1
|
Guadalupi L, Vanni V, Balletta S, Caioli S, De Vito F, Fresegna D, Sanna K, Nencini M, Donninelli G, Volpe E, Mariani F, Battistini L, Stampanoni Bassi M, Gilio L, Bruno A, Dolcetti E, Buttari F, Mandolesi G, Centonze D, Musella A. Interleukin-9 protects from microglia- and TNF-mediated synaptotoxicity in experimental multiple sclerosis. J Neuroinflammation 2024; 21:128. [PMID: 38745307 PMCID: PMC11092167 DOI: 10.1186/s12974-024-03120-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a progressive neurodegenerative disease of the central nervous system characterized by inflammation-driven synaptic abnormalities. Interleukin-9 (IL-9) is emerging as a pleiotropic cytokine involved in MS pathophysiology. METHODS Through biochemical, immunohistochemical, and electrophysiological experiments, we investigated the effects of both peripheral and central administration of IL-9 on C57/BL6 female mice with experimental autoimmune encephalomyelitis (EAE), a model of MS. RESULTS We demonstrated that both systemic and local administration of IL-9 significantly improved clinical disability, reduced neuroinflammation, and mitigated synaptic damage in EAE. The results unveil an unrecognized central effect of IL-9 against microglia- and TNF-mediated neuronal excitotoxicity. Two main mechanisms emerged: first, IL-9 modulated microglial inflammatory activity by enhancing the expression of the triggering receptor expressed on myeloid cells-2 (TREM2) and reducing TNF release. Second, IL-9 suppressed neuronal TNF signaling, thereby blocking its synaptotoxic effects. CONCLUSIONS The data presented in this work highlight IL-9 as a critical neuroprotective molecule capable of interfering with inflammatory synaptopathy in EAE. These findings open new avenues for treatments targeting the neurodegenerative damage associated with MS, as well as other inflammatory and neurodegenerative disorders of the central nervous system.
Collapse
Affiliation(s)
- Livia Guadalupi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Sara Balletta
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | - Silvia Caioli
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | | | - Diego Fresegna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Krizia Sanna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Monica Nencini
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Gloria Donninelli
- Molecular Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome, 00143, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome, 00143, Italy
| | - Fabrizio Mariani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Luca Battistini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, Rome, 00143, Italy
| | | | - Luana Gilio
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | - Antonio Bruno
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
- Ph.D. Program in Neuroscience, Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Ettore Dolcetti
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
- Ph.D. Program in Neuroscience, Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Fabio Buttari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, 00166, Italy
| | - Diego Centonze
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy.
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077, Italy.
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, 00166, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, 00166, Italy
| |
Collapse
|
2
|
Tichauer JE, Arellano G, Acuña E, González LF, Kannaiyan NR, Murgas P, Panadero-Medianero C, Ibañez-Vega J, Burgos PI, Loda E, Miller SD, Rossner MJ, Gebicke-Haerter PJ, Naves R. Interferon-gamma ameliorates experimental autoimmune encephalomyelitis by inducing homeostatic adaptation of microglia. Front Immunol 2023; 14:1191838. [PMID: 37334380 PMCID: PMC10272814 DOI: 10.3389/fimmu.2023.1191838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Compelling evidence has shown that interferon (IFN)-γ has dual effects in multiple sclerosis and in its animal model of experimental autoimmune encephalomyelitis (EAE), with results supporting both a pathogenic and beneficial function. However, the mechanisms whereby IFN-γ may promote neuroprotection in EAE and its effects on central nervous system (CNS)-resident cells have remained an enigma for more than 30 years. In this study, the impact of IFN-γ at the peak of EAE, its effects on CNS infiltrating myeloid cells (MC) and microglia (MG), and the underlying cellular and molecular mechanisms were investigated. IFN-γ administration resulted in disease amelioration and attenuation of neuroinflammation associated with significantly lower frequencies of CNS CD11b+ myeloid cells and less infiltration of inflammatory cells and demyelination. A significant reduction in activated MG and enhanced resting MG was determined by flow cytometry and immunohistrochemistry. Primary MC/MG cultures obtained from the spinal cord of IFN-γ-treated EAE mice that were ex vivo re-stimulated with a low dose (1 ng/ml) of IFN-γ and neuroantigen, promoted a significantly higher induction of CD4+ regulatory T (Treg) cells associated with increased