findings implicate that even relatively small BBB disturbances are sustained long term, and render nearby astrocytes dysfunctional, likely at the cost of neuronal health and function.
Collapse
Affiliation(s)
- Kijana K. George
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Roanoke, Virginia, USA
| | - Benjamin P. Heithoff
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Oleksii Shandra
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Stefanie Robel
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA.,Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Address correspondence to: Stefanie Robel, PhD, University of Alabama at Birmingham, Department of Cell, Developmental, and Integrative Biology, McCallum Basic Health Sciences Building, 9th floor, 1720 2nd Avenue South, Birmingham, AL 35294-0006, USA
| |
Collapse
|
65
|
Abstract
Astroglia are key regulators of synaptic function, playing central roles in homeostatic ion buffering, energy dynamics, transmitter uptake, maintenance of neurotransmitter pools, and regulation of synaptic plasticity through release of neuroactive chemicals. Given the myriad of crucial homeostatic and signaling functions attributed to astrocytes and the variety of neurotransmitter receptors expressed by astroglia, they serve as prime cellular candidates for establishing maladaptive synaptic plasticity following drug exposure. Initial studies on astroglia and addiction have placed drug-mediated disruptions in the homeostatic regulation of glutamate as a central aspect of relapse vulnerability. However, the generation of sophisticated tools to study and manipulate astroglia have proven that the interaction between addictive substances, astroglia, and relapse-relevant synaptic plasticity extends far beyond the homeostatic regulation of glutamate. Here we present astroglial systems impacted by drug exposure and discuss how changes in astroglial biology contribute to addiction biology.
Collapse
|
66
|
Jo J, Woo J, Cristobal CD, Choi JM, Wang C, Ye Q, Smith JA, Ung K, Liu G, Cortes D, Jung SY, Arenkiel BR, Lee HK. Regional heterogeneity of astrocyte morphogenesis dictated by the formin protein, Daam2, modifies circuit function. EMBO Rep 2021; 22:e53200. [PMID: 34633730 PMCID: PMC8647146 DOI: 10.15252/embr.202153200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 01/07/2023] Open
Abstract
Astrocytes display extraordinary morphological complexity that is essential to support brain circuit development and function. Formin proteins are key regulators of the cytoskeleton; however, their role in astrocyte morphogenesis across diverse brain regions and neural circuits is unknown. Here, we show that loss of the formin protein Daam2 in astrocytes increases morphological complexity in the cortex and olfactory bulb, but elicits opposing effects on astrocytic calcium dynamics. These differential physiological effects result in increased excitatory synaptic activity in the cortex and increased inhibitory synaptic activity in the olfactory bulb, leading to altered olfactory behaviors. Proteomic profiling and immunoprecipitation experiments identify Slc4a4 as a binding partner of Daam2 in the cortex, and combined deletion of Daam2 and Slc4a4 restores the morphological alterations seen in Daam2 mutants. Our results reveal new mechanisms regulating astrocyte morphology and show that congruent changes in astrocyte morphology can differentially influence circuit function.
Collapse
Affiliation(s)
- Juyeon Jo
- Department of PediatricsSection of NeurologyBaylor College of MedicineHoustonTXUSA
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
| | - Junsung Woo
- Center for Cell and Gene TherapyBaylor College of MedicineHoustonTXUSA
| | - Carlo D Cristobal
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
- Program in Integrative Molecular and Biomedical SciencesBaylor College of MedicineHoustonTXUSA
| | - Jong Min Choi
- Center for Molecular DiscoveryDepartment of Biochemistry and Molecular BiologyBaylor College of MedicineHoustonTXUSA
| | - Chih‐Yen Wang
- Department of PediatricsSection of NeurologyBaylor College of MedicineHoustonTXUSA
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
| | - Qi Ye
- Department of PediatricsSection of NeurologyBaylor College of MedicineHoustonTXUSA
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
| | - Joshua A Smith
- Department of PediatricsSection of NeurologyBaylor College of MedicineHoustonTXUSA
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
| | - Kevin Ung
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Gary Liu
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
| | - Diego Cortes
- Department of PediatricsSection of NeurologyBaylor College of MedicineHoustonTXUSA
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
| | - Sung Yun Jung
- Center for Molecular DiscoveryDepartment of Biochemistry and Molecular BiologyBaylor College of MedicineHoustonTXUSA
| | - Benjamin R Arenkiel
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
| | - Hyun Kyoung Lee
- Department of PediatricsSection of NeurologyBaylor College of MedicineHoustonTXUSA
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalHoustonTXUSA
- Program in Integrative Molecular and Biomedical SciencesBaylor College of MedicineHoustonTXUSA
- Program in Developmental BiologyBaylor College of MedicineHoustonTXUSA
- Department of NeuroscienceBaylor College of MedicineHoustonTXUSA
| |
Collapse
|
67
|
Serafini G, Trabucco A, Corsini G, Escelsior A, Amerio A, Aguglia A, Nasrallah H, Amore M. The potential of microRNAs as putative biomarkers in major depressive disorder and suicidal behavior. Biomark Neuropsychiatry 2021. [DOI: 10.1016/j.bionps.2021.100035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
68
|
Heard KJ, Shokhirev MN, Becronis C, Fredlender C, Zahid N, Le AT, Ji Y, Skime M, Nelson T, Hall-Flavin D, Weinshilboum R, Gage FH, Vadodaria KC. Chronic cortisol differentially impacts stem cell-derived astrocytes from major depressive disorder patients. Transl Psychiatry 2021; 11:608. [PMID: 34848679 PMCID: PMC8632962 DOI: 10.1038/s41398-021-01733-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023] Open
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric disorder, and exposure to stress is a robust risk factor for MDD. Clinical data and rodent models have indicated the negative impact of chronic exposure to stress-induced hormones like cortisol on brain volume, memory, and cell metabolism. However, the cellular and transcriptomic changes that occur in the brain after prolonged exposure to cortisol are less understood. Furthermore, the astrocyte-specific contribution to cortisol-induced neuropathology remains understudied. Here, we have developed an in vitro model of "chronic stress" using human induced pluripotent stem cell (iPSC)-derived astrocytes treated with cortisol for 7 days. Whole transcriptome sequencing reveals differentially expressed genes (DEGs) uniquely regulated in chronic cortisol compared to acute cortisol treatment. Utilizing this paradigm, we examined the stress response transcriptome of astrocytes generated from MDD patient iPSCs. The MDD-specific DEGs are related to GPCR ligand binding, synaptic signaling, and ion homeostasis. Together, these data highlight the unique role astrocytes play in the central nervous system and present interesting genes for future study into the relationship between chronic stress and MDD.
Collapse
Affiliation(s)
- Kelly J. Heard
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Maxim N. Shokhirev
- grid.250671.70000 0001 0662 7144The Razavi Newman Integrative Genomics and Bioinformatics Core (IGC), The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA USA
| | - Caroline Becronis
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Callie Fredlender
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Nadia Zahid
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Amy T. Le
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Yuan Ji
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA ,grid.223827.e0000 0001 2193 0096University of Utah School of Medicine, Salt Lake City, UT USA
| | - Michelle Skime
- grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic, Rochester, MN USA
| | - Timothy Nelson
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | - Daniel Hall-Flavin
- grid.66875.3a0000 0004 0459 167XDepartment of Psychiatry and Psychology, Mayo Clinic, Rochester, MN USA
| | - Richard Weinshilboum
- grid.66875.3a0000 0004 0459 167XDepartment of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA
| | - Fred H. Gage
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Krishna C. Vadodaria
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| |
Collapse
|
69
|
Ohlig S, Clavreul S, Thorwirth M, Simon-Ebert T, Bocchi R, Ulbricht S, Kannayian N, Rossner M, Sirko S, Smialowski P, Fischer-Sternjak J, Götz M. Molecular diversity of diencephalic astrocytes reveals adult astrogenesis regulated by Smad4. EMBO J 2021; 40:e107532. [PMID: 34549820 PMCID: PMC8561644 DOI: 10.15252/embj.2020107532] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 08/09/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022] Open
Abstract
Astrocytes regulate brain‐wide functions and also show region‐specific differences, but little is known about how general and region‐specific functions are aligned at the single‐cell level. To explore this, we isolated adult mouse diencephalic astrocytes by ACSA‐2‐mediated magnetic‐activated cell sorting (MACS). Single‐cell RNA‐seq revealed 7 gene expression clusters of astrocytes, with 4 forming a supercluster. Within the supercluster, cells differed by gene expression related to ion homeostasis or metabolism, with the former sharing gene expression with other regions and the latter being restricted to specific regions. All clusters showed expression of proliferation‐related genes, and proliferation of diencephalic astrocytes was confirmed by immunostaining. Clonal analysis demonstrated low level of astrogenesis in the adult diencephalon, but not in cerebral cortex grey matter. This led to the identification of Smad4 as a key regulator of diencephalic astrocyte in vivo proliferation and in vitro neurosphere formation. Thus, astrocytes show diverse gene expression states related to distinct functions with some subsets being more widespread while others are more regionally restricted. However, all share low‐level proliferation revealing the novel concept of adult astrogenesis in the diencephalon.
