51
|
Sulimai N, Lominadze D. Fibrinogen and Neuroinflammation During Traumatic Brain Injury. Mol Neurobiol 2020; 57:4692-4703. [PMID: 32776201 DOI: 10.1007/s12035-020-02012-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Many neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis, and traumatic brain injury (TBI) are associated with systemic inflammation. Inflammation itself results in increased blood content of fibrinogen (Fg), called hyperfibrinogenemia (HFg). Fg is not only considered an acute phase protein and a marker of inflammation, but has been shown that it can cause inflammatory responses. Fibrin deposits have been associated with memory reduction in neuroinflammatory diseases such as AD and TBI. Reduction in short-term memory has been seen during the most common form of TBI, mild-to-moderate TBI. Fibrin deposits have been found in brains of patients with mild-to-moderate TBI. The vast majority of the literature emphasizes the role of fibrin-activated microglia as the mediator in the neuroinflammation pathway. However, the recent discovery that astrocytes, which constitute approximately 30% of the cells in the mammalian central nervous system, manifest different reactive states warrants further investigations in the causative role of HFg in astrocyte-mediated neuroinflammation. Our previous study showed that Fg deposited in the vasculo-astrocyte interface-activated astrocytes. However, little is known of how Fg directly affects astrocytes and neurons. In this review, we summarize studies that show the effect of Fg on different types of cells in the vasculo-neuronal unit. We will also discuss the possible mechanism of HFg-induced neuroinflammation during TBI.
Collapse
Affiliation(s)
- Nurul Sulimai
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - David Lominadze
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
- Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
52
|
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: 1.2] [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
|
53
|
Colombo E, Bassani C, De Angelis A, Ruffini F, Ottoboni L, Comi G, Martino G, Farina C. Siponimod (BAF312) Activates Nrf2 While Hampering NFκB in Human Astrocytes, and Protects From Astrocyte-Induced Neurodegeneration. Front Immunol 2020; 11:635. [PMID: 32322257 PMCID: PMC7156595 DOI: 10.3389/fimmu.2020.00635] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/19/2020] [Indexed: 01/12/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory neurodegenerative disease of the central nervous system (CNS) with heterogeneous pathophysiology. In its progressive course oligodendrocyte and neuroaxonal damage is sustained by compartmentalized inflammation due to glial dysregulation. Siponimod (BAF312), a modulator of two sphingosine-1-phosphate (S1P) receptors (S1P1 and S1P5) is the first oral treatment specifically approved for active secondary progressive MS. To address potential direct effects of BAF312 on glial function and glia-neuron interaction, we set up a series of in vitro functional assays with astrocytes generated from human fibroblasts. These cells displayed the typical morphology and markers of astroglia, and were susceptible to the action of inflammatory mediators and BAF312, because expressing receptors for IL1, IL17, and S1P (namely S1P1 and S1P3). Targeting of S1P signaling by BAF312 inhibited NFκB translocation evoked by inflammatory cytokines, indicating a direct anti-inflammatory activity of the drug on the human astrocyte. Further, while glia cells exposed to IL1 or IL17 downregulated protein expression of glutamate transporters, BAF312-treated astrocytes maintained high levels of GLAST and GLT1 regardless of the presence of inflammatory mediators. Interestingly, despite potential glial susceptibility to S1P signaling via S1P3, which is not targeted by BAF312, NFκB translocation and downregulation of glutamate transporters in response to S1P were inhibited at similar levels by BAF312 and FTY720, another S1P signaling modulator targeting also S1P3. Accordingly, specific inhibition of S1P1 via NIBR-0213 blocked S1P-evoked NFκB translocation, demonstrating that modulation of S1P1 is sufficient to dampen signaling via other S1P receptors. Considering that NFκB-dependent responses are regulated by Nrf2, we measured activation of this critical transcription factor for anti-oxidant reactions, and observed that BAF312 rapidly induced nuclear translocation of Nrf2, but this effect was attenuated in the presence of an inflammatory milieu. Finally, in vitro experiments with spinal neurons exposed to astrocyte-conditioned media showed that modulation of S1P or cytokine signaling in astrocytes via BAF312 prevented neurons from astrocyte-induced degeneration. Overall, these experiments on human astrocytes suggest that during neuroinflammation targeting of S1P1 via BAF312 may modulate key astrocyte functions and thereby attain neuroprotection indirectly.
Collapse
Affiliation(s)
- Emanuela Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Bassani
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Anthea De Angelis
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ruffini
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Linda Ottoboni
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Giancarlo Comi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
54
|
Zhu X, Sun-Waterhouse D, Tao Q, Li W, Shu D, Cui C. The enhanced serotonin (5-HT) synthesis and anti-oxidative roles of Trp oligopeptide in combating anxious depression C57BL/6 mice. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
55
|
Charkviani M, Muradashvili N, Lominadze D. Vascular and non-vascular contributors to memory reduction during traumatic brain injury. Eur J Neurosci 2019; 50:2860-2876. [PMID: 30793398 PMCID: PMC6703968 DOI: 10.1111/ejn.14390] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is an increasing health problem. It is a complex, progressive disease that consists of many factors affecting memory. Studies have shown that increased blood-brain barrier (BBB) permeability initiates pathological changes in neuro-vascular network but the role of cerebrovascular dysfunction and its mediated mechanisms associated with memory reduction during TBI are still not well understood. Changes in BBB, inflammation, extravasation of blood plasma components, activation of neuroglia lead to neurodegeneration. Extravasated proteins such as amyloid-beta, fibrinogen, and cellular prion protein may form degradation resistant complexes that can lead to neuronal dysfunction and degeneration. They also have the ability to activate astrocytes, and thus, can be involved in memory impairment. Understanding the triggering mechanisms and the places they originate in vasculature or in extravascular tissue may help to identify potential therapeutic targets to ameliorate memory reduction during TBI. The goal of this review is to discuss conceptual mechanisms that lead to short-term memory reduction during non-severe TBI considering distinction between vascular and non-vascular effects on neurons. Some aspects of these mechanisms need to be confirmed further. Therefore, we hope that the discussion presented bellow may lead to experiments that may clarify the triggering mechanisms of memory reduction after head trauma.
Collapse
Affiliation(s)
- Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, KY, USA
| |
Collapse
|
56
|
Halpern M, Brennand KJ, Gregory J. Examining the relationship between astrocyte dysfunction and neurodegeneration in ALS using hiPSCs. Neurobiol Dis 2019; 132:104562. [PMID: 31381978 DOI: 10.1016/j.nbd.2019.104562] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/28/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex and fatal neurodegenerative disease for which the causes of disease onset and progression remain unclear. Recent advances in human induced pluripotent stem cell (hiPSC)-based models permit the study of the genetic factors associated with ALS in patient-derived neural cell types, including motor neurons and glia. While astrocyte dysfunction has traditionally been thought to exacerbate disease progression, astrocytic dysfunction may play a more direct role in disease initiation and progression. Such non-cell autonomous mechanisms expand the potential targets of therapeutic intervention, but only a handful of ALS risk-associated genes have been examined for their impact on astrocyte dysfunction and neurodegeneration. This review summarizes what is currently known about astrocyte function in ALS and suggests ways in which hiPSC-based models can be used to more effectively study the role of astrocytes in neurodegenerative disease.
Collapse
Affiliation(s)
- Madeline Halpern
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| | - James Gregory
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, United States of America.
| |
Collapse
|
57
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
58
|
Ulivieri C, De Tommaso D, Finetti F, Ortensi B, Pelicci G, D'Elios MM, Ballerini C, Baldari CT. A T Cell Suppressive Circuitry Mediated by CD39 and Regulated by ShcC/Rai Is Induced in Astrocytes by Encephalitogenic T Cells. Front Immunol 2019; 10:1041. [PMID: 31134091 PMCID: PMC6524536 DOI: 10.3389/fimmu.2019.01041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease caused by autoreactive immune cell infiltration into the central nervous system leading to inflammation, demyelination, and neuronal loss. While myelin-reactive Th1 and Th17 are centrally implicated in multiple sclerosis pathogenesis, the local CNS microenvironment, which is shaped by both infiltrated immune cells and central nervous system resident cells, has emerged a key player in disease onset and progression. We have recently demonstrated that ShcC/Rai is as a novel astrocytic adaptor whose loss in mice protects from experimental autoimmune encephalomyelitis. Here, we have explored the mechanisms that underlie the ability of Rai-/- astrocytes to antagonize T cell-dependent neuroinflammation. We show that Rai deficiency enhances the ability of astrocytes to upregulate the expression and activity of the ectonucleotidase CD39, which catalyzes the conversion of extracellular ATP to the immunosuppressive metabolite adenosine, through both contact-dependent and-independent mechanisms. As a result, Rai-deficient astrocytes acquire an enhanced ability to suppress T-cell proliferation, which involves suppression of T cell receptor signaling and upregulation of the inhibitory receptor CTLA-4. Additionally, Rai-deficient astrocytes preferentially polarize to the neuroprotective A2 phenotype. These results identify a new mechanism, to which Rai contributes to a major extent, by which astrocytes modulate the pathogenic potential of autoreactive T cells.