transforming growth factor (TGF)-β secretion. Additionally, IFN-γ-treated primary MC/MG cultures produced significantly lower nitrite in response to LPS challenge than control MC/MG. IFN-γ-treated EAE mice had a significantly higher frequency of CX3CR1high MC/MG and expressed lower levels of program death ligand 1 (PD-L1) than PBS-treated mice. Most CX3CR1highPD-L1lowCD11b+Ly6G- cells expressed MG markers (Tmem119, Sall2, and P2ry12), indicating that they represented an enriched MG subset (CX3CR1highPD-L1low MG). Amelioration of clinical symptoms and induction of CX3CR1highPD-L1low MG by IFN-γ were dependent on STAT-1. RNA-seq analyses revealed that in vivo treatment with IFN-γ promoted the induction of homeostatic CX3CR1highPD-L1low MG, upregulating the expression of genes associated with tolerogenic and anti-inflammatory roles and down-regulating pro-inflammatory genes. These analyses highlight the master role that IFN-γ plays in regulating microglial activity and provide new insights into the cellular and molecular mechanisms involved in the therapeutic activity of IFN-γ in EAE.
Collapse
Affiliation(s)
- Juan E. Tichauer
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Gabriel Arellano
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Eric Acuña
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Luis F. González
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Nirmal R. Kannaiyan
- Molecular Neurobiology, Department of Psychiatry & Psychotherapy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Paola Murgas
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile
| | | | - Jorge Ibañez-Vega
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Paula I. Burgos
- Department of Clinical Immunology and Rheumatology , School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eileah Loda
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Stephen D. Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Moritz J. Rossner
- Molecular Neurobiology, Department of Psychiatry & Psychotherapy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Peter J. Gebicke-Haerter
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, Mannheim, Germany
| | - Rodrigo Naves
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
3
|
Cui Y, Yu H, Bu Z, Wen L, Yan L, Feng J. Focus on the Role of the NLRP3 Inflammasome in Multiple Sclerosis: Pathogenesis, Diagnosis, and Therapeutics. Front Mol Neurosci 2022; 15:894298. [PMID: 35694441 PMCID: PMC9175009 DOI: 10.3389/fnmol.2022.894298] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is initiated with an aberrant innate immune response in the central nervous system (CNS) and is involved in many neurological diseases. Inflammasomes are intracellular multiprotein complexes that can be used as platforms to induce the maturation and secretion of proinflammatory cytokines and pyroptosis, thus playing a pivotal role in neuroinflammation. Among the inflammasomes, the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasome is well-characterized and contributes to many neurological diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), and ischemic stroke. MS is a chronic autoimmune disease of the CNS, and its hallmarks include chronic inflammation, demyelination, and neurodegeneration. Studies have demonstrated a relationship between MS and the NLRP3 inflammasome. To date, the pathogenesis of MS is not fully understood, and clinical studies on novel therapies are still underway. Here, we review the activation mechanism of the NLRP3 inflammasome, its role in MS, and therapies targeting related molecules, which may be beneficial in MS.
Collapse
|
4
|
Neuroprotective Effect of Glatiramer Acetate on Neurofilament Light Chain Leakage and Glutamate Excess in an Animal Model of Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms222413419. [PMID: 34948217 PMCID: PMC8707261 DOI: 10.3390/ijms222413419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/29/2022] Open
Abstract
Axonal and neuronal pathologies are a central constituent of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), induced by the myelin oligodendrocyte glycoprotein (MOG) 35–55 peptide. In this study, we investigated neurodegenerative manifestations in chronic MOG 35–55 induced EAE and the effect of glatiramer acetate (GA) treatment on these manifestations. We report that the neuronal loss seen in this model is not attributed to apoptotic neuronal cell death. In EAE-affected mice, axonal damage prevails from the early disease phase, as revealed by analysis of neurofilament light (NFL) leakage into the sera along the disease duration, as well as by immunohistological examination. Elevation of interstitial glutamate concentrations measured in the cerebrospinal fluid (CSF) implies that glutamate excess plays a role in the damage processes inflicted by this disease. GA applied as a therapeutic regimen to mice with apparent clinical symptoms significantly reduces the pathological manifestations, namely apoptotic cell death, NFL leakage, histological tissue damage, and glutamate excess, thus corroborating the neuroprotective consequences of this treatment.