Collapse
Affiliation(s)
- Stefanie Ohlig
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Solène Clavreul
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Manja Thorwirth
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Tatiana Simon-Ebert
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Riccardo Bocchi
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Sabine Ulbricht
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Nirmal Kannayian
- Molecular Neurobiology, Department of Psychiatry, LMU Munich, Munich, Germany
| | - Moritz Rossner
- Molecular Neurobiology, Department of Psychiatry, LMU Munich, Munich, Germany
| | - Swetlana Sirko
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Pawel Smialowski
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Judith Fischer-Sternjak
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany
| | - Magdalena Götz
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, Munich, Germany.,Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Neuherberg, Germany.,SYNERGY, Excellence cluster of Systems Neurology, LMU Munich, Munich, Germany
| |
Collapse
|
70
|
Han J, Yoon S, Park H. Endocytic BDNF secretion regulated by Vamp3 in astrocytes. Sci Rep 2021; 11:21203. [PMID: 34707216 PMCID: PMC8551197 DOI: 10.1038/s41598-021-00693-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/15/2021] [Indexed: 11/26/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) regulates diverse brain functions via TrkB receptor signaling. Due to the expression of TrkB receptors, astrocytes can internalize extracellular BDNF proteins via receptor-mediated endocytosis. Endocytosed BDNF can be re-secreted upon stimulation, but the molecular mechanism underlying this phenomenon remains unrecognized. Our study reveals that vesicle-associated membrane protein 3 (Vamp3) selectively regulates the release of endocytic BDNF from astrocytes. By using quantum dot (QD)-conjugated mature BDNF (QD-BDNF) as a proxy for the extracellular BDNF protein, we monitored the uptake, transport, and secretion of BDNF from cultured cortical astrocytes. Our data showed that endocytic QD-BDNF particles were enriched in Vamp3-containing vesicles in astrocytes and that ATP treatment sufficiently triggered either the antero- or retrograde transport and exocytosis of QD-BDNF-containing vesicles. Downregulation of Vamp3 expression disrupted endocytic BDNF secretion from astrocytes but did not affect uptake or transport. Collectively, these results provide evidence of the selective ability of astrocytic Vamp3 to control endocytic BDNF secretion during BDNF recycling.
Collapse
Affiliation(s)
- Jeongho Han
- Research Group of Neurovascular Unit, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Sungryeong Yoon
- Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, South Korea
| | - Hyungju Park
- Research Group of Neurovascular Unit, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea. .,Department of Brain and Cognitive Sciences, DGIST, Daegu, 42988, South Korea.
| |
Collapse
|
71
|
Wahid RM, Samy W, El-Sayed SF. Cognitive impairment in obese rat model: role of glial cells. Int J Obes (Lond) 2021; 45:2191-2196. [PMID: 34140627 DOI: 10.1038/s41366-021-00880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/03/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Obesity is a worldwide problem. Some studies revealed that it leads to deterioration of the cognitive function, regardless of age. AIM OF THE STUDY explore the effect of obesity on cognitive function in a rat model of obesity highlighting the role of glial cells. MATERIALS AND METHODS twenty adult male albino rats were assigned to two groups: group I: consumed normal diet, group II: consumed high-fat diet. Body Mass Index (BMI), serum glucose, insulin, HOMA IR and lipid profile were measured. Also, hippocampal expression of Brain derived neurotrophic factor (Bdnf), synapsin, Ionized calcium binding adaptor molecule 1 (Iba), nuclear factor erythroid -related factor 2 (Nrf2), Myelin basic protein (Mbp) were measured by real-time polymerase chain reaction. The Morris Water Maze is a test used to assess spatial learning and memory capacities of rats. RESULTS There was a high significant increase in lipid profile, serum glucose, insulin serum levels and HOMA-IR in obese groups with impaired Morris water maze performance compared to control group. There was a significant downregulation in hippocampal Bdnf and synapsin mRNA expression. In addition to decrease in Mbp mRNA expression (P < 0.001). This could be explained by oxidative stress through significant downregulation of Nrf2 mRNA, and inflammation observed in significant upregulation Iba mRNA gene expression in the obese group. CONCLUSION Many factors contribute to obesity associated cognitive impairment. In our study, we figured out the crucial roles of glial cells including microglial activation and oligodendrocytes affection with other underlying mechanisms including oxidative stress and hippocampal inflammation.
Collapse
Affiliation(s)
- Reham M Wahid
- Physiology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Walaa Samy
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sherein F El-Sayed
- Physiology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
72
|
Liu X, Ying J, Wang X, Zheng Q, Zhao T, Yoon S, Yu W, Yang D, Fang Y, Hua F. Astrocytes in Neural Circuits: Key Factors in Synaptic Regulation and Potential Targets for Neurodevelopmental Disorders. Front Mol Neurosci 2021; 14:729273. [PMID: 34658786 PMCID: PMC8515196 DOI: 10.3389/fnmol.2021.729273] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes are the major glial cells in the brain, which play a supporting role in the energy and nutritional supply of neurons. They were initially regarded as passive space-filling cells, but the latest progress in the study of the development and function of astrocytes highlights their active roles in regulating synaptic transmission, formation, and plasticity. In the concept of "tripartite synapse," the bidirectional influence between astrocytes and neurons, in addition to their steady-state and supporting function, suggests that any negative changes in the structure or function of astrocytes will affect the activity of neurons, leading to neurodevelopmental disorders. The role of astrocytes in the pathophysiology of various neurological and psychiatric disorders caused by synaptic defects is increasingly appreciated. Understanding the roles of astrocytes in regulating synaptic development and the plasticity of neural circuits could help provide new treatments for these diseases.
Collapse
Affiliation(s)
- Xing Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Tiancheng Zhao
- Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Sungtae Yoon
- Helping Minds International Charitable Foundation, New York, NY, United States
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Danying Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| |
Collapse
|
73
|
Prowse N, Hayley S. Microglia and BDNF at the crossroads of stressor related disorders: Towards a unique trophic phenotype. Neurosci Biobehav Rev 2021; 131:135-163. [PMID: 34537262 DOI: 10.1016/j.neubiorev.2021.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
Stressors ranging from psychogenic/social to neurogenic/injury to systemic/microbial can impact microglial inflammatory processes, but less is known regarding their effects on trophic properties of microglia. Recent studies do suggest that microglia can modulate neuronal plasticity, possibly through brain derived neurotrophic factor (BDNF). This is particularly important given the link between BDNF and neuropsychiatric and neurodegenerative pathology. We posit that certain activated states of microglia play a role in maintaining the delicate balance of BDNF release onto neuronal synapses. This focused review will address how different "activators" influence the expression and release of microglial BDNF and address the question of tropomyosin receptor kinase B (TrkB) expression on microglia. We will then assess sex-based differences in microglial function and BDNF expression, and how microglia are involved in the stress response and related disorders such as depression. Drawing on research from a variety of other disorders, we will highlight challenges and opportunities for modulators that can shift microglia to a "trophic" phenotype with a view to potential therapeutics relevant for stressor-related disorders.