Collapse
Affiliation(s)
| | | | | | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | |
Collapse
|
59
|
The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol 2019; 137:757-783. [PMID: 30847559 DOI: 10.1007/s00401-019-01980-7] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation is the coordinated response of the central nervous system (CNS) to threats to its integrity posed by a variety of conditions, including autoimmunity, pathogens and trauma. Activated astrocytes, in concert with other cellular elements of the CNS and immune system, are important players in the modulation of the neuroinflammatory response. During neurological disease, they produce and respond to cellular signals that often lead to dichotomous processes, which can promote further damage or contribute to repair. This occurs also in multiple sclerosis (MS), where astrocytes are now recognized as key components of its immunopathology. Evidence supporting this role has emerged not only from studies in MS patients, but also from animal models, among which the experimental autoimmune encephalomyelitis (EAE) model has proved especially instrumental. Based on this premise, the purpose of the present review is to summarize the current knowledge of astrocyte behavior in MS and EAE. Following a brief description of the pathological characteristics of the two diseases and the main functional roles of astrocytes in CNS physiology, we will delve into the specific responses of this cell population, analyzing MS and EAE in parallel. We will define the temporal and anatomical profile of astroglial activation, then focus on key processes they participate in. These include: (1) production and response to soluble mediators (e.g., cytokines and chemokines), (2) regulation of oxidative stress, and (3) maintenance of BBB integrity and function. Finally, we will review the state of the art on the available methods to measure astroglial activation in vivo in MS patients, and how this could be exploited to optimize diagnosis, prognosis and treatment decisions. Ultimately, we believe that integrating the knowledge obtained from studies in MS and EAE may help not only better understand the pathophysiology of MS, but also uncover new signals to be targeted for therapeutic intervention.
Collapse
|
60
|
Fletcher JL, Murray SS, Xiao J. Brain-Derived Neurotrophic Factor in Central Nervous System Myelination: A New Mechanism to Promote Myelin Plasticity and Repair. Int J Mol Sci 2018; 19:ijms19124131. [PMID: 30572673 PMCID: PMC6321406 DOI: 10.3390/ijms19124131] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/27/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays vitally important roles in neural development and plasticity in both health and disease. Recent studies using mutant mice to selectively manipulate BDNF signalling in desired cell types, in combination with animal models of demyelinating disease, have demonstrated that BDNF not only potentiates normal central nervous system myelination in development but enhances recovery after myelin injury. However, the precise mechanisms by which BDNF enhances myelination in development and repair are unclear. Here, we review some of the recent progress made in understanding the influence BDNF exerts upon the myelinating process during development and after injury, and discuss the cellular and molecular mechanisms underlying its effects. In doing so, we raise new questions for future research.
Collapse
Affiliation(s)
- Jessica L Fletcher
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, 3010, VIC, Australia.
| | - Simon S Murray
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, 3010, VIC, Australia.
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, 3010, VIC, Australia.
| |
Collapse
|
61
|
Vahidi-Ferdowsi P, Mehrzad J, Malvandi A, Hosseinkhani S. Bioluminescence-based detection of astrocytes apoptosis and ATP depletion induced by biologically relevant level aflatoxin B1. WORLD MYCOTOXIN J 2018. [DOI: 10.3920/wmj2017.2275] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although brain accumulation of aflatoxin B1 (AFB1) suggests potential impact on brain cells, including astrocytes, there still exists a scarcity of research on this issue within the literature. This research investigates the apoptosis effect of AFB1 on primary mouse astrocytes. To this aim, a MTT colorimetric assay on astrocytes was performed to measure the toxicity/LC50 of various concentrations (0-320,000 nM) of AFB1 for 24 h. Further, the astrocytes were exposed to concentrations of 8, 16 and 32 nM of AFB1 for 24, 48 and 72 h. Concentration of intracellular ATP) and caspase-3/7 activity was then determined by luciferase-dependent bioluminescence. Furthermore, the percentage of apoptotic cells was obtained using flow cytometry (annexin V+/propidium iodide (PI)−; cytochrome c release from mitochondria, a hallmark of cell damage, was carried out by Western blot as well. MTT assay at post-exposure hours (PEH) 24 revealed that the LC50 of AFB1 was ~80,000 nM. Though at PEH 48 only 32 nM of AFB1 resulted in a significant diminished intracellular ATP content, at PEH 72 both 8 and 32 nM of AFB1 led to a significant ATP depletion in astrocytes. Similar patterns of changes were observed in bioluminescence intensity of AFB1-treated astrocytes. Flow cytometry-based annexin V and PI staining of astrocytes at PEH 24, 48 and 72 showed that 32 nM of AFB1 significantly and time dependently increased the percentage of apoptotic astrocytes (annexin V+/PI−). With 32 nM of AFB1, caspase-3/7 activity in astrocytes was increased ~4-fold at PEH 72. A remarkable release of cytochrome c was only detected in astrocytes exposed to 32 nM AFB1 for PEH 72. The results indicated that a biologically relevant level of AFB1 (32 nM) induces apoptosis in astrocytes through ATP depletion and caspases activation.
Collapse
Affiliation(s)
- P. Vahidi-Ferdowsi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, 14115-111 Tehran, Iran
| | - J. Mehrzad
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, 1419963111 Tehran, Iran
| | - A.M. Malvandi
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Giovanni Battista Grassi 74, 20157 Milan, Italy
| | - S. Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, 14115-111 Tehran, Iran
| |
Collapse
|
62
|
Clark VD, Layson A, Charkviani M, Muradashvili N, Lominadze D. Hyperfibrinogenemia-mediated astrocyte activation. Brain Res 2018; 1699:158-165. [PMID: 30153459 DOI: 10.1016/j.brainres.2018.08.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/10/2018] [Accepted: 08/22/2018] [Indexed: 12/26/2022]
Abstract
Fibrinogen (Fg)-containing plaques are associated with memory loss during various inflammatory neurodegenerative diseases such as Alzheimer's disease, multiple sclerosis, stroke, and traumatic brain injury. However, mechanisms of its action in neurovascular unit are not clear. As Fg is a high molecular weight blood protein and cannot translocate far from the vessel after extravasation, we hypothesized that it may interact with astrocytes first causing their activation. Cultured mouse cortical astrocytes were treated with Fg in the presence or absence of function-blocking anti-mouse intercellular adhesion molecule 1 (ICAM-1) antibody, or with medium alone (control). Expressions of ICAM-1 and tyrosine receptor kinase B (TrkB) as markers of astrocyte activation, and phosphorylation of TrkB (pTrkB) were assessed. Fg dose-dependently increased activation of astrocytes defined by their shape change, retraction of processes, and enhanced expressions of ICAM-1 and TrkB, and increased pTrkB. Blocking of ICAM-1 function ameliorated these Fg effects. Data suggest that Fg interacts with astrocytes causing overexpression of ICAM-1 and TrkB, and TrkB phosphorylation, and thus, astrocyte activation. Since TrkB is known to be involved in neurodegeneration, interaction of Fg with astrocytes and the resultant activation of TrkB can be a possible mechanism involved in memory reduction, which were observed in previous studies and were associated with formation of complexes of Fg deposited in extravascular space with proteins such as Amyloid beta or prion, the proteins involved in development of dementia.
Collapse
Affiliation(s)
- Vincent D Clark
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Ailey Layson
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA; Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA; Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
63
|
Quetiapine prevents Aβ25-35-induced cell death in cultured neuron by enhancing brain-derived neurotrophic factor release from astrocyte. Neuroreport 2018; 29:92-98. [PMID: 29120942 DOI: 10.1097/wnr.0000000000000911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
β-Amyloid (Aβ)-induced neuronal toxicity is an early event in the pathogenesis of Alzheimer's disease. Quetiapine (QTP) is an atypical antipsychotic drug that has neuroprotectant properties, but little is known about its direct protective effects on neurons against the Aβ-induced cell toxicity. In the present study, we investigated the neuroprotective effects of QTP on Aβ25-35-induced cell death and the possible underlying mechanisms in primary cultures of neurons. Exposure of cortical neurons to 10 μM or more Aβ25-35 caused significant viability loss in a MTT assay, and the toxic effects were not significantly prevented by the simultaneous coadministration of QTP. However, pretreated astrocyte conditioned medium (ACM) with QTP (ACMQTP) for 24 h markedly protected the neurons against the amyloid-induced cell loss. Furthermore, we revealed that QTP increased both the release of brain-derived neurotrophic factor from cultured astrocytes and the phosphorylation of extracellular signal-regulated kinase after 24 h of treatment, which might be responsible for its protective effects on neurons. Consistent with the aforementioned findings, the protective effects of ACM on neurons could potentially be abolished by the extracellular signal-regulated kinase inhibitor and tropomyosin receptor kinase B receptor blocker. In conclusion, our data demonstrated that QTP exerted its neuroprotective effects against amyloid toxicity by enhancing the brain-derived neurotrophic factor release from astrocytes.