Collapse
|
5
|
Molecular Effects of FDA-Approved Multiple Sclerosis Drugs on Glial Cells and Neurons of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21124229. [PMID: 32545828 PMCID: PMC7352301 DOI: 10.3390/ijms21124229] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is characterized by peripheral and central inflammatory features, as well as demyelination and neurodegeneration. The available Food and Drug Administration (FDA)-approved drugs for MS have been designed to suppress the peripheral immune system. In addition, however, the effects of these drugs may be partially attributed to their influence on glial cells and neurons of the central nervous system (CNS). We here describe the molecular effects of the traditional and more recent FDA-approved MS drugs Fingolimod, Dimethyl Fumarate, Glatiramer Acetate, Interferon-β, Teriflunomide, Laquinimod, Natalizumab, Alemtuzumab and Ocrelizumab on microglia, astrocytes, neurons and oligodendrocytes. Furthermore, we point to a possible common molecular effect of these drugs, namely a key role for NFκB signaling, causing a switch from pro-inflammatory microglia and astrocytes to anti-inflammatory phenotypes of these CNS cell types that recently emerged as central players in MS pathogenesis. This notion argues for the need to further explore the molecular mechanisms underlying MS drug action.
Collapse
|
6
|
Musella A, Fresegna D, Rizzo FR, Gentile A, De Vito F, Caioli S, Guadalupi L, Bruno A, Dolcetti E, Buttari F, Bullitta S, Vanni V, Centonze D, Mandolesi G. 'Prototypical' proinflammatory cytokine (IL-1) in multiple sclerosis: role in pathogenesis and therapeutic targeting. Expert Opin Ther Targets 2020; 24:37-46. [PMID: 31899994 DOI: 10.1080/14728222.2020.1709823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: It has been recognized for about 20 years that interleukin (IL)-1 signaling is implicated in Multiple Sclerosis (MS), a disabling, chronic, inflammatory and neurodegenerative disease of the central nervous system (CNS). Only recently, multifaceted roles of IL-1 emerged in MS pathophysiology as a result of both clinical and preclinical studies. Notably, drugs that directly target the IL-1 system have not been tested so far in MS.Areas covered: Recent studies in animal models, together with the development of ex vivo chimeric MS models, have disclosed a critical role for IL-1 not only at the peripheral level but also within the CNS. In the present review, we highlight the IL-1-dependent neuropathological aspects of MS, by providing an overview of the cells of the immune and CNS systems that respond to IL-1 signaling, and by emphasizing the subsequent effects on the CNS, from demyelinating processes, to synaptopathy, and excitotoxicity.Expert opinion: Drugs that act on the IL-1 system show a therapeutic potential in several autoinflammatory diseases and preclinical studies have highlighted the effects of these compounds in MS. We will discuss why anti-IL-1 therapies in MS have been neglected to date.