Collapse
Affiliation(s)
- Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
74
|
Jaudon F, Albini M, Ferroni S, Benfenati F, Cesca F. A developmental stage- and Kidins220-dependent switch in astrocyte responsiveness to brain-derived neurotrophic factor. J Cell Sci 2021; 134:jcs258419. [PMID: 34279618 DOI: 10.1242/jcs.258419] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022] Open
Abstract
Astroglial cells are key to maintain nervous system homeostasis. Neurotrophins are known for their pleiotropic effects on neuronal physiology but also exert complex functions to glial cells. Here, we investigated (i) the signaling competence of mouse embryonic and postnatal primary cortical astrocytes exposed to brain-derived neurotrophic factor (BDNF) and, (ii) the role of kinase D-interacting substrate of 220 kDa (Kidins220), a transmembrane scaffold protein that mediates neurotrophin signaling in neurons. We found a shift from a kinase-based response in embryonic cells to a response predominantly relying on intracellular Ca2+ transients [Ca2+]i within postnatal cultures, associated with a decrease in the synthesis of full-length BDNF receptor TrkB, with Kidins220 contributing to the BDNF-activated kinase and [Ca2+]i pathways. Finally, Kidins220 participates in the homeostatic function of astrocytes by controlling the expression of the ATP-sensitive inward rectifier potassium channel 10 (Kir4.1) and the metabolic balance of embryonic astrocytes. Overall, our data contribute to the understanding of the complex role played by astrocytes within the central nervous system, and identify Kidins220 as a novel actor in the increasing number of pathologies characterized by astrocytic dysfunctions. This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
| | - Martina Albini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Stefano Ferroni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
75
|
Zhu J, Chen F, Luo L, Wu W, Dai J, Zhong J, Lin X, Chai C, Ding P, Liang L, Wang S, Ding X, Chen Y, Wang H, Qiu J, Wang F, Sun C, Zeng Y, Fang J, Jiang X, Liu P, Tang G, Qiu X, Zhang X, Ruan Y, Jiang S, Li J, Zhu S, Xu X, Li F, Liu Z, Cao G, Chen D. Single-cell atlas of domestic pig cerebral cortex and hypothalamus. Sci Bull (Beijing) 2021; 66:1448-1461. [PMID: 36654371 DOI: 10.1016/j.scib.2021.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/07/2020] [Accepted: 03/12/2021] [Indexed: 01/20/2023]
Abstract
The brain of the domestic pig (Sus scrofa domesticus) has drawn considerable attention due to its high similarities to that of humans. However, the cellular compositions of the pig brain (PB) remain elusive. Here we investigated the single-nucleus transcriptomic profiles of five regions of the PB (frontal lobe, parietal lobe, temporal lobe, occipital lobe, and hypothalamus) and identified 21 cell subpopulations. The cross-species comparison of mouse and pig hypothalamus revealed the shared and specific gene expression patterns at the single-cell resolution. Furthermore, we identified cell types and molecular pathways closely associated with neurological disorders, bridging the gap between gene mutations and pathogenesis. We reported, to our knowledge, the first single-cell atlas of domestic pig cerebral cortex and hypothalamus combined with a comprehensive analysis across species, providing extensive resources for future research regarding neural science, evolutionary developmental biology, and regenerative medicine.
Collapse
Affiliation(s)
- Jiacheng Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Fang Chen
- BGI-Shenzhen, Shenzhen 518083, China; MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Lihua Luo
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Weiying Wu
- BGI-Shenzhen, Shenzhen 518083, China; Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310031, China
| | - Jinxia Dai
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jixing Zhong
- School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiumei Lin
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Chaochao Chai
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Peiwen Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Langchao Liang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Shiyou Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Xiangning Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Yin Chen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Haoyu Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Jiaying Qiu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | | | - Chengcheng Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China; School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yuying Zeng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China; College of Life Science, South China Agricultural University, Guangzhou 510642, China
| | - Jian Fang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xiaosen Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Ping Liu
- BGI-Shenzhen, Shenzhen 518083, China; MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Gen Tang
- Shenzhen Children's Hospital, Shenzhen 518083, China
| | - Xin Qiu
- Shenzhen Children's Hospital, Shenzhen 518083, China
| | | | - Yetian Ruan
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | | | | | - Shida Zhu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Fang Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China.
| | | |
Collapse
|
76
|
Guo L, Gao T, Gao C, Jia X, Ni J, Han C, Wang Y. Stimulation of astrocytic sigma-1 receptor is sufficient to ameliorate inflammation- induced depression. Behav Brain Res 2021; 410:113344. [PMID: 33961912 DOI: 10.1016/j.bbr.2021.113344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/16/2022]
Abstract
Astrocytes play important roles in the development of depression. As a promising target for antidepressant development, sigma-1 receptor (Sig-1R) is reported to promote activation of astrocyte in chronic stress-induced depression in our previous study. However, astrocytes are hyper-activated in inflammation-induced depression, raising concerns of whether stimulation of astrocytic Sig-1R would exert antidepressant-like effect in inflammation-induced depression. Here we reported that specific stimulation of astrocytic Sig-1R using adeno-associated virus (AAV) significantly attenuated lipopolysaccharide (LPS)- induced depressive-like behavior in the forced swim test (FST), tail suspension test (TST), sucrose preference test, and improved the memory function in novel object recognition test. Besides, specific stimulation of astrocytic Sig-1R decreased the activation of astrocyte and microglia, as well as increased brain-derived neurotrophic factor (BDNF) in LPS-induced depression. In primary cultured astrocytes, overexpression of Sig-1R also reduced the expression of IL-1β, TNF-α, iNOS during inflammation-treated astrocyte. Taken together, the results suggest that specific stimulation of astrocytic Sig-1R ameliorates inflammation-induced depressive-like behavior, providing the evidence that astrocytic Sig-1R could represent a reliable therapeutic target for depression.
Collapse
Affiliation(s)
- Lin Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, China; Department of Pharmacy, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tianyu Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, China
| | - Ce Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, China
| | - Xiaoxia Jia
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, China
| | - Jing Ni
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, China
| | - Chaojun Han
- Department of Pharmacology, College of Pharmacy, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Yun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, China.
| |
Collapse
|
77
|
Colombo E, Triolo D, Bassani C, Bedogni F, Di Dario M, Dina G, Fredrickx E, Fermo I, Martinelli V, Newcombe J, Taveggia C, Quattrini A, Comi G, Farina C. Dysregulated copper transport in multiple sclerosis may cause demyelination via astrocytes. Proc Natl Acad Sci U S A 2021; 118:e2025804118. [PMID: 34183414 PMCID: PMC8271600 DOI: 10.1073/pnas.2025804118] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Demyelination is a key pathogenic feature of multiple sclerosis (MS). Here, we evaluated the astrocyte contribution to myelin loss and focused on the neurotrophin receptor TrkB, whose up-regulation on the astrocyte finely demarcated chronic demyelinated areas in MS and was paralleled by neurotrophin loss. Mice lacking astrocyte TrkB were resistant to demyelination induced by autoimmune or toxic insults, demonstrating that TrkB signaling in astrocytes fostered oligodendrocyte damage. In vitro and ex vivo approaches highlighted that astrocyte TrkB supported scar formation and glia proliferation even in the absence of neurotrophin binding, indicating TrkB transactivation in response to inflammatory or toxic mediators. Notably, our neuropathological studies demonstrated copper dysregulation in MS and model lesions and TrkB-dependent expression of copper transporter (CTR1) on glia cells during neuroinflammation. In vitro experiments evidenced that TrkB was critical for the generation of glial intracellular calcium flux and CTR1 up-regulation induced by stimuli distinct from neurotrophins. These events led to copper uptake and release by the astrocyte, and in turn resulted in oligodendrocyte loss. Collectively, these data demonstrate a pathogenic demyelination mechanism via the astrocyte release of copper and open up the possibility of restoring copper homeostasis in the white matter as a therapeutic target in MS.
Collapse
Affiliation(s)
- Emanuela Colombo
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Daniela Triolo
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Claudia Bassani
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Francesco Bedogni
- San Raffaele Rett Research Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marco Di Dario
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Giorgia Dina
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Evelien Fredrickx
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Isabella Fermo
- Division of Immunology, Transplantation, and Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Vittorio Martinelli
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Jia Newcombe
- NeuroResource, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, WC1N 1PJ, London, UK
| | - Carla Taveggia
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Angelo Quattrini
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Giancarlo Comi
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Cinthia Farina
- Division of Neuroscience, Institute of Experimental Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132, Milan, Italy;
| |
Collapse
|
78
|
De Nicola AF, Meyer M, Garay L, Kruse MS, Schumacher M, Guennoun R, Gonzalez Deniselle MC. Progesterone and Allopregnanolone Neuroprotective Effects in the Wobbler Mouse Model of Amyotrophic Lateral Sclerosis. Cell Mol Neurobiol 2021; 42:23-40. [PMID: 34138412 DOI: 10.1007/s10571-021-01118-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023]
Abstract
Progesterone regulates a number of processes in neurons and glial cells not directly involved in reproduction or sex behavior. Several neuroprotective effects are better observed under pathological conditions, as shown in the Wobbler mouse model of amyotrophic laterals sclerosis (ALS). Wobbler mice are characterized by forelimb atrophy due to motoneuron degeneration in the spinal cord, and include microgliosis and astrogliosis. Here we summarized current evidence on progesterone reversal of Wobbler neuropathology. We demonstrated that progesterone decreased motoneuron vacuolization with preservation of mitochondrial respiratory complex I activity, decreased mitochondrial expression and activity of nitric oxide synthase, increased Mn-dependent superoxide dismutase, stimulated brain-derived neurotrophic factor, increased the cholinergic phenotype of motoneurons, and enhanced survival with a concomitant decrease of death-related pathways. Progesterone also showed differential effects on glial cells, including increased oligodendrocyte density and downregulation of astrogliosis and microgliosis. These changes associate with reduced anti-inflammatory markers. The enhanced neurochemical parameters were accompanied by longer survival and increased muscle strength in tests of motor behavior. Because progesterone is locally metabolized to allopregnanolone (ALLO) in nervous tissues, we also studied neuroprotection by this derivative. Treatment of Wobbler mice with ALLO decreased oxidative stress and glial pathology, increased motoneuron viability and clinical outcome in a progesterone-like manner, suggesting that ALLO could mediate some progesterone effects in the spinal cord. In conclusion, the beneficial effects observed in different parameters support the versatile properties of progesterone and ALLO in a mouse model of motoneuron degeneration. The studies foresee future therapeutic opportunities with neuroactive steroids for deadly diseases like ALS.