Collapse
|
64
|
Cao C, Li X, Qin L, Luo J, Zhang M, Ou Z, Wang K. High Selenium Yeast mitigates aluminum-induced cerebral inflammation by increasing oxidative stress and blocking NO production. Biometals 2018; 31:835-843. [DOI: 10.1007/s10534-018-0128-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/08/2018] [Indexed: 12/15/2022]
|
65
|
Marie C, Pedard M, Quirié A, Tessier A, Garnier P, Totoson P, Demougeot C. Brain-derived neurotrophic factor secreted by the cerebral endothelium: A new actor of brain function? J Cereb Blood Flow Metab 2018; 38:935-949. [PMID: 29557702 PMCID: PMC5998997 DOI: 10.1177/0271678x18766772] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Low cerebral levels of brain-derived neurotrophic factor (BDNF), which plays a critical role in many brain functions, have been implicated in neurodegenerative, neurological and psychiatric diseases. Thus, increasing BDNF levels in the brain is considered an attractive possibility for the prevention/treatment of various brain diseases. To date, BDNF-based therapies have largely focused on neurons. However, given the cross-talk between endothelial cells and neurons and recent evidence that BDNF expressed by the cerebral endothelium largely accounts for BDNF levels present in the brain, it is likely that BDNF-based therapies would be most effective if they also targeted the cerebral endothelium. In this review, we summarize the available knowledge about the biology and actions of BDNF derived from endothelial cells of the cerebral microvasculature and we emphasize the remaining gaps and shortcomings.
Collapse
Affiliation(s)
- Christine Marie
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France
| | - Martin Pedard
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France.,2 Service de Neurologie, CHRU, Dijon, France
| | - Aurore Quirié
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France
| | - Anne Tessier
- 1 INSERM U1093, Univ. Bourgogne Franche-Comté, Dijon, France
| | | | - Perle Totoson
- 3 EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
| | - Céline Demougeot
- 3 EA4267 PEPITE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
66
|
Kowiański P, Lietzau G, Czuba E, Waśkow M, Steliga A, Moryś J. BDNF: A Key Factor with Multipotent Impact on Brain Signaling and Synaptic Plasticity. Cell Mol Neurobiol 2018; 38:579-593. [PMID: 28623429 PMCID: PMC5835061 DOI: 10.1007/s10571-017-0510-4] [Citation(s) in RCA: 866] [Impact Index Per Article: 123.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/08/2017] [Indexed: 12/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most widely distributed and extensively studied neurotrophins in the mammalian brain. Among its prominent functions, one can mention control of neuronal and glial development, neuroprotection, and modulation of both short- and long-lasting synaptic interactions, which are critical for cognition and memory. A wide spectrum of processes are controlled by BDNF, and the sometimes contradictory effects of its action can be explained based on its specific pattern of synthesis, comprising several intermediate biologically active isoforms that bind to different types of receptor, triggering several signaling pathways. The functions of BDNF must be discussed in close relation to the stage of brain development, the different cellular components of nervous tissue, as well as the molecular mechanisms of signal transduction activated under physiological and pathological conditions. In this review, we briefly summarize the current state of knowledge regarding the impact of BDNF on regulation of neurophysiological processes. The importance of BDNF for future studies aimed at disclosing mechanisms of activation of signaling pathways, neuro- and gliogenesis, as well as synaptic plasticity is highlighted.
Collapse
Affiliation(s)
- Przemysław Kowiański
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland.
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland.
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| | - Monika Waśkow
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland
| | - Aleksandra Steliga
- Department of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte Str., 76-200, Słupsk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki Street, 80-211, Gdańsk, Poland
| |
Collapse
|
67
|
Abstract
The role traditionally assigned to astrocytes in the pathogenesis of multiple sclerosis (MS) lesions has been the formation of the glial scar once inflammation has subsided. Astrocytes are now recognized to be early and highly active players during lesion formation and key for providing peripheral immune cells access to the central nervous system. Here, we review the role of astrocytes in the formation and evolution of MS lesions, including the recently described functional polarization of astrocytes, discuss prototypical pathways for astrocyte activation, and summarize mechanisms by which MS treatments affect astrocyte function.
Collapse
Affiliation(s)
- Gerald Ponath
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Calvin Park
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
68
|
Cragnolini AB, Montenegro G, Friedman WJ, Mascó DH. Brain-region specific responses of astrocytes to an in vitro injury and neurotrophins. Mol Cell Neurosci 2018; 88:240-248. [PMID: 29444457 DOI: 10.1016/j.mcn.2018.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/22/2018] [Accepted: 02/09/2018] [Indexed: 11/17/2022] Open
Abstract
Astrocytes are a heterogeneous population of glial cells that react to brain insults through a process referred to as astrogliosis. Reactive astrocytes are characterized by an increase in proliferation, size, migration to the injured zone and release of a plethora of chemical mediators such as NGF and BDNF. The aim of this study was to determine whether there are brain region-associated responses of astrocytes to an injury and to the neurotrophins NGF and BDNF. We used the scratch injury model to study the closure of a wound inflicted on a monolayer of astrocytes obtained from cortex, hippocampus or striatum. Our results indicate that the response of astrocytes to a mechanical lesion differ according to brain regions. Astrocytes from the striatum proliferate and repopulate the injury site more rapidly than astrocytes from cortex or hippocampus. We found that the scratch injury induced the upregulation of neurotrophin receptor p75NTR and TrkB.t in astrocytes from all brain regions studied. When astrocytes from all regions were treated with NGF, the neurotrophin induced migration of the astrocytes (assessed in Boyden chambers) and induced wound closure but did not affect proliferation. In contrast, BDNF induced wound closure but only in astrocytes from striatum. Our overall findings show the heterogeneity in astrocyte functions based on their brain region of origin, and how this functional diversity may determine their responses to an injury and to neurotrophins.
Collapse
Affiliation(s)
- Andrea Beatriz Cragnolini
- IIByT-UNC CONICET, CEBICEM, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, X5016GCA Córdoba, Argentina
| | - Gonzalo Montenegro
- IIByT-UNC CONICET, CEBICEM, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, X5016GCA Córdoba, Argentina
| | - Wilma J Friedman
- Department of Biological Sciences, Rutgers University, 225 University Avenue, Newark, N.J. 07102, United States
| | - Daniel Hugo Mascó
- IIByT-UNC CONICET, CEBICEM, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, X5016GCA Córdoba, Argentina.
| |
Collapse
|
69
|
Medelin M, Giacco V, Aldinucci A, Castronovo G, Bonechi E, Sibilla A, Tanturli M, Torcia M, Ballerini L, Cozzolino F, Ballerini C. Bridging pro-inflammatory signals, synaptic transmission and protection in spinal explants in vitro. Mol Brain 2018; 11:3. [PMID: 29334986 PMCID: PMC5769440 DOI: 10.1186/s13041-018-0347-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/04/2018] [Indexed: 01/30/2023] Open
Abstract
Multiple sclerosis is characterized by tissue atrophy involving the brain and the spinal cord, where reactive inflammation contributes to the neurodegenerative processes. Recently, the presence of synapse alterations induced by the inflammatory responses was suggested by experimental and clinical observations, in experimental autoimmune encephalomyelitis mouse model and in patients, respectively. Further knowledge on the interplay between pro-inflammatory agents, neuroglia and synaptic dysfunction is crucial to the design of unconventional protective molecules. Here we report the effects, on spinal cord circuits, of a cytokine cocktail that partly mimics the signature of T lymphocytes sub population Th1. In embryonic mouse spinal organ-cultures, containing neuronal cells and neuroglia, cytokines induced inflammatory responses accompanied by a significant increase in spontaneous synaptic activity. We suggest that cytokines specifically altered signal integration in spinal networks by speeding the decay of GABAA responses. This hypothesis is supported by the finding that synapse protection by a non-peptidic NGF mimetic molecule prevented both the changes in the time course of GABA events and in network activity that were left unchanged by the cytokine production from astrocytes and microglia present in the cultured tissue. In conclusion, we developed an important tool for the study of synaptic alterations induced by inflammation, that takes into account the role of neuronal and not neuronal resident cells.
Collapse
Affiliation(s)
- M Medelin
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy.,International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy
| | - V Giacco
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy
| | - A Aldinucci
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy
| | - G Castronovo
- Department of DSBSC, University of Florence, 50134, Florence, Italy
| | - E Bonechi
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy
| | - A Sibilla
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy
| | - M Tanturli
- Department of DSBSC, University of Florence, 50134, Florence, Italy
| | - M Torcia
- Department of DMSC, University of Florence, 50134, Florence, Italy
| | - L Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
| | - F Cozzolino
- Department of DSBSC, University of Florence, 50134, Florence, Italy
| | - C Ballerini
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy.
| |
Collapse
|
70
|
Mancini A, Tantucci M, Mazzocchetti P, de Iure A, Durante V, Macchioni L, Giampà C, Alvino A, Gaetani L, Costa C, Tozzi A, Calabresi P, Di Filippo M. Microglial activation and the nitric oxide/cGMP/PKG pathway underlie enhanced neuronal vulnerability to mitochondrial dysfunction in experimental multiple sclerosis. Neurobiol Dis 2018; 113:97-108. [PMID: 29325869 DOI: 10.1016/j.nbd.2018.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/07/2017] [Accepted: 01/04/2018] [Indexed: 12/24/2022] Open
Abstract
During multiple sclerosis (MS), a close link has been demonstrated to occur between inflammation and neuro-axonal degeneration, leading to the hypothesis that immune mechanisms may promote neurodegeneration, leading to irreversible disease progression. Energy deficits and inflammation-driven mitochondrial dysfunction seem to be involved in this process. In this work we investigated, by the use of striatal electrophysiological field-potential recordings, if the inflammatory process associated with experimental autoimmune encephalomyelitis (EAE) is able to influence neuronal vulnerability to the blockade of mitochondrial complex IV, a crucial component for mitochondrial activity responsible of about 90% of total cellular oxygen consumption. We showed that during the acute relapsing phase of EAE, neuronal susceptibility to mitochondrial complex IV inhibition is markedly enhanced. This detrimental effect was counteracted by the pharmacological inhibition of microglia, of nitric oxide (NO) synthesis and its intracellular pathway (involving soluble guanylyl cyclase, sGC, and protein kinase G, PKG). The obtained results suggest that mitochondrial complex IV exerts an important role in maintaining neuronal energetic homeostasis during EAE. The pathological processes associated with experimental MS, and in particular the activation of microglia and of the NO pathway, lead to an increased neuronal vulnerability to mitochondrial complex IV inhibition, representing promising pharmacological targets.