Collapse
Affiliation(s)
- Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele University, Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Francesca Romana Rizzo
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | - Silvia Caioli
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Antonio Bruno
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Ettore Dolcetti
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Silvia Bullitta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Diego Centonze
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy.,Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele University, Rome, Italy
| |
Collapse
|
7
|
Gentile A, De Vito F, Fresegna D, Rizzo FR, Bullitta S, Guadalupi L, Vanni V, Buttari F, Stampanoni Bassi M, Leuti A, Chiurchiù V, Marfia GA, Mandolesi G, Centonze D, Musella A. Peripheral T cells from multiple sclerosis patients trigger synaptotoxic alterations in central neurons. Neuropathol Appl Neurobiol 2019; 46:160-170. [DOI: 10.1111/nan.12569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 05/22/2019] [Indexed: 01/03/2023]
Affiliation(s)
- A. Gentile
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - F. De Vito
- Unit of Neurology IRCCS Neuromed Pozzilli Italy
| | - D. Fresegna
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - F. R. Rizzo
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
| | - S. Bullitta
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - L. Guadalupi
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - V. Vanni
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - F. Buttari
- Unit of Neurology IRCCS Neuromed Pozzilli Italy
| | | | - A. Leuti
- Department of Medicine Campus Bio‐Medico University of Rome Rome Italy
- European Center for Brain Research (CERC)/IRCCS Fondazione Santa Lucia Rome Italy
| | - V. Chiurchiù
- Department of Medicine Campus Bio‐Medico University of Rome Rome Italy
- European Center for Brain Research (CERC)/IRCCS Fondazione Santa Lucia Rome Italy
| | - G. A. Marfia
- Multiple Sclerosis Research Unit Department of Systems Medicine Tor Vergata University Rome Italy
| | - G. Mandolesi
- Synaptic Immunopathology Lab IRCCS San Raffaele Pisana and University San Raffaele Rome Italy
| | - D. Centonze
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Unit of Neurology IRCCS Neuromed Pozzilli Italy
| | - A. Musella
- Synaptic Immunopathology Lab IRCCS San Raffaele Pisana and University San Raffaele Rome Italy
| |
Collapse
|
8
|
Lazo-Gomez R, Velázquez GDLLG, Mireles-Jacobo D, Sotomayor-Sobrino MA. Mechanisms of neurobehavioral abnormalities in multiple sclerosis: Contributions from neural and immune components. Clin Neurophysiol Pract 2019; 4:39-46. [PMID: 30911699 PMCID: PMC6416523 DOI: 10.1016/j.cnp.2019.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis-related neurobehavioral abnormalities are one of the main components of disability in this disease. The same pathological processes that explain demyelination periods and neurodegeneration also allow the comprehension of neurobehavioral abnormalities. Inflammation in the central nervous system caused by cells of the immune system, especially lymphocytes, and by resident cells, such as astrocytes and microglia, directly modulate neurotransmission and synaptic physiology, resulting in behavioral changes (such as sickness behavior) and amplifying the degenerative mechanisms that occur in multiple sclerosis. In addition, neuronal death caused by glutamate-mediated excitotoxicity, alterations in GABAergic, serotonergic, and dopaminergic neurotransmission, and the mechanisms of axon damage are of foremost importance to explain the reduction in brain volume and the associated cognitive decline. Neuroinflammation and neurodegeneration are not isolated phenomena and various instances of interaction between them have been described. This presents attractive targets for the development of therapeutic strategies for this neglected component of multiple sclerosis related disability.
Collapse
Affiliation(s)
- Rafael Lazo-Gomez
- Neuroscience franchise, Novartis Pharma México, Calzada de Tlalpan 1779, San Diego Churubusco, 04120 Coyoacán, CDMX, Mexico
| | | | - Diego Mireles-Jacobo
- Neuroscience franchise, Novartis Pharma México, Calzada de Tlalpan 1779, San Diego Churubusco, 04120 Coyoacán, CDMX, Mexico
| | | |
Collapse
|
9
|
Pittaluga A. CCL5-Glutamate Cross-Talk in Astrocyte-Neuron Communication in Multiple Sclerosis. Front Immunol 2017; 8:1079. [PMID: 28928746 PMCID: PMC5591427 DOI: 10.3389/fimmu.2017.01079] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/18/2017] [Indexed: 12/17/2022] Open
Abstract
The immune system (IS) and the central nervous system (CNS) are functionally coupled, and a large number of endogenous molecules (i.e., the chemokines for the IS and the classic neurotransmitters for the CNS) are shared in common between the two systems. These interactions are key elements for the elucidation of the pathogenesis of central inflammatory diseases. In recent years, evidence has been provided supporting the role of chemokines as modulators of central neurotransmission. It is the case of the chemokines CCL2 and CXCL12 that control pre- and/or post-synaptically the chemical transmission. This article aims to review the functional cross-talk linking another endogenous pro-inflammatory factor released by glial cells, i.e., the chemokine Regulated upon Activation Normal T-cell Expressed and Secreted (CCL5) and the principal neurotransmitter in CNS (i.e., glutamate) in physiological and pathological conditions. In particular, the review discusses preclinical data concerning the role of CCL5 as a modulator of central glutamatergic transmission in healthy and demyelinating disorders. The CCL5-mediated control of glutamate release at chemical synapses could be relevant either to the onset of psychiatric symptoms that often accompany the development of multiple sclerosis (MS), but also it might indirectly give a rationale for the progression of inflammation and demyelination. The impact of disease-modifying therapies for the cure of MS on the endogenous availability of CCL5 in CNS will be also summarized. We apologize in advance for omission in our coverage of the existing literature.