Collapse
Affiliation(s)
- Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina. .,Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425, Buenos Aires, Argentina.
| | - María Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Laura Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina.,Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425, Buenos Aires, Argentina
| | - Maria Sol Kruse
- Laboratory of Neurobiology, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Michael Schumacher
- U1195 INSERM and University Paris Sud "Neuroprotective, Neuroregenerative and Remyelinating Small Molecules, 94276, Kremlin-Bicetre, France
| | - Rachida Guennoun
- U1195 INSERM and University Paris Sud "Neuroprotective, Neuroregenerative and Remyelinating Small Molecules, 94276, Kremlin-Bicetre, France
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina.,Department of Physiological Sciences, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425, Buenos Aires, Argentina
| |
Collapse
|
79
|
Pérez-Sisqués L, Sancho-Balsells A, Solana-Balaguer J, Campoy-Campos G, Vives-Isern M, Soler-Palazón F, Anglada-Huguet M, López-Toledano MÁ, Mandelkow EM, Alberch J, Giralt A, Malagelada C. RTP801/REDD1 contributes to neuroinflammation severity and memory impairments in Alzheimer's disease. Cell Death Dis 2021; 12:616. [PMID: 34131105 PMCID: PMC8206344 DOI: 10.1038/s41419-021-03899-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023]
Abstract
RTP801/REDD1 is a stress-regulated protein whose upregulation is necessary and sufficient to trigger neuronal death. Its downregulation in Parkinson's and Huntington's disease models ameliorates the pathological phenotypes. In the context of Alzheimer's disease (AD), the coding gene for RTP801, DDIT4, is responsive to Aβ and modulates its cytotoxicity in vitro. Also, RTP801 mRNA levels are increased in AD patients' lymphocytes. However, the involvement of RTP801 in the pathophysiology of AD has not been yet tested. Here, we demonstrate that RTP801 levels are increased in postmortem hippocampal samples from AD patients. Interestingly, RTP801 protein levels correlated with both Braak and Thal stages of the disease and with GFAP expression. RTP801 levels are also upregulated in hippocampal synaptosomal fractions obtained from murine 5xFAD and rTg4510 mice models of the disease. A local RTP801 knockdown in the 5xFAD hippocampal neurons with shRNA-containing AAV particles ameliorates cognitive deficits in 7-month-old animals. Upon RTP801 silencing in the 5xFAD mice, no major changes were detected in hippocampal synaptic markers or spine density. Importantly, we found an unanticipated recovery of several gliosis hallmarks and inflammasome key proteins upon neuronal RTP801 downregulation in the 5xFAD mice. Altogether our results suggest that RTP801 could be a potential future target for theranostic studies since it could be a biomarker of neuroinflammation and neurotoxicity severity of the disease and, at the same time, a promising therapeutic target in the treatment of AD.
Collapse
Affiliation(s)
- Leticia Pérez-Sisqués
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Júlia Solana-Balaguer
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Genís Campoy-Campos
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marcel Vives-Isern
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ferran Soler-Palazón
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marta Anglada-Huguet
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
| | | | - Eva-Maria Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Catalonia, Spain
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Catalonia, Spain.
| | - Cristina Malagelada
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
80
|
Minge D, Domingos C, Unichenko P, Behringer C, Pauletti A, Anders S, Herde MK, Delekate A, Gulakova P, Schoch S, Petzold GC, Henneberger C. Heterogeneity and Development of Fine Astrocyte Morphology Captured by Diffraction-Limited Microscopy. Front Cell Neurosci 2021; 15:669280. [PMID: 34149361 PMCID: PMC8211899 DOI: 10.3389/fncel.2021.669280] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/06/2021] [Indexed: 12/26/2022] Open
Abstract
The fine processes of single astrocytes can contact many thousands of synapses whose function they can modulate through bi-directional signaling. The spatial arrangement of astrocytic processes and neuronal structures is relevant for such interactions and for the support of neuronal signaling by astrocytes. At the same time, the geometry of perisynaptic astrocyte processes is variable and dynamically regulated. Studying these fine astrocyte processes represents a technical challenge, because many of them cannot be fully resolved by diffraction-limited microscopy. Therefore, we have established two indirect parameters of astrocyte morphology, which, while not fully resolving local geometry by design, provide statistical measures of astrocyte morphology: the fraction of tissue volume that astrocytes occupy and the density of resolvable astrocytic processes. Both are straightforward to obtain using widely available microscopy techniques. We here present the approach and demonstrate its robustness across various experimental conditions using mainly two-photon excitation fluorescence microscopy in acute slices and in vivo as well as modeling. Using these indirect measures allowed us to analyze the morphology of relatively large populations of astrocytes. Doing so we captured the heterogeneity of astrocytes within and between the layers of the hippocampal CA1 region and the developmental profile of astrocyte morphology. This demonstrates that volume fraction (VF) and segment density are useful parameters for describing the structure of astrocytes. They are also suitable for online monitoring of astrocyte morphology with widely available microscopy techniques.
Collapse
Affiliation(s)
- Daniel Minge
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Cátia Domingos
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Petr Unichenko
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Charlotte Behringer
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alberto Pauletti
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Stefanie Anders
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Michel K Herde
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andrea Delekate
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Polina Gulakova
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Susanne Schoch
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Division of Vascular Neurology, University Hospital Bonn, Bonn, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
81
|
Cao L, Ali S, Queen NJ. Hypothalamic gene transfer of BDNF promotes healthy aging. VITAMINS AND HORMONES 2021; 115:39-66. [PMID: 33706955 DOI: 10.1016/bs.vh.2020.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The aging process and age-related diseases all involve metabolic decline and impaired ability to cope with adversity. Environmental enrichment (EE)-a housing environment which recapitulates aspects of active lifestyle-exerts a wide range of health benefits in laboratory rodents. Brain-derived neurotrophic factor (BDNF) in the hypothalamus orchestrates autonomic and neuroendocrine processes, serving as one key brain mediator of EE-induced resistance to obesity, cancer, and autoimmunity. Recombinant adeno-associated virus (AAV)-mediated hypothalamic BDNF gene transfer alleviates obesity, diabetes, and metabolic syndromes in both diet-induced and genetic models. One recent study by our lab demonstrates the efficacy and safety of a built-in autoregulatory system to control transgene BDNF expression, mimicking the body's natural feedback systems in middle-age mice. Twelve-month old mice were treated with autoregulatory BDNF vector and monitored for 7months. BDNF gene transfer prevented age-associated metabolic decline by: reducing adiposity, preventing the decline of brown fat activity, increasing adiponectin while reducing leptin and insulin in circulation, improving glucose tolerance, increasing energy expenditure, alleviating hepatic steatosis, and suppressing inflammatory genes in the hypothalamus and adipose tissues. Furthermore, BDNF treatment reduced anxiety-like and depression-like behaviors. This chapter summarizes this work and discusses potential roles that hypothalamic BDNF might play in promoting healthy aging.
Collapse
Affiliation(s)
- Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States; The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.
| | - Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States; The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Nicholas J Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States; The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
82
|
Xie L, Hu Y, Yan D, McQuillan P, Liu Y, Zhu S, Zhu Z, Jiang Y, Hu Z. The relationship between exposure to general anesthetic agents and the risk of developing an impulse control disorder. Pharmacol Res 2021; 165:105440. [PMID: 33493656 DOI: 10.1016/j.phrs.2021.105440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022]
Abstract
Most studies examining the effect of extended exposure to general anesthetic agents (GAAs) have demonstrated that extended exposure induces both structural and functional changes in the central nervous system. These changes are frequently accompanied by neurobehavioral changes that include impulse control disorders that are generally characterized by deficits in behavioral inhibition and executive function. In this review, we will.