Collapse
Affiliation(s)
- Andrea Mancini
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Michela Tantucci
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Petra Mazzocchetti
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Antonio de Iure
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Valentina Durante
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Lara Macchioni
- Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Carmela Giampà
- Università Cattolica del Sacro Cuore, Istituto di Anatomia Umana e Biologia Cellulare, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessandra Alvino
- Università Cattolica del Sacro Cuore, Istituto di Anatomia Umana e Biologia Cellulare, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Lorenzo Gaetani
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Cinzia Costa
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Alessandro Tozzi
- Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy; IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Paolo Calabresi
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy; IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Massimiliano Di Filippo
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy.
| |
Collapse
|
71
|
Beneventano M, Spampinato SF, Merlo S, Chisari M, Platania P, Ragusa M, Purrello M, Nicoletti F, Sortino MA. Shedding of Microvesicles from Microglia Contributes to the Effects Induced by Metabotropic Glutamate Receptor 5 Activation on Neuronal Death. Front Pharmacol 2017; 8:812. [PMID: 29170640 PMCID: PMC5684115 DOI: 10.3389/fphar.2017.00812] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/26/2017] [Indexed: 01/08/2023] Open
Abstract
Metabotropic glutamate (mGlu) receptor 5 is involved in neuroinflammation and has been shown to mediate reduced inflammation and neurotoxicity and to modify microglia polarization. On the other hand, blockade of mGlu5 receptor results in inhibition of microglia activation. To dissect this controversy, we investigated whether microvesicles (MVs) released from microglia BV2 cells could contribute to the communication between microglia and neurons and whether this interaction was modulated by mGlu5 receptor. Activation of purinergic ionotropic P2X7 receptor with the stable ATP analog benzoyl-ATP (100 μM) caused rapid MVs shedding from BV2 cells. Ionic currents through P2X7 receptor increased in BV2 cells pretreated for 24 h with the mGlu5 receptor agonist CHPG (200 μM) as by patch-clamp recording. This increase was blunted when microglia cells were activated by exposure to lipopolysaccharide (LPS; 0.1 μg/ml for 6 h). Accordingly, a greater amount of MVs formed after CHPG treatment, an effect prevented by the mGlu5 receptor antagonist MTEP (100 μM), as measured by expression of flotillin, a membrane protein enriched in MVs. Transferred MVs were internalized by SH-SY5Y neurons where they did not modify neuronal death induced by a low concentration of rotenone (0.1 μM for 24 h), but significantly increased rotenone neurotoxicity when shed from CHPG-treated BV2 cells. miR146a was increased in CHPG-treated MVs, an effect concealed in MVs from LPS-activated BV2 cells that showed per se an increase in miRNA146a levels. The present data support a role for microglia-shed MVs in mGlu5-mediated modulation of neuronal death and identify miRNAs as potential critical mediators of this interaction.
Collapse
Affiliation(s)
- Martina Beneventano
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Simona F Spampinato
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Merlo
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Mariangela Chisari
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paola Platania
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Marco Ragusa
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Michele Purrello
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - Maria Angela Sortino
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
72
|
Mehrzad J, Malvandi AM, Alipour M, Hosseinkhani S. Environmentally relevant level of aflatoxin B 1 elicits toxic pro-inflammatory response in murine CNS-derived cells. Toxicol Lett 2017; 279:96-106. [DOI: 10.1016/j.toxlet.2017.07.902] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/26/2017] [Accepted: 07/31/2017] [Indexed: 01/08/2023]
|
73
|
Neural Stem Cell Transplantation Induces Stroke Recovery by Upregulating Glutamate Transporter GLT-1 in Astrocytes. J Neurosci 2017; 36:10529-10544. [PMID: 27733606 DOI: 10.1523/jneurosci.1643-16.2016] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/09/2016] [Indexed: 01/10/2023] Open
Abstract
Ischemic stroke is the leading cause of disability, but effective therapies are currently widely lacking. Recovery from stroke is very much dependent on the possibility to develop treatments able to both halt the neurodegenerative process as well as to foster adaptive tissue plasticity. Here we show that ischemic mice treated with neural precursor cell (NPC) transplantation had on neurophysiological analysis, early after treatment, reduced presynaptic release of glutamate within the ipsilesional corticospinal tract (CST), and an enhanced NMDA-mediated excitatory transmission in the contralesional CST. Concurrently, NPC-treated mice displayed a reduced CST degeneration, increased axonal rewiring, and augmented dendritic arborization, resulting in long-term functional amelioration persisting up to 60 d after ischemia. The enhanced functional and structural plasticity relied on the capacity of transplanted NPCs to localize in the peri-ischemic and ischemic area, to promote the upregulation of the glial glutamate transporter 1 (GLT-1) on astrocytes and to reduce peri-ischemic extracellular glutamate. The upregulation of GLT-1 induced by transplanted NPCs was found to rely on the secretion of VEGF by NPCs. Blocking VEGF during the first week after stroke reduced GLT-1 upregulation as well as long-term behavioral recovery in NPC-treated mice. Our results show that NPC transplantation, by modulating the excitatory-inhibitory balance and stroke microenvironment, is a promising therapy to ameliorate disability, to promote tissue recovery and plasticity processes after stroke. SIGNIFICANCE STATEMENT Tissue damage and loss of function occurring after stroke can be constrained by fostering plasticity processes of the brain. Over the past years, stem cell transplantation for repair of the CNS has received increasing interest, although underlying mechanism remain elusive. We here show that neural stem/precursor cell transplantation after ischemic stroke is able to foster axonal rewiring and dendritic plasticity and to induce long-term functional recovery. The observed therapeutic effect of neural precursor cells seems to underlie their capacity to upregulate the glial glutamate transporter on astrocytes through the vascular endothelial growth factor inducing favorable changes in the electrical and molecular stroke microenvironment. Cell-based approaches able to influence plasticity seem particularly suited to favor poststroke recovery.
Collapse
|
74
|
Dietary eicosapentaenoic acid normalizes hippocampal omega-3 and 6 polyunsaturated fatty acid profile, attenuates glial activation and regulates BDNF function in a rodent model of neuroinflammation induced by central interleukin-1β administration. Eur J Nutr 2017; 57:1781-1791. [DOI: 10.1007/s00394-017-1462-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/19/2017] [Indexed: 12/26/2022]
|
75
|
Transient glutathione depletion in the substantia nigra compacta is associated with neuroinflammation in rats. Neuroscience 2016; 335:207-20. [DOI: 10.1016/j.neuroscience.2016.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 01/21/2023]
|
76
|
Coulibaly AP, Isaacson LG. Increased Cx32 expression in spinal cord TrkB oligodendrocytes following peripheral axon injury. Neurosci Lett 2016; 627:115-20. [PMID: 27246301 PMCID: PMC4971883 DOI: 10.1016/j.neulet.2016.05.064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 10/21/2022]
Abstract
Following injury to motor axons in the periphery, retrograde influences from the injury site lead to glial cell plasticity in the vicinity of the injured neurons. Following the transection of peripherally located preganglionic axons of the cervical sympathetic trunk (CST), a population of oligodendrocyte (OL) lineage cells expressing full length TrkB, the cognate receptor for brain derived neurotrophic factor (BDNF), is significantly increased in number in the spinal cord. Such robust plasticity in OL lineage cells in the spinal cord following peripheral axon transection led to the hypothesis that the gap junction communication protein connexin 32 (Cx32), which is specific to OL lineage cells, was influenced by the injury. Following CST transection, Cx32 expression in the spinal cord intermediolateral cell column (IML), the location of the parent cell bodies, was significantly increased. The increased Cx32 expression was localized specifically to TrkB OLs in the IML, rather than other cell types in the OL cell lineage, with the population of Cx32/TrkB cells increased by 59%. Cx32 expression in association with OPCs was significantly decreased at one week following the injury. The results of this study provide evidence that peripheral axon injury can differentially affect the gap junction protein expression in OL lineage cells in the adult rat spinal cord. We conclude that the retrograde influences originating from the peripheral injury site elicit dramatic changes in the CNS expression of Cx32, which in turn may mediate the plasticity of OL lineage cells observed in the spinal cord following peripheral axon injury.