Collapse
Affiliation(s)
- Linghua Xie
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuhan Hu
- Department of Cell Biology, Yale University, New Haven, CT, USA
| | - Dandan Yan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - P McQuillan
- Department of Anesthesiology, Penn State Hershey Medical Centre, Penn State College of Medicine, Hershey, PA, USA
| | - Yue Liu
- Department of Anesthesiology, The Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhirui Zhu
- Department of Anesthesiology, The Children Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yilei Jiang
- Department of Anesthesiology, The Children Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
83
|
Charvériat M, Guiard BP. Serotonergic neurons in the treatment of mood disorders: The dialogue with astrocytes. PROGRESS IN BRAIN RESEARCH 2021; 259:197-228. [PMID: 33541677 DOI: 10.1016/bs.pbr.2021.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Astrocytes were traditionally regarded as cells important to neuronal activity, providing both metabolic and structural supports. Recent evidence suggests that they may also play a crucial role in the control of higher brain functions. In keeping with this hypothesis, it is now well accepted that astrocytes contribute to stress but also react to antidepressant drugs as they express serotonergic transporters and receptors. However, the downstream mechanisms leading to the fine-tuned regulation of mood are still unknown. This chapter pays attention to the role of astrocytes in the regulation of emotional behavior and related serotonergic neurotransmission. In particular, it gives a current state of the clinical and preclinical evidence showing that astrocytes respond to environmental conditions and antidepressant drugs through the release of gliotransmitters and neurotrophic factors which in turn, influence serotonergic tone in discrete brain areas. This state-of-the-art review aims at demonstrating the remarkable potential for novel therapeutic antidepressant strategies targeting these glial cells.
Collapse
Affiliation(s)
| | - Bruno P Guiard
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, Toulouse, France.
| |
Collapse
|
84
|
Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins in Adult Neurogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:95-115. [PMID: 34453295 DOI: 10.1007/978-3-030-74046-7_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell survival during adult neurogenesis and the modulation of each step, namely, proliferation, lineage differentiation, migration, maturation, and functional integration of the newborn cells into the existing circuitry, is regulated by intrinsic and extrinsic factors. Transduction of extracellular niche signals triggers the activation of intracellular mechanisms that regulate adult neurogenesis by affecting gene expression. While the intrinsic factors include transcription factors and epigenetic regulators, the extrinsic factors are molecular signals that are present in the neurogenic niche microenvironment. These include morphogens, growth factors, neurotransmitters, and signaling molecules secreted as soluble factors or associated to the extracellular matrix. Among these molecular mechanisms are neurotrophins and neurotrophin receptors which have been implicated in the regulation of adult neurogenesis at different levels, with brain-derived neurotrophic factor (BDNF) being the most studied neurotrophin. In this chapter, we review the current knowledge about the role of neurotrophins in the regulation of adult neurogenesis in both the subventricular zone (SVZ) and the hippocampal subgranular zone (SGZ).
Collapse
|
85
|
Yanpallewar S, Fulgenzi G, Tomassoni-Ardori F, Barrick C, Tessarollo L. Delayed onset of inherited ALS by deletion of the BDNF receptor TrkB.T1 is non-cell autonomous. Exp Neurol 2020; 337:113576. [PMID: 33359475 DOI: 10.1016/j.expneurol.2020.113576] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/04/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022]
Abstract
The pathophysiology of Amyotrophic Lateral Sclerosis (ALS), a disease caused by the gradual degeneration of motoneurons, is still largely unknown. Insufficient neurotrophic support has been cited as one of the causes of motoneuron cell death. Neurotrophic factors such as BDNF have been evaluated in ALS human clinical trials, but yielded disappointing results attributed to the poor pharmacokinetics and pharmacodynamics of BDNF. In the inherited ALS G93A SOD1 animal model, deletion of the BDNF receptor TrkB.T1 delays spinal cord motoneuron cell death and muscle weakness through an unknown cellular mechanism. Here we show that TrkB.T1 is expressed ubiquitously in the spinal cord and its deletion does not change the SOD1 mutant spinal cord inflammatory state suggesting that TrkB.T1 does not influence microglia or astrocyte activation. Although TrkB.T1 knockout in astrocytes preserves muscle strength and co-ordination at early stages of disease, its specific conditional deletion in motoneurons or astrocytes does not delay motoneuron cell death during the early stage of the disease. These data suggest that TrkB.T1 may limit the neuroprotective BDNF signaling to motoneurons via a non-cell autonomous mechanism providing new understanding into the reasons for past clinical failures and insights into the design of future clinical trials employing TrkB agonists in ALS.
Collapse
Affiliation(s)
| | - Gianluca Fulgenzi
- Neural Development Section, Mouse Cancer Genetics Program, CCR, NCI, NIH, USA
| | | | - Colleen Barrick
- Neural Development Section, Mouse Cancer Genetics Program, CCR, NCI, NIH, USA
| | - Lino Tessarollo
- Neural Development Section, Mouse Cancer Genetics Program, CCR, NCI, NIH, USA.
| |
Collapse
|
86
|
Kinboshi M, Ikeda A, Ohno Y. Role of Astrocytic Inwardly Rectifying Potassium (Kir) 4.1 Channels in Epileptogenesis. Front Neurol 2020; 11:626658. [PMID: 33424762 PMCID: PMC7786246 DOI: 10.3389/fneur.2020.626658] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/08/2020] [Indexed: 12/25/2022] Open
Abstract
Astrocytes regulate potassium and glutamate homeostasis via inwardly rectifying potassium (Kir) 4.1 channels in synapses, maintaining normal neural excitability. Numerous studies have shown that dysfunction of astrocytic Kir4.1 channels is involved in epileptogenesis in humans and animal models of epilepsy. Specifically, Kir4.1 channel inhibition by KCNJ10 gene mutation or expressional down-regulation increases the extracellular levels of potassium ions and glutamate in synapses and causes hyperexcitation of neurons. Moreover, recent investigations demonstrated that inhibition of Kir4.1 channels facilitates the expression of brain-derived neurotrophic factor (BDNF), an important modulator of epileptogenesis, in astrocytes. In this review, we summarize the current understanding on the role of astrocytic Kir4.1 channels in epileptogenesis, with a focus on functional and expressional changes in Kir4.1 channels and their regulation of BDNF secretion. We also discuss the potential of Kir4.1 channels as a therapeutic target for the prevention of epilepsy.
Collapse
Affiliation(s)
- Masato Kinboshi
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, Japan.,Department of Epilepsy, Movement Disorders and Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akio Ikeda
- Department of Epilepsy, Movement Disorders and Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukihiro Ohno
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, Takatsuki, Japan
| |
Collapse
|
87
|
Bruzelius A, Hidalgo I, Boza-Serrano A, Hjelmér AG, Tison A, Deierborg T, Bengzon J, Ramos-Moreno T. The human bone marrow harbors a CD45 - CD11B + cell progenitor permitting rapid microglia-like cell derivative approaches. Stem Cells Transl Med 2020; 10:582-597. [PMID: 33295698 PMCID: PMC7980218 DOI: 10.1002/sctm.20-0127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/23/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia, the immune sentinel of the central nervous system (CNS), are generated from yolk sac erythromyeloid progenitors that populate the developing CNS. Interestingly, a specific type of bone marrow-derived monocyte is able to express a yolk sac microglial signature and populate CNS in disease. Here we have examined human bone marrow (hBM) in an attempt to identify novel cell sources for generating microglia-like cells to use in cell-based therapies and in vitro modeling. We demonstrate that hBM stroma harbors a progenitor cell that we name stromal microglial progenitor (STR-MP). STR-MP single-cell gene analysis revealed the expression of the consensus genetic microglial signature and microglial-specific genes present in development and CNS pathologies. STR-MPs can be expanded and generate microglia-like cells in vitro, which we name stromal microglia (STR-M). STR-M cells show phagocytic ability, classically activate, and survive and phagocyte in human brain tissue. Thus, our results reveal that hBM harbors a source of microglia-like precursors that can be used in patient-centered fast derivative approaches.