Collapse
Affiliation(s)
- Aminata P Coulibaly
- Center for Neuroscience and Behavior, Graduate Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, United States.
| | - Lori G Isaacson
- Center for Neuroscience and Behavior, Graduate Program in Cell, Molecular, and Structural Biology, Miami University, Oxford, OH 45056, United States.
| |
Collapse
|
77
|
Colombo E, Farina C. Astrocytes: Key Regulators of Neuroinflammation. Trends Immunol 2016; 37:608-620. [PMID: 27443914 DOI: 10.1016/j.it.2016.06.006] [Citation(s) in RCA: 662] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 01/09/2023]
Abstract
Astrocytes are crucial regulators of innate and adaptive immune responses in the injured central nervous system. Depending on timing and context, astrocyte activity may exacerbate inflammatory reactions and tissue damage, or promote immunosuppression and tissue repair. Recent literature has unveiled key factors and intracellular signaling pathways that govern astrocyte behavior during neuroinflammation. Here we have re-visited in vivo studies on astrocyte signaling in neuroinflammatory models focusing on evidences obtained from the analysis of transgenic mice where distinct genes involved in ligand binding, transcriptional regulation and cell communication have been manipulated in astrocytes. The integration of in vivo observations with in vitro data clarifies precise signaling steps, highlights the crosstalk among pathways and identifies shared effector mechanisms in neuroinflammation.
Collapse
Affiliation(s)
- Emanuela Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
78
|
Ulivieri C, Savino MT, Luccarini I, Fanigliulo E, Aldinucci A, Bonechi E, Benagiano M, Ortensi B, Pelicci G, D'Elios MM, Ballerini C, Baldari CT. The Adaptor Protein Rai/ShcC Promotes Astrocyte-Dependent Inflammation during Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2016; 197:480-90. [PMID: 27288534 DOI: 10.4049/jimmunol.1502063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 05/13/2016] [Indexed: 01/13/2023]
Abstract
Th17 cells have been casually associated to the pathogenesis of autoimmune disease. We have previously demonstrated that Rai/ShcC, a member of the Shc family of adaptor proteins, negatively regulates Th17 cell differentiation and lupus autoimmunity. In this study, we have investigated the pathogenic outcome of the Th17 bias associated with Rai deficiency on multiple sclerosis development, using the experimental autoimmune encephalomyelitis (EAE) mouse model. We found that, unexpectedly, EAE was less severe in Rai(-/-) mice compared with their wild-type counterparts despite an enhanced generation of myelin-specific Th17 cells that infiltrated into the CNS. Nevertheless, when adoptively transferred into immunodeficient Rai(+/+) mice, these cells promoted a more severe disease compared with wild-type encephalitogenic Th17 cells. This paradoxical phenotype was caused by a dampened inflammatory response of astrocytes, which were found to express Rai, to IL-17. The results provide evidence that Rai plays opposite roles in Th17 cell differentiation and astrocyte activation, with the latter dominant over the former in EAE, highlighting this adaptor as a potential novel target for the therapy of multiple sclerosis.
Collapse
Affiliation(s)
- Cristina Ulivieri
- Department of Life Sciences, University of Siena, 2 53100, Siena, Italy;
| | | | | | | | | | - Elena Bonechi
- Department of Neurosciences, 6 50134 Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, 3 50134 Florence, Italy; and
| | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, 16 20139 Milan, Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, 16 20139 Milan, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, 3 50134 Florence, Italy; and
| | | | | |
Collapse
|
79
|
Makar TK, Nimmagadda VK, Singh IS, Lam K, Mubariz F, Judge SI, Trisler D, Bever CT. TrkB agonist, 7,8-dihydroxyflavone, reduces the clinical and pathological severity of a murine model of multiple sclerosis. J Neuroimmunol 2016; 292:9-20. [DOI: 10.1016/j.jneuroim.2016.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/23/2015] [Accepted: 01/04/2016] [Indexed: 01/05/2023]
|
80
|
Nitric oxide mediates glial-induced neurodegeneration in Alexander disease. Nat Commun 2015; 6:8966. [PMID: 26608817 PMCID: PMC4674772 DOI: 10.1038/ncomms9966] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/22/2015] [Indexed: 01/07/2023] Open
Abstract
Glia play critical roles in maintaining the structure and function of the nervous system; however, the specific contribution that astroglia make to neurodegeneration in human disease states remains largely undefined. Here we use Alexander disease, a serious degenerative neurological disorder caused by astrocyte dysfunction, to identify glial-derived NO as a signalling molecule triggering astrocyte-mediated neuronal degeneration. We further find that NO acts through cGMP signalling in neurons to promote cell death. Glial cells themselves also degenerate, via the DNA damage response and p53. Our findings thus define a specific mechanism for glial-induced non-cell autonomous neuronal cell death, and identify a potential therapeutic target for reducing cellular toxicity in Alexander disease, and possibly other neurodegenerative disorders with glial dysfunction. Alexander disease is a rare neurological disorder caused by mutations in GFAP, yet it is unclear how glial disruptions lead to neural death. Here, Wang et al. identify a mechanism by which glial-derived nitric oxide leads to neuronal degeneration in fly and mouse models of the disease.
Collapse
|
81
|
Poddar R, Rajagopal S, Shuttleworth CW, Paul S. Zn2+-dependent Activation of the Trk Signaling Pathway Induces Phosphorylation of the Brain-enriched Tyrosine Phosphatase STEP: MOLECULAR BASIS FOR ZN2+-INDUCED ERK MAPK ACTIVATION. J Biol Chem 2015; 291:813-25. [PMID: 26574547 DOI: 10.1074/jbc.m115.663468] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Indexed: 11/06/2022] Open
Abstract
Excessive release of Zn(2+) in the brain is implicated in the progression of acute brain injuries. Although several signaling cascades have been reported to be involved in Zn(2+)-induced neurotoxicity, a potential contribution of tyrosine phosphatases in this process has not been well explored. Here we show that exposure to high concentrations of Zn(2+) led to a progressive increase in phosphorylation of the striatal-enriched phosphatase (STEP), a component of the excitotoxic-signaling pathway that plays a role in neuroprotection. Zn(2+)-mediated phosphorylation of STEP61 at multiple sites (hyperphosphorylation) was induced by the up-regulation of brain-derived neurotropic factor (BDNF), tropomyosin receptor kinase (Trk) signaling, and activation of cAMP-dependent PKA (protein kinase A). Mutational studies further show that differential phosphorylation of STEP61 at the PKA sites, Ser-160 and Ser-221 regulates the affinity of STEP61 toward its substrates. Consistent with these findings we also show that BDNF/Trk/PKA mediated signaling is required for Zn(2+)-induced phosphorylation of extracellular regulated kinase 2 (ERK2), a substrate of STEP that is involved in Zn(2+)-dependent neurotoxicity. The strong correlation between the temporal profile of STEP61 hyperphosphorylation and ERK2 phosphorylation indicates that loss of function of STEP61 through phosphorylation is necessary for maintaining sustained ERK2 phosphorylation. This interpretation is further supported by the findings that deletion of the STEP gene led to a rapid and sustained increase in ERK2 phosphorylation within minutes of exposure to Zn(2+). The study provides further insight into the mechanisms of regulation of STEP61 and also offers a molecular basis for the Zn(2+)-induced sustained activation of ERK2.
Collapse
Affiliation(s)
| | | | - C William Shuttleworth
- Neurosciences University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131
| | - Surojit Paul
- From the Departments of Neurology and Neurosciences University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131
| |
Collapse
|
82
|
Di Dario M, Colombo E, Govi C, De Feo D, Messina MJ, Romeo M, Sangalli F, Moiola L, Rodegher M, Martino G, Martinelli V, Comi G, Farina C. Myeloid cells as target of fingolimod action in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e157. [PMID: 26587553 PMCID: PMC4635549 DOI: 10.1212/nxi.0000000000000157] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/05/2015] [Indexed: 01/03/2023]
Abstract
Objective: To track the effects of fingolimod, an approved drug for multiple sclerosis (MS), on the activation of myeloid cells from the periphery to the CNS. Methods: In vitro and ex vivo immunologic studies coupled with flow cytometry were performed to evaluate the action of fingolimod on lipopolysaccharide (LPS)–induced expression of activation markers in human monocytes from healthy participants, participants with untreated MS, and participants with fingolimod-treated MS. In vivo administration of fingolimod during experimental autoimmune encephalomyelitis (EAE) was established to verify the activation state of splenic, CNS infiltrating, and CNS resident myeloid cells ex vivo at flow cytometer. Results: We found that in vitro exposure of human monocytes to fingolimod inhibited LPS-induced CD25 and CD150 expression and tumor necrosis factor–α (TNF-α) secretion without altering immune cell survival. Further, EAE treatment with fingolimod led to reduced amounts of TNF-α produced by myeloid cells in vivo in the spleen and CNS. Finally, while displaying normal induction of CD25 and CD150 levels at high LPS concentration, monocytes from patients with fingolimod-treated MS showed significantly higher activation threshold at suboptimal LPS stimulation than controls. Conclusions: The inhibition of myeloid cell activation may be part of the immunosuppressive action of fingolimod and take place in the periphery and in the CNS.
Collapse
Affiliation(s)
- Marco Di Dario
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuela Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Govi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Donatella De Feo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria José Messina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marzia Romeo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sangalli
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mariaemma Rodegher
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vittorio Martinelli
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giancarlo Comi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
83
|
Yuste JE, Tarragon E, Campuzano CM, Ros-Bernal F. Implications of glial nitric oxide in neurodegenerative diseases. Front Cell Neurosci 2015; 9:322. [PMID: 26347610 PMCID: PMC4538301 DOI: 10.3389/fncel.2015.00322] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/03/2015] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) is a pleiotropic janus-faced molecule synthesized by nitric oxide synthases (NOS) which plays a critical role in a number of physiological and pathological processes in humans. The physiological roles of NO depend on its local concentrations, as well as its availability and the nature of downstream target molecules. Its double-edged sword action has been linked to neurodegenerative disorders. Excessive NO production, as the evoked by inflammatory signals, has been identified as one of the major causative reasons for the pathogenesis of several neurodegenerative diseases. Moreover, excessive NO synthesis under neuroinflammation leads to the formation of reactive nitrogen species and neuronal cell death. There is an intimate relation between microglial activation, NO and neuroinflammation in the human brain. The role of NO in neuroinflammation has been defined in animal models where this neurotransmitter can modulate the inflammatory process acting on key regulatory pathways, such as those associated with excitotoxicity processes induced by glutamate accumulation and microglial activation. Activated glia express inducible NOS and produce NO that triggers calcium mobilization from the endoplasmic reticulum, activating the release of vesicular glutamate from astroglial cells resulting in neuronal death. This change in microglia potentially contributes to the increased age-associated susceptibility and neurodegeneration. In the current review, information is provided about the role of NO, glial activation and age-related processes in the central nervous system (CNS) that may be helpful in the isolation of new therapeutic targets for aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jose Enrique Yuste
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain
| | - Ernesto Tarragon
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain ; Département des Sciences Biomédicales et Précliniques/Biochimie et Physiologie du Système Nerveux, Centre de Recherche du Cyclotron, Université de Liège Liège, Belgium
| | - Carmen María Campuzano
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain
| | - Francisco Ros-Bernal
- Neurobiotechnology Group, Departament of Medicine, Facultat de Ciències de la Salut, Universitat Jaume I Castelló de la Plana, Spain
| |
Collapse
|
84
|
Carandini T, Colombo F, Finardi A, Casella G, Garzetti L, Verderio C, Furlan R. Microvesicles: What is the Role in Multiple Sclerosis? Front Neurol 2015; 6:111. [PMID: 26074867 PMCID: PMC4443736 DOI: 10.3389/fneur.2015.00111] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/04/2015] [Indexed: 12/21/2022] Open
Abstract
Microvesicles are a recently described way of cell communication that has been implicated in a number of biological processes, including neuroinflammation. Widely investigated as biomarkers in oncology and neurological disorders, little is known of the role of microvesicles in the pathogenesis of diseases such as multiple sclerosis (MS). Several evidences suggest that pro-inflammatory microglia and infiltrating macrophages release microvesicles that spread inflammatory signals and alter neuronal functions. We review here available information on microvesicles, with a special focus on microglia and macrophage microvesicles, in the pathogenesis of MS, and as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Tiziana Carandini
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute , Milan , Italy
| | - Federico Colombo
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute , Milan , Italy
| | - Annamaria Finardi
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute , Milan , Italy
| | - Giacomo Casella
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute , Milan , Italy
| | - Livia Garzetti
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute , Milan , Italy
| | - Claudia Verderio
- CNR Institute of Neuroscience , Milan , Italy ; IRCCS Humanitas , Rozzano , Italy
| | - Roberto Furlan
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute , Milan , Italy
| |
Collapse
|
85
|
Dixon AR, Philbert MA. Morphometric assessment of toxicant induced neuronal degeneration in full and restricted contact co-cultures of embryonic cortical rat neurons and astrocytes: using m-Dinitrobezene as a model neurotoxicant. Toxicol In Vitro 2015; 29:564-74. [PMID: 25553915 PMCID: PMC4418429 DOI: 10.1016/j.tiv.2014.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 11/05/2014] [Accepted: 12/11/2014] [Indexed: 01/22/2023]
Abstract
With m-Dinitrobenzene (m-DNB) as a selected model neurotoxicant, we demonstrate how to assess neurotoxicity, using morphology based measurement of neurite degeneration, in a conventional "full-contact" and a modern "restricted-contact" co-culture of rat cortical neurons and astrocytes. In the "full-contact" co-culture, neurons and astrocytes in complete physical contact are "globally" exposed to m-DNB. A newly emergent "restricted-contact" co-culture is attained with a microfluidic device that polarizes neuron somas and neurites into separate compartments, and the neurite compartment is "selectively" exposed to m-DNB. Morphometric analysis of the neuronal area revealed that m-DNB exposure produced no significant change in mean neuronal cell area in "full-contact" co-cultures, whereas a significant decrease was observed for neuron monocultures. Neurite elaboration into a neurite exclusive compartment in a compartmentalized microfluidic device, for both monocultures (no astrocytes) and "restricted" co-cultures (astrocytes touching neurites), decreased with exposure to increasing concentrations of m-DNB, but the average neurite area was higher in co-cultures. By using co-culture systems that more closely approach biological and architectural complexities, and the directionality of exposure found in the brain, this study provides a methodological foundation for unraveling the role of physical contact between astrocytes and neurons in mitigating the toxic effects of chemicals such as m-DNB.
Collapse
Affiliation(s)
- Angela R Dixon
- Toxicology Program, School of Public Health, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Martin A Philbert
- Toxicology Program, School of Public Health, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
86
|
Abstract
The brain under immunological attack does not surrender quietly. Investigation of brain lesions in multiple sclerosis (MS) reveals a coordinated molecular response involving various proteins and small molecules ranging from heat shock proteins to small lipids, neurotransmitters, and even gases, which provide protection and foster repair. Reduction of inflammation serves as a necessary prerequisite for effective recovery and regeneration. Remarkably, many lesion-resident molecules activate pathways leading to both suppression of inflammation and promotion of repair mechanisms. These guardian molecules and their corresponding physiologic pathways could potentially be exploited to silence inflammation and repair the injured and degenerating brain and spinal cord in both relapsing-remitting and progressive forms of MS and may be beneficial in other neurologic and psychiatric conditions.
Collapse
|
87
|
Neuronal BDNF signaling is necessary for the effects of treadmill exercise on synaptic stripping of axotomized motoneurons. Neural Plast 2015; 2015:392591. [PMID: 25918648 PMCID: PMC4397030 DOI: 10.1155/2015/392591] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 03/09/2015] [Accepted: 03/16/2015] [Indexed: 11/18/2022] Open
Abstract
The withdrawal of synaptic inputs from the somata and proximal dendrites of spinal motoneurons following peripheral nerve injury could contribute to poor functional recovery. Decreased availability of neurotrophins to afferent terminals on axotomized motoneurons has been implicated as one cause of the withdrawal. No reduction in contacts made by synaptic inputs immunoreactive to the vesicular glutamate transporter 1 and glutamic acid decarboxylase 67 is noted on axotomized motoneurons if modest treadmill exercise, which stimulates the production of neurotrophins by spinal motoneurons, is applied after nerve injury. In conditional, neuron-specific brain-derived neurotrophic factor (BDNF) knockout mice, a reduction in synaptic contacts onto motoneurons was noted in intact animals which was similar in magnitude to that observed after nerve transection in wild-type controls. No further reduction in coverage was found if nerves were cut in knockout mice. Two weeks of moderate daily treadmill exercise following nerve injury in these BDNF knockout mice did not affect synaptic inputs onto motoneurons. Treadmill exercise has a profound effect on synaptic inputs to motoneurons after peripheral nerve injury which requires BDNF production by those postsynaptic cells.