Collapse
Affiliation(s)
- Andreas Bruzelius
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Department of Experimental Medical Science and Lund Stem Cell Center BMC, Lund University, Lund, Sweden
| | - Isabel Hidalgo
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Institution for Laboratory Medicine, Division of Molecular Hematology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Departamento de Bioquimica y Biologia Molecular, Facultad de Farmacia e Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain.,Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| | - Anna-Giorgia Hjelmér
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Amelie Tison
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| | - Johan Bengzon
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Tania Ramos-Moreno
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| |
Collapse
|
88
|
Qiao LY, Tiwari N. Spinal neuron-glia-immune interaction in cross-organ sensitization. Am J Physiol Gastrointest Liver Physiol 2020; 319:G748-G760. [PMID: 33084399 PMCID: PMC7792669 DOI: 10.1152/ajpgi.00323.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), historically considered as regional gastrointestinal disorders with heightened colonic sensitivity, are increasingly recognized to have concurrent dysfunction of other visceral and somatic organs, such as urinary bladder hyperactivity, leg pain, and skin hypersensitivity. The interorgan sensory cross talk is, at large, termed "cross-organ sensitization." These organs, anatomically distant from one another, physiologically interlock through projecting their sensory information into dorsal root ganglia (DRG) and then the spinal cord for integrative processing. The fundamental question of how sensitization of colonic afferent neurons conveys nociceptive information to activate primary afferents that innervate distant organs remains ambiguous. In DRG, primary afferent neurons are surrounded by satellite glial cells (SGCs) and macrophage accumulation in response to signals of injury to form a neuron-glia-macrophage triad. Astrocytes and microglia are major resident nonneuronal cells in the spinal cord to interact, physically and chemically, with sensory synapses. Cumulative evidence gathered so far indicate the indispensable roles of paracrine/autocrine interactions among neurons, glial cells, and immune cells in sensory cross-activation. Dichotomizing afferents, sensory convergency in the spinal cord, spinal nerve comingling, and extensive sprouting of central axons of primary afferents each has significant roles in the process of cross-organ sensitization; however, more results are required to explain their functional contributions. DRG that are located outside the blood-brain barrier and reside upstream in the cascade of sensory flow from one organ to the other in cross-organ sensitization could be safer therapeutic targets to produce less central adverse effects.
Collapse
Affiliation(s)
- Liya Y. Qiao
- 1Department of Physiology and Biophysics, Commonwealth University School of Medicine, Richmond, Virginia,2Department of Internal Medicine, Commonwealth University School of Medicine, Richmond, Virginia
| | - Namrata Tiwari
- 1Department of Physiology and Biophysics, Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
89
|
Tendilla-Beltrán H, Sanchez-Islas NDC, Marina-Ramos M, Leza JC, Flores G. The prefrontal cortex as a target for atypical antipsychotics in schizophrenia, lessons of neurodevelopmental animal models. Prog Neurobiol 2020; 199:101967. [PMID: 33271238 DOI: 10.1016/j.pneurobio.2020.101967] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Prefrontal cortex (PFC) inflammatory imbalance, oxidative/nitrosative stress (O/NS) and impaired neuroplasticity in schizophrenia are thought to have neurodevelopmental origins. Animal models are not only useful to test this hypothesis, they are also effective to establish a relationship among brain disturbances and behavior with the atypical antipsychotics (AAPs) effects. Here we review data of PFC post-mortem and in vivo neuroimaging, human induced pluripotent stem cells (hiPSC), and peripheral blood studies of inflammatory, O/NS, and neuroplasticity alterations in the disease as well as about their modulation by AAPs. Moreover, we reviewed the PFC alterations and the AAP mechanisms beyond their canonical antipsychotic action in four neurodevelopmental animal models relevant to the study of schizophrenia with a distinct approach in the generation of schizophrenia-like phenotypes, but all converge in O/NS and altered neuroplasticity in the PFC. These animal models not only reinforce the neurodevelopmental risk factor model of schizophrenia but also arouse some novel potential therapeutic targets for the disease including the reestablishment of the antioxidant response by the perineuronal nets (PNNs) and the nuclear factor erythroid 2-related factor (Nrf2) pathway, as well as the dendritic spine dynamics in the PFC pyramidal cells.
Collapse
Affiliation(s)
- Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico; Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico
| | | | - Mauricio Marina-Ramos
- Departamento de Ciencias de la Salud, Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM. Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital, 12 de Octubre (Imas12), Madrid, Spain
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
90
|
Shi J, Sun Y, Hua J. Functional Genetic Variation in the 3'-UTRNTRK2 is Associated with Risk of Ischemic Stroke. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:577-584. [PMID: 33209049 PMCID: PMC7669521 DOI: 10.2147/pgpm.s270319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/30/2020] [Indexed: 12/05/2022]
Abstract
Background Stroke is a leading cause of death and disability worldwide. It remains difficult to treat brain injury and improve functional rehabilitation after cerebral ischemia. Brain-derived neurotrophic factor (BDNF) is involved in ischemic stroke (IS) through interactions in the CREB1-BDNF-NTRk2 pathway. In this study, we aimed to determine the association of NTRK2 gene polymorphisms and the effects of intergenetic interactions in the Chinese population. Materials and Methods A total of 400 patients diagnosed with IS and 400 healthy controls were enrolled for genotyping. Detailed sequence-based analysis was predicted through bioinformatical investigation. Polymorphisms associated with miRNA were analyzed by a dual-luciferase reporter assay system. Results Analysis of clinical characteristics revealed that IS was highly associated with exposure to cigarette smoking, alcohol intake, as well as metabolic diseases, such as diabetes, hypertension, and higher serum triglyceride concentration. Three polymorphisms in NTRK2 located in the 3ʹ-untranslated region (3ʹ-UTR) were genotyped. Logistic regression analysis showed that IS patients with rs11140793, rs7047042, and rs1221 polymorphisms had a higher risk of stroke and indicated a worse short-term recovery. The mRNA level of NTRK2 was suppressed in a mutant genotype compared with wild genotype. The suppression of NTRK2 was induced by the gain-of-binding ability of certain miRNAs through the direct binding of 3ʹ-UTR. Conclusion Our research indicated that, by influencing the expression of NTRK2, the SNPs rs11140793, rs7047042, and rs1221 in the 3′UTR of NTRK2 can be used as risk factors for IS patients.
Collapse
Affiliation(s)
- Jiajia Shi
- Department of Rehabilitation Medicine, Kunshan Rehabilitation Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Ying Sun
- Department of Rehabilitation Medicine, Kunshan Rehabilitation Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Jiajia Hua
- Department of Rehabilitation Medicine, The Sixth People's Hospital of Nantong, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
91
|
Muñoz-Ballester C, Umans RA, Robel S. Leveraging Zebrafish To Study Bona Fide Astrocytes. Trends Neurosci 2020; 44:77-79. [PMID: 33213859 DOI: 10.1016/j.tins.2020.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 11/17/2022]
Abstract
In a recent study, Chen and colleagues demonstrated that zebrafish spinal cord radial glia differentiate into cells that are similar to mammalian astrocytes. This study highlights the validity of the zebrafish model for discovering molecular mechanisms governing astrocyte function.
Collapse
Affiliation(s)
- Carmen Muñoz-Ballester
- Center for Glial Biology in Health, Disease, and Cancer, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
| | - Robyn A Umans
- Center for Glial Biology in Health, Disease, and Cancer, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
| | - Stefanie Robel
- Center for Glial Biology in Health, Disease, and Cancer, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA; School of Neuroscience, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
92
|
Miranda-Lourenço C, Ribeiro-Rodrigues L, Fonseca-Gomes J, Tanqueiro SR, Belo RF, Ferreira CB, Rei N, Ferreira-Manso M, de Almeida-Borlido C, Costa-Coelho T, Freitas CF, Zavalko S, Mouro FM, Sebastião AM, Xapelli S, Rodrigues TM, Diógenes MJ. Challenges of BDNF-based therapies: From common to rare diseases. Pharmacol Res 2020; 162:105281. [PMID: 33161136 DOI: 10.1016/j.phrs.2020.105281] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022]
Abstract
Neurotrophins are a well-known family of neurotrophic factors that play an important role both in the central and peripheral nervous systems, where they modulate neuronal survival, development, function and plasticity. Brain-derived neurotrophic factor (BDNF) possesses diverse biological functions which are mediated by the activation of two main classes of receptors, the tropomyosin-related kinase (Trk) B and the p75 neurotrophin receptor (p75NTR). The therapeutic potential of BDNF has drawn attention since dysregulation of its signalling cascades has been suggested to underlie the pathogenesis of both common and rare diseases. Multiple strategies targeting this neurotrophin have been tested; most have found obstacles that ultimately hampered their effectiveness. This review focuses on the involvement of BDNF and its receptors in the pathophysiology of Alzheimer's disease (AD), Amyotrophic Lateral Sclerosis (ALS) and Rett Syndrome (RTT). We describe the known mechanisms leading to the impairment of BDNF/TrkB signalling in these disorders. Such mechanistic insight highlights how BDNF signalling compromise can take various shapes, nearly disease-specific. Therefore, BDNF-based therapeutic strategies must be specifically tailored and are more likely to succeed if a combination of resources is employed.