Collapse
|
88
|
Razavi S, Nazem G, Mardani M, Esfandiari E, Salehi H, Esfahani SHZ. Neurotrophic factors and their effects in the treatment of multiple sclerosis. Adv Biomed Res 2015; 4:53. [PMID: 25802822 PMCID: PMC4361963 DOI: 10.4103/2277-9175.151570] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/24/2014] [Indexed: 12/24/2022] Open
Abstract
Neurotrophins are small molecules of polypeptides, which include nerve growth factor (NGF) family, glial cell line–derived neurotrophic factor (GDNF) family ligands, and neuropoietic cytokines. These factors have an important role in neural regeneration, remyelination, and regulating the development of the peripheral and central nervous systems (PNS and CNS, respectively) by intracellular signaling through specific receptors. It has been suggested that the pathogenesis of human neurodegenerative disorders may be due to an alteration in the neurotrophic factors and their receptors. The use of neurotrophic factors as therapeutic agents is a novel strategy for restoring and maintaining neuronal function during neurodegenerative disorders such as multiple sclerosis. Innate and adaptive immune responses contribute to pathology of neurodegenerative disorders. Furthermore, autoimmune and mesenchymal stem cells, by the release of neurotrophic factors, have the ability to protect neuronal population and can efficiently suppress the formation of new lesions. So, these cells may be an alternative source for delivering neurotrophic factors into the CNS.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghasemi Nazem
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Mardani
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
89
|
Guerau-de-Arellano M, Liu Y, Meisen WH, Pitt D, Racke MK, Lovett-Racke AE. Analysis of miRNA in Normal Appearing White Matter to Identify Altered CNS Pathways in Multiple Sclerosis. ACTA ACUST UNITED AC 2015; 1. [PMID: 26894232 DOI: 10.21767/2471-8153.100006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Genetic studies suggest that the immune system is the greatest genetic contributor to multiple sclerosis (MS) susceptibility. Yet, these immune-related genes do not explain why inflammation is limited to the CNS in MS. We hypothesize that there is an underlying dysregulation in the CNS of MS patients that makes them more vulnerable to CNS inflammation. The sparsity of CNS-related genes associated with MS suggests that epigenetic changes in the CNS may play a role. Thus, a miRNA profiling study was performed in NAWM of MS patients and control subjects to determine if specific CNS pathways can be identified that may be altered due to miRNA-mediated post-transcriptional dysregulation. There were 15 differentially expressed miRNAs found in the MS patients' NAWM. Pathway analysis indicated that the MAPK pathway and pathways associated with the blood-brain barrier were predicted to be significantly affected by these miRNAs. Using target predication and mRNA analysis, an inverse relationship was found between miR-191 and BDNF, SOX4, FZD5 and WSB1. The pathway and target analysis of the MS-associated miRNAs suggests that MS patients' CNS is more prone to inflammation and less capable of repair, yet enriched in neuroprotective mechanisms.
Collapse
Affiliation(s)
- Mireia Guerau-de-Arellano
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yue Liu
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Walter H Meisen
- Biomedical Sciences Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - David Pitt
- Department of Neurology, Yale School of Medicine, New Haven, CT.; Department of Neurology, The Ohio State University Wexner MedicalCenter, Columbus, OH, USA
| | - Michael K Racke
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neurology, The Ohio State University Wexner MedicalCenter, Columbus, OH, USA
| | - Amy E Lovett-Racke
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
90
|
Watson AJ, Henson K, Dorsey SG, Frank MG. The truncated TrkB receptor influences mammalian sleep. Am J Physiol Regul Integr Comp Physiol 2014; 308:R199-207. [PMID: 25502751 DOI: 10.1152/ajpregu.00422.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin hypothesized to play an important role in mammalian sleep expression and regulation. In order to investigate the role of the truncated receptor for BDNF, TrkB.T1, in mammalian sleep, we examined sleep architecture and sleep regulation in adult mice constitutively lacking this receptor. We find that TrkB.T1 knockout mice have increased REM sleep time, reduced REM sleep latency, and reduced sleep continuity. These results demonstrate a novel role for the TrkB.T1 receptor in sleep expression and provide new insights into the relationship between BDNF, psychiatric illness, and sleep.
Collapse
Affiliation(s)
- Adam J Watson
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kyle Henson
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan G Dorsey
- School of Nursing, University of Maryland, Baltimore, Maryland; and
| | - Marcos G Frank
- College of Medical Sciences, Sleep and Performance Research Center, Washington State University Spokane, Spokane, Washington
| |
Collapse
|
91
|
Repulsive Guidance Molecule-a Is Involved in Th17-Cell-Induced Neurodegeneration in Autoimmune Encephalomyelitis. Cell Rep 2014; 9:1459-70. [DOI: 10.1016/j.celrep.2014.10.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 09/22/2014] [Accepted: 10/14/2014] [Indexed: 12/21/2022] Open
|
92
|
dos Santos NAG, Martins NM, Silva RDB, Ferreira RS, Sisti FM, dos Santos AC. Caffeic acid phenethyl ester (CAPE) protects PC12 cells from MPP+ toxicity by inducing the expression of neuron-typical proteins. Neurotoxicology 2014; 45:131-8. [PMID: 25454720 DOI: 10.1016/j.neuro.2014.09.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/29/2014] [Accepted: 09/23/2014] [Indexed: 01/18/2023]
Abstract
Neurite loss is an early event in neurodegenerative diseases; therefore, the regeneration of the network of neurites constitutes an interesting strategy of treatment for such disorders. Neurotrophic factors play a critical role in neuronal regeneration, but their clinical use is limited by their inability to cross the blood brain barrier. Oxidative and inflammatory events are implicated in neurodegeneration and antioxidant compounds have been suggested as potential neuroprotectors. The protective potential of CAPE (caffeic acid phenethyl ester) has been shown in different models of neurotoxicity (in vitro and in vivo) and it has been associated with immune-modulatory, antioxidant and anti-inflammatory properties; however, other mechanisms might be involved. The present study demonstrates that CAPE protects PC12 cells from the cellular death induced by the dopaminergic neurotoxin MPP(+) by increasing the network of neurites. Results showed that CAPE induced the formation, elongation and ramification of neurites in PC12 cells non-stimulated with NGF (nerve growth factor) and inhibited the shortage of neurites induced by the dopaminergic neurotoxin. These effects were associated with increased expression of neuron-typical proteins responsible for axonal growth (GAP-43) and synaptogenesis (synaptophysin and synapsin I). It is noteworthy that, unlike neurotrophins, CAPE would be able to cross the blood brain barrier and exert its neurotrophic effects in the brain. This study corroborates the therapeutic potential of CAPE in neurodegenerative diseases while proposes the involvement of neuroplasticity in the mechanism of neuroprotection.
Collapse
Affiliation(s)
- Neife Aparecida Guinaim dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Nádia Maria Martins
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Roberto de Barros Silva
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Flávia Malvestio Sisti
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio Cardozo dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
93
|
Sayre NL, Chen Y, Sifuentes M, Stoveken B, Lechleiter JD. Purinergic receptor stimulation decreases ischemic brain damage by energizing astrocyte mitochondria. ADVANCES IN NEUROBIOLOGY 2014; 11:121-50. [PMID: 25236727 DOI: 10.1007/978-3-319-08894-5_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
As a leading cause of death in the world, cerebral ischemic stroke has limited treatment options. The lack of glucose and oxygen after stroke is particularly harmful in the brain because neuronal metabolism accounts for significantly more energy consumption per gram of body weight compared to other organs. Our laboratory has identified mitochondrial metabolism of astrocytes to be a key target for pharmacologic intervention, not only because astrocytes play a central role in regulating brain metabolism, but also because they are essential for neuronal health and support. Here we review current literature pertaining to the pathobiology of stroke, along with the role of astrocytes and metabolism in stroke. We also discuss our research, which has revealed that pharmacologic stimulation of metabotropic P2Y1 receptor signaling in astrocytes can increase mitochondrial energy production and also reduce damage after stroke.
Collapse
Affiliation(s)
- Naomi L Sayre
- Department of Cellular and Structural Biology, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | | | | | | | | |
Collapse
|
94
|
Namekata K, Kimura A, Harada C, Yoshida H, Matsumoto Y, Harada T. Dock3 protects myelin in the cuprizone model for demyelination. Cell Death Dis 2014; 5:e1395. [PMID: 25165881 PMCID: PMC4454328 DOI: 10.1038/cddis.2014.357] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 07/11/2014] [Accepted: 07/21/2014] [Indexed: 12/18/2022]
Abstract
Dedicator of cytokinesis 3 (Dock3) belongs to an atypical family of the guanine nucleotide exchange factors. It is predominantly expressed in the neural tissues and causes cellular morphological changes by activating the small GTPase Rac1. We previously reported that Dock3 overexpression protects retinal ganglion cells from excitotoxic cell death. Oligodendrocytes are the myelinating cells of axons in the central nervous system and these cells are damaged in demyelinating disorders including multiple sclerosis (MS) and optic neuritis. In this study, we examined if Dock3 is expressed in oligodendrocytes and if increasing Dock3 signals can suppress demyelination in a cuprizone-induced demyelination model, an animal model of MS. We demonstrate that Dock3 is expressed in oligodendrocytes and Dock3 overexpression protects myelin in the corpus callosum following cuprizone treatment. Furthermore, we show that cuprizone demyelinates optic nerves and the extent of demyelination is ameliorated in mice overexpressing Dock3. Cuprizone treatment impairs visual function, which was demonstrated by multifocal electroretinograms, an established non-invasive method, and Dock3 overexpression prevented this effect. In mice overexpressing Dock3, Erk activation is increased, suggesting this may at least partly explain the observed protective effects. Our findings suggest that Dock3 may be a therapeutic target for demyelinating disorders including optic neuritis.