Collapse
Affiliation(s)
- Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - João Fonseca-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Rita F Belo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Catarina B Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mafalda Ferreira-Manso
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Carolina de Almeida-Borlido
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Tiago Costa-Coelho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Céline Felicidade Freitas
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Svitlana Zavalko
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Tiago M Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Institute of Molecular and Clinical Ophthalmology Basel (IOB), Mittlere Strasse 91, 4031 Basel, Switzerland
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
93
|
Iwata K, Wu Q, Ferdousi F, Sasaki K, Tominaga K, Uchida H, Arai Y, Szele FG, Isoda H. Sugarcane ( Saccharum officinarum L.) Top Extract Ameliorates Cognitive Decline in Senescence Model SAMP8 Mice: Modulation of Neural Development and Energy Metabolism. Front Cell Dev Biol 2020; 8:573487. [PMID: 33123536 PMCID: PMC7573230 DOI: 10.3389/fcell.2020.573487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related biological alterations in brain function increase the risk of mild cognitive impairment and dementia, a global problem exacerbated by aging populations in developed nations. Limited pharmacological therapies have resulted in attention turning to the promising role of medicinal plants and dietary supplements in the treatment and prevention of dementia. Sugarcane (Saccharum officinarum L.) top, largely considered as a by-product because of its low sugar content, in fact contains the most abundant amounts of antioxidant polyphenols relative to the rest of the plant. Given the numerous epidemiological studies on the effects of polyphenols on cognitive function, in this study, we analyzed polyphenolic constituents of sugarcane top and examined the effect of sugarcane top ethanolic extract (STEE) on a range of central nervous system functions in vitro and in vivo. Orally administrated STEE rescued spatial learning and memory deficit in the senescence-accelerated mouse prone 8 (SAMP8) mice, a non-transgenic strain that spontaneously develops a multisystemic aging phenotype including pathological features of Alzheimer's disease. This could be correlated with an increased number of hippocampal newborn neurons and restoration of cortical monoamine levels in STEE-fed SAMP8 mice. Global genomic analysis by microarray in cerebral cortices showed multiple potential mechanisms for the cognitive improvement. Gene set enrichment analysis (GSEA) revealed biological processes such as neurogenesis, neuron differentiation, and neuron development were significantly enriched in STEE-fed mice brain compared to non-treated SAMP8 mice. Furthermore, STEE treatment significantly regulated genes involved in neurotrophin signaling, glucose metabolism, and neural development in mice brain. Our in vitro results suggest that STEE treatment enhances the metabolic activity of neuronal cells promoting glucose metabolism with significant upregulation of genes, namely PGK1, PGAM1, PKM, and PC. STEE also stimulated proliferation of human neural stem cells (hNSCs), regulated bHLH factor expression and induced neuronal differentiation and astrocytic process lengthening. Altogether, our findings suggest the potential of STEE as a dietary intervention, with promising implications as a novel nutraceutical for cognitive health.
Collapse
Affiliation(s)
- Kengo Iwata
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan.,Nippo Co., Ltd., Daito, Japan
| | - Qingqing Wu
- Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Kazunori Sasaki
- Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | - Kenichi Tominaga
- AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan
| | | | | | - Francis G Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Hiroko Isoda
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan.,Alliance for Research on the Mediterranean and North Africa, University of Tsukuba, Tsukuba, Japan.,AIST-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), AIST, University of Tsukuba, Tsukuba, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
94
|
Saba J, López Couselo F, Turati J, Carniglia L, Durand D, de Laurentiis A, Lasaga M, Caruso C. Astrocytes from cortex and striatum show differential responses to mitochondrial toxin and BDNF: implications for protection of striatal neurons expressing mutant huntingtin. J Neuroinflammation 2020; 17:290. [PMID: 33023623 PMCID: PMC7542133 DOI: 10.1186/s12974-020-01965-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Background Evidence shows significant heterogeneity in astrocyte gene expression and function. We previously demonstrated that brain-derived neurotrophic factor (BDNF) exerts protective effects on whole brain primary cultured rat astrocytes treated with 3-nitropropionic acid (3NP), a mitochondrial toxin widely used as an in vitro model of Huntington’s disease (HD). Therefore, we now investigated 3NP and BDNF effects on astrocytes from two areas involved in HD: the striatum and the entire cortex, and their involvement in neuron survival. Methods We prepared primary cultured rat cortical or striatal astrocytes and treated them with BDNF and/or 3NP for 24 h. In these cells, we assessed expression of astrocyte markers, BDNF receptor, and glutamate transporters, and cytokine release. We prepared astrocyte-conditioned medium (ACM) from cortical and striatal astrocytes and tested its effect on a cellular model of HD. Results BDNF protected astrocytes from 3NP-induced death, increased expression of its own receptor, and activation of ERK in both cortical and striatal astrocytes. However, BDNF modulated glutamate transporter expression differently by increasing GLT1 and GLAST expression in cortical astrocytes but only GLT1 expression in striatal astrocytes. Striatal astrocytes released higher amounts of tumor necrosis factor-α than cortical astrocytes in response to 3NP but BDNF decreased this effect in both populations. 3NP decreased transforming growth factor-β release only in cortical astrocytes, whereas BDNF treatment increased its release only in striatal astrocytes. Finally, we evaluated ACM effect on a cellular model of HD: the rat striatal neuron cell line ST14A expressing mutant human huntingtin (Q120) or in ST14A cells expressing normal human huntingtin (Q15). Neither striatal nor cortical ACM modified the viability of Q15 cells. Only ACM from striatal astrocytes treated with BDNF and ACM from 3NP + BDNF-treated striatal astrocytes protected Q120 cells, whereas ACM from cortical astrocytes did not. Conclusions Data suggest that cortical and striatal astrocytes respond differently to mitochondrial toxin 3NP and BDNF. Moreover, striatal astrocytes secrete soluble neuroprotective factors in response to BDNF that selectively protect neurons expressing mutant huntingtin implicating that BDNF modulation of striatal astrocyte function has therapeutic potential against neurodegeneration. Graphical abstract ![]()
Collapse
Affiliation(s)
- Julieta Saba
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico López Couselo
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Turati
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lila Carniglia
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andrea de Laurentiis
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO). UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas (INBIOMED), UBA-CONICET, Paraguay 2155, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
95
|
Di Consiglio E, Pistollato F, Mendoza-De Gyves E, Bal-Price A, Testai E. Integrating biokinetics and in vitro studies to evaluate developmental neurotoxicity induced by chlorpyrifos in human iPSC-derived neural stem cells undergoing differentiation towards neuronal and glial cells. Reprod Toxicol 2020; 98:174-188. [PMID: 33011216 PMCID: PMC7772889 DOI: 10.1016/j.reprotox.2020.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Human iPSC-derived NSCs undergoing differentiation possess some metabolic competence. CPF entered the cells and was biotrasformed into its two main metabolites (CPFO and TCP). After repeated exposure, very limited bioaccumulation of CPF was observed. Treatment with CPF decreased neurite outgrowth, synapse number and electrical activity. Treatment with CPF increased BDNF levels and the percentage of astrocytes.
For some complex toxicological endpoints, chemical safety assessment has conventionally relied on animal testing. Apart from the ethical issues, also scientific considerations have been raised concerning the traditional approach, highlighting the importance for considering real life exposure scenario. Implementation of flexible testing strategies, integrating multiple sources of information, including in vitro reliable test methods and in vitro biokinetics, would enhance the relevance of the obtained results. Such an approach could be pivotal in the evaluation of developmental neurotoxicity (DNT), especially when applied to human cell-based models, mimicking key neurodevelopmental processes, relevant to human brain development. Here, we integrated the kinetic behaviour with the toxicodynamic alterations of chlorpyrifos (CPF), such as in vitro endpoints specific for DNT evaluation, after repeated exposure during differentiation of human neural stem cells into a mixed culture of neurons and astrocytes. The upregulation of some cytochrome P450 and glutathione S-transferase genes during neuronal differentiation and the formation of the two major CPF metabolites (due to bioactivation and detoxification) supported the metabolic competence of the used in vitro model. The alterations in the number of synapses, neurite outgrowth, brain derived neurotrophic factor, the proportion of neurons and astrocytes, as well as spontaneous electrical activity correlated well with the CPF ability to enter the cells and be bioactivated to CPF-oxon. Overall, our results confirm that combining in vitro biokinetics and assays to evaluate effects on neurodevelopmental endpoints in human cells should be regarded as a key strategy for a quantitative characterization of DNT effects.
Collapse
Affiliation(s)
- Emma Di Consiglio
- Istituto Superiore di Sanità, Environment and Health Department, Mechanisms, Biomarkers and Models Unit, Rome, Italy
| | | | | | - Anna Bal-Price
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Emanuela Testai
- Istituto Superiore di Sanità, Environment and Health Department, Mechanisms, Biomarkers and Models Unit, Rome, Italy
| |
Collapse
|
96
|
Charkviani M, Muradashvili N, Sulimai N, Lominadze D. Fibrinogen-cellular prion protein complex formation on astrocytes. J Neurophysiol 2020; 124:536-543. [PMID: 32697670 DOI: 10.1152/jn.00224.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most common neurological disorders causing memory reduction, particularly short-term memory (STM). We showed that, during TBI-induced inflammation, increased blood content of fibrinogen (Fg) enhanced vascular protein transcytosis and deposition of extravasated Fg in vasculo-astrocyte interfaces. In addition, we found that deposition of cellular prion protein (PrPC) was also increased in the vasculo-astrocyte endfeet interface. However, association of Fg and PrPC was not confirmed. Presently, we aimed to define whether Fg can associate with PrPC on astrocytes and cause their activation. Cultured mouse brain astrocytes were treated with medium alone (control), Fg (2 mg/mL or 4 mg/mL), 4 mg/mL of Fg in the presence of a function-blocking anti-PrPC peptide or anti-mouse IgG, function-blocking anti-PrPC peptide, or anti-mouse IgG alone. After treatment, either cell lysates were collected and analyzed via Western blot or coimmunoprecipitation was performed, or astrocytes were fixed and their activation was assessed with immunohistochemistry. Results showed that Fg dose-dependently activated astrocytes, increased expressions of PrPC and tyrosine (tropomyosin) receptor kinase B (TrkB), and PrP gene. Blocking the function of PrPC reduced these effects. Coimmunoprecipitation demonstrated Fg and PrPC association. Since it is known that prion protein has a greater effect on memory reduction than amyloid beta, and that activation of TrkB is involved in neurodegeneration, our findings confirming the possible formation of Fg-PrPC and Fg-induced overexpression of TrkB on astrocytes suggest a possible triggering mechanism for STM reduction that was seen previously during mild-to-moderate TBI.NEW & NOTEWORTHY For the first time we showed that fibrinogen (Fg) can associate with cellular prion protein (PrPC) on the surface of cultured mouse brain astrocytes. At high levels, Fg causes upregulation of astrocyte PrPC and astrocyte activation accompanied with overexpression of tyrosine receptor kinase B (TrkB), which results in nitric oxide (NO) production and generation of reactive oxygen species (ROS). Fg/PrPC interaction can be a triggering mechanism for TrkB-NO-ROS axis activation and the resultant astrocyte-mediated neurodegeneration.
Collapse
Affiliation(s)
- Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, Kentucky
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, Kentucky.,Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - Nurul Sulimai
- Department of Surgery, USF Health-Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, Kentucky.,Department of Surgery, USF Health-Morsani College of Medicine, University of South Florida, Tampa, Florida.,Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, Kentucky
| |
Collapse
|
97
|
Fernández-García S, Sancho-Balsells A, Longueville S, Hervé D, Gruart A, Delgado-García JM, Alberch J, Giralt A. Astrocytic BDNF and TrkB regulate severity and neuronal activity in mouse models of temporal lobe epilepsy. Cell Death Dis 2020; 11:411. [PMID: 32483154 PMCID: PMC7264221 DOI: 10.1038/s41419-020-2615-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
Astrocytes have emerged as crucial regulators of neuronal network activity, synapse formation, and underlying behavioral and cognitive processes. Despite some pathways have been identified, the communication between astrocytes and neurons remains to be completely elucidated. Unraveling this communication is crucial to design potential treatments for neurological disorders like temporal lobe epilepsy (TLE). The BDNF and TrkB molecules have emerged as very promising therapeutic targets. However, their modulation can be accompanied by several off-target effects such as excitotoxicity in case of uncontrolled upregulation or dementia, amnesia, and other memory disorders in case of downregulation. Here, we show that BDNF and TrkB from astrocytes modulate neuronal dysfunction in TLE models. First, conditional overexpression of BDNF from astrocytes worsened the phenotype in the lithium-pilocarpine mouse model. Our evidences pointed out to the astrocytic pro-BDNF isoform as a major player of this altered phenotype. Conversely, specific genetic deletion of BDNF in astrocytes prevented the increase in the number of firing neurons and the global firing rate in an in vitro model of TLE. Regarding to the TrkB, we generated mice with a genetic deletion of TrkB specifically in hippocampal neurons or astrocytes. Interestingly, both lines displayed neuroprotection in the lithium-pilocarpine model but only the mice with genetic deletion of TrkB in astrocytes showed significantly preserved spatial learning skills. These data identify the astrocytic BDNF and TrkB molecules as promising therapeutic targets for the treatment of TLE.
Collapse
Affiliation(s)
- Sara Fernández-García
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Sophie Longueville
- Inserm UMR-S 1270, 75005, Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005, Paris, France.,Institut du Fer a Moulin, 75005, Paris, France
| | - Denis Hervé
- Inserm UMR-S 1270, 75005, Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005, Paris, France.,Institut du Fer a Moulin, 75005, Paris, France
| | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, 41013, Seville, Spain
| | | | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036, Barcelona, Spain
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, 08036, Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain. .,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036, Barcelona, Spain.
| |
Collapse
|
98
|
Cao T, Matyas JJ, Renn CL, Faden AI, Dorsey SG, Wu J. Function and Mechanisms of Truncated BDNF Receptor TrkB.T1 in Neuropathic Pain. Cells 2020; 9:cells9051194. [PMID: 32403409 PMCID: PMC7290366 DOI: 10.3390/cells9051194] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/11/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a major focus for regenerative therapeutics, has been lauded for its pro-survival characteristics and involvement in both development and recovery of function within the central nervous system (CNS). However, studies of tyrosine receptor kinase B (TrkB), a major receptor for BDNF, indicate that certain effects of the TrkB receptor in response to disease or injury may be maladaptive. More specifically, imbalance among TrkB receptor isoforms appears to contribute to aberrant signaling and hyperpathic pain. A truncated isoform of the receptor, TrkB.T1, lacks the intracellular kinase domain of the full length receptor and is up-regulated in multiple CNS injury models. Such up-regulation is associated with hyperpathic pain, and TrkB.T1 inhibition reduces neuropathic pain in various experimental paradigms. Deletion of TrkB.T1 also limits astrocyte changes in vitro, including proliferation, migration, and activation. Mechanistically, TrkB.T1 is believed to act through release of intracellular calcium in astrocytes, as well as through interactions with neurotrophins, leading to cell cycle activation. Together, these studies support a potential role for astrocytic TrkB.T1 in hyperpathic pain and suggest that targeted strategies directed at this receptor may have therapeutic potential.
Collapse
Affiliation(s)
- Tuoxin Cao
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
| | - Jessica J. Matyas
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
| | - Cynthia L. Renn
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD 21201, USA; (C.L.R.); (S.G.D.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
| | - Alan I. Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
| | - Susan G. Dorsey
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD 21201, USA; (C.L.R.); (S.G.D.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; (T.C.); (J.J.M.); (A.I.F.)
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-410-706-5189
| |
Collapse
|
99
|
Xie W, Xiang L, Song Y, Tian X. The Downregulation of Truncated TrkB Receptors Modulated by MicroRNA-185 Activates Full-Length TrkB Signaling and Suppresses the Epileptiform Discharges in Cultured Hippocampal Neurons. Neurochem Res 2020; 45:1647-1660. [DOI: 10.1007/s11064-020-03013-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 11/30/2022]
|
100
|
Abstract
Astrocytes are the most abundant cell type in the central nervous system and have diverse functions in blood–brain barrier maintenance, neural circuitry formation and function, and metabolic regulation. To better understand the diverse roles of astrocytes, we will summarize what is known about astrocyte development and the challenges limiting our understanding of this process. We will also discuss new approaches and technologies advancing the field.
Collapse
Affiliation(s)
- Ekin Su Akdemir
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna Yu-Szu Huang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|