Collapse
Affiliation(s)
- K Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - A Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - C Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - H Yoshida
- Department of Neuro-ophthalmology, Tokyo Metropolitan Neurological Hospital, Tokyo, Japan
| | - Y Matsumoto
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - T Harada
- 1] Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan [2] Department of Neuro-ophthalmology, Tokyo Metropolitan Neurological Hospital, Tokyo, Japan
| |
Collapse
|
95
|
Conditional ablation of astroglial CCL2 suppresses CNS accumulation of M1 macrophages and preserves axons in mice with MOG peptide EAE. J Neurosci 2014; 34:8175-85. [PMID: 24920622 DOI: 10.1523/jneurosci.1137-14.2014] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Current multiple sclerosis (MS) therapies only partially prevent chronically worsening neurological deficits, which are largely attributable to progressive loss of CNS axons. Prior studies of experimental autoimmune encephalomyelitis (EAE) induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein peptide 35-55 (MOG peptide), a model of MS, documented continued axon loss for months after acute CNS inflammatory infiltrates had subsided, and massive astroglial induction of CCL2 (MCP-1), a chemokine for CCR2(+) monocytes. We now report that conditional deletion of astroglial CCL2 significantly decreases CNS accumulation of classically activated (M1) monocyte-derived macrophages and microglial expression of M1 markers during the initial CNS inflammatory phase of MOG peptide EAE, reduces the acute and long-term severity of clinical deficits and slows the progression of spinal cord axon loss. In addition, lack of astroglial-derived CCL2 results in increased accumulation of Th17 cells within the CNS in these mice, but also in greater confinement of CD4(+) lymphocytes to CNS perivascular spaces. These findings suggest that therapies designed to inhibit astroglial CCL2-driven trafficking of monocyte-derived macrophages to the CNS during acute MS exacerbations have the potential to significantly reduce CNS axon loss and slow progression of neurological deficits.
Collapse
|
96
|
Distribution and phenotype of TrkB oligodendrocyte lineage cells in the adult rat spinal cord. Brain Res 2014; 1582:21-33. [PMID: 25072185 DOI: 10.1016/j.brainres.2014.07.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
The distribution and phenotype of a previously undescribed population of nonneuronal cells in the intact spinal cord that expresses TrkB, the cognate receptor for brain derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4), were characterized by examining the extent of co-localization of TrkB with NG2, which identifies oligodendrocyte progenitors (OPCs) or CC1, a marker for mature oligodendrocytes (OLs). All TrkB nonneuronal cells expressed Olig2, confirming their role in the OL lineage. Similar to OPCs and OLs, TrkB cells resided in gray and white matter of the spinal cord in similar abundance. Less than 2% of TrkB cells expressed NG2, while over 80% of TrkB cells in the adult spinal cord co-expressed CC1. Most OPCs did not express detectable levels of TrkB, however a small OPC pool (~5%) showed TrkB immunoreactivity. The majority of mature OLs (~65%) expressed TrkB, but a population of mature OLs (~36%) did not express TrkB at detectable levels, and 17% of TrkB nonneuronal cells did not express NG2 or CC1. Approximately 20% of the TrkB nonneuronal population in the ventral horn resided in close proximity to motor neurons and were categorized as perineuronal. We conclude that TrkB is expressed by several pools of OL lineage cells in the adult spinal cord. These findings are important in understanding the neurotrophin regulation of OL lineage cells in the adult spinal cord.
Collapse
|
97
|
Colombo E, Di Dario M, Capitolo E, Chaabane L, Newcombe J, Martino G, Farina C. Fingolimod may support neuroprotection via blockade of astrocyte nitric oxide. Ann Neurol 2014; 76:325-37. [PMID: 25043204 DOI: 10.1002/ana.24217] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 07/01/2014] [Accepted: 07/01/2014] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Although astrocytes participate in glial scar formation and tissue repair, dysregulation of the NFκB pathway and of nitric oxide (NO) production in these glia cells contributes to neuroinflammation and neurodegeneration. Here we investigated the role of the crosstalk between sphingosine-1-phosphate (S1P) and cytokine signaling cascades in astrocyte activation and inflammation-mediated neurodegeneration, and addressed the effects of fingolimod on astrocyte-neuron interaction and NO synthesis in vivo. METHODS Immunohistochemistry, immunofluorescence, and confocal microscopy were used to detect S1P receptors, interleukin (IL) 1R, IL17RA, and nitrosative stress in multiple sclerosis (MS) plaques, experimental autoimmune encephalomyelitis (EAE) spinal cord, and the spinal cord of fingolimod-treated EAE mice. An in vitro model was established to study the effects of S1P, IL1, and IL17 stimulation on NFkB translocation and NO production in astrocytes, on spinal neuron survival, and on astrocyte-neuron interaction. Furthermore, fingolimod efficacy in blocking astrocyte-mediated neurodegeneration was evaluated. RESULTS We found coordinated upregulation of IL1R, IL17RA, S1P1, and S1P3 together with nitrosative markers in astrocytes within MS and EAE lesions. In vitro studies revealed that S1P, IL17, and IL1 induced NFκB translocation and NO production in astrocytes, and astrocyte conditioned media triggered neuronal death. Importantly, fingolimod blocked the 2 activation events evoked in astrocytes by either S1P or inflammatory cytokines, resulting in inhibition of astrocyte-mediated neurodegeneration. Finally, therapeutic administration of fingolimod to EAE mice hampered astrocyte activation and NO production. INTERPRETATION A neuroprotective effect of fingolimod in vivo may result from its inhibitory action on key astrocyte activation steps.
Collapse
Affiliation(s)
- Emanuela Colombo
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
98
|
Lieury A, Chanal M, Androdias G, Reynolds R, Cavagna S, Giraudon P, Confavreux C, Nataf S. Tissue remodeling in periplaque regions of multiple sclerosis spinal cord lesions. Glia 2014; 62:1645-58. [DOI: 10.1002/glia.22705] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/23/2014] [Accepted: 05/23/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Alice Lieury
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team; Lyon France
- University Lyon 1; Lyon France
| | - Marie Chanal
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team; Lyon France
- University Lyon 1; Lyon France
| | - Géraldine Androdias
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team; Lyon France
- University Lyon 1; Lyon France
- Service de Neurologie A and Eugène Devic Foundation, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon (Lyon University Hospital); Bron France
| | - Richard Reynolds
- Wolfson Neuroscience Laboratories, Hammersmith Hospital Campus, Imperial College Faculty of Medicine; London United Kingdom
| | - Sylvie Cavagna
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team; Lyon France
| | - Pascale Giraudon
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team; Lyon France
| | - Christian Confavreux
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team; Lyon France
- University Lyon 1; Lyon France
- Service de Neurologie A and Eugène Devic Foundation, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon (Lyon University Hospital); Bron France
| | - Serge Nataf
- INSERM U1028, CNRS UMR 5292, Lyon Neuroscience Research Center, Neuro-Oncology and Neuro-Inflammation Team; Lyon France
- University Lyon 1; Lyon France
- Banque de Cellules et de Tissus, Hôpital Edouard Herriot, Hospices Civils de Lyon (Lyon University Hospital); Lyon France
| |
Collapse
|
99
|
Colombo E, Tentorio P, Musio S, Rajewsky K, Pedotti R, Casola S, Farina C. Skewed B cell differentiation affects lymphoid organogenesis but not T cell-mediated autoimmunity. Clin Exp Immunol 2014; 176:58-65. [PMID: 24325711 DOI: 10.1111/cei.12250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2013] [Indexed: 01/23/2023] Open
Abstract
B cell receptor (BCR) signalling determines B cell differentiation and may potentially alter T cell-mediated immune responses. In this study we used two transgenic strains of BCR-deficient mice expressing Epstein-Barr virus latent membrane protein (LMP)2A in B cells, where either follicular and marginal zone differentiation (D(H)LMP2A mice) or B-1 cell development (V(H)LMP2A mice) were supported, and evaluated the effects of skewed B lymphocyte differentiation on lymphoid organogenesis and T cell responses in vivo. Compared to wild-type animals, both transgenic strains displayed alterations in the composition of lymphoid organs and in the dynamics of distinct immune cell subsets following immunization with the self-antigen PLP₁₈₅₋₂₀₆. However, ex-vivo T cell proliferation to PLP₁₈₅₋₂₀₆ peptide measured in immunized D(H)LMP2A and V(H)LMP2A mice was similar to that detected in immunized control mice. Further, clinical expression of experimental autoimmune encephalitis in both LMP2A strains was identical to that of wild-type mice. In conclusion, mice with skewed B cell differentiation driven by LMP2A expression in BCR-negative B cells do not show changes in the development of a T cell mediated disease model of autoimmunity, suggesting that compensatory mechanisms support the generation of T cell responses.
Collapse
Affiliation(s)
- E Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy; Neuroimmunology and Neuromuscular Disorders Unit, Foundation IRCCS-Neurological Institute Carlo Besta, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Multiple sclerosis (MS) is the most frequent chronic inflammatory disease of the CNS, and imposes major burdens on young lives. Great progress has been made in understanding and moderating the acute inflammatory components of MS, but the pathophysiological mechanisms of the concomitant neurodegeneration--which causes irreversible disability--are still not understood. Chronic inflammatory processes that continuously disturb neuroaxonal homeostasis drive neurodegeneration, so the clinical outcome probably depends on the balance of stressor load (inflammation) and any remaining capacity for neuronal self-protection. Hence, suitable drugs that promote the latter state are sorely needed. With the aim of identifying potential novel therapeutic targets in MS, we review research on the pathological mechanisms of neuroaxonal dysfunction and injury, such as altered ion channel activity, and the endogenous neuroprotective pathways that counteract oxidative stress and mitochondrial dysfunction. We focus on mechanisms inherent to neurons and their axons, which are separable from those acting on inflammatory responses and might, therefore, represent bona fide neuroprotective drug targets with the capability to halt MS progression.
Collapse
|