Collapse
Affiliation(s)
- Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
10
|
von Euler Chelpin M, Vorup-Jensen T. Targets and Mechanisms in Prevention of Parkinson's Disease through Immunomodulatory Treatments. Scand J Immunol 2017; 85:321-330. [PMID: 28231624 DOI: 10.1111/sji.12542] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 01/13/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world; however, there is no cure for it. Current treatments only relieve some of the symptoms, without ceasing the disease, and lose efficacy with prolonged treatment. Considerable evidence shows that persistent inflammatory responses, involving T cell infiltration and glial cell activation, are common characteristics of human patients and play a crucial role in the degeneration of dopaminergic neurons. Therefore, it is important to develop therapeutic strategies that can impede or halt the disease through the modulation of the peripheral immune system by aiming at controlling the existing neuroinflammation. Most of the immunomodulatory therapies designed for the treatment of Parkinson's disease are based on vaccines using AS or antibodies against it; yet, it is of significant interest to explore other formulations that could be used as therapeutic agents. Several vaccination procedures have shown that inducing regulatory T cells in the periphery is protective in PD animal models. In this regard, the formulation glatiramer acetate (Copaxone® ), extensively used for the treatment of multiple sclerosis, could be a suitable candidate due to its capability to increase the number and suppressor capacity of regulatory T cells. In this review, we will present some of the recent immunomodulatory therapies for PD including vaccinations with AS or glatiramoids, or both, as treatments of PD pathology.
Collapse
Affiliation(s)
| | - T Vorup-Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Annovazzi P, Bertolotto A, Brescia Morra V, Gasperini C, Montanari E, Navarra P, Patti F, Sormani MP, Ghezzi A. A Comprehensive Review on Copemyl ®. Neurol Ther 2017; 6:161-173. [PMID: 28762192 PMCID: PMC5700901 DOI: 10.1007/s40120-017-0079-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Indexed: 01/17/2023] Open
Abstract
Economic sustainability is of paramount importance in the rapidly evolving therapeutic scenario of multiple sclerosis (MS). Glatiramoids are a class of drugs whose forefather, glatiramer acetate, has been used as a disease modifying drug (DMD) in patients with MS for over 20 years. Its patent expired in 2015; new versions of such drug are nowadays available on the market, potentially contributing to lowering prices and enhancing a better allocation of economic resources. In this review, we analyze the recommendations underlying the approval of both generic drugs and biosimilars by regulatory authorities, and we provide methodological tools to contextualize the design of studies on these new classes of drugs. We examine in more detail the preclinical and clinical data of Copemyl®, a new member of the glatiramoid class, focusing on its biological and immunological properties and illustrating randomized controlled trials that led to its authorization.
Collapse
Affiliation(s)
- Pietro Annovazzi
- Multiple Sclerosis Study Center, ASST Valle Olona, Gallarate, VA, Italy.
| | - Antonio Bertolotto
- Neurology and Multiple Sclerosis Regional Reference Center (CRESM), AOU San Luigi, Orbassano, Italy
| | - Vincenzo Brescia Morra
- Multiple Sclerosis Clinical Care and Research Center, Departement of Neuroscience (NSRO), Federico II University, Naples, Italy
| | - Claudio Gasperini
- Multiple Sclerosis Center, S. Camillo-Forlanini Hospital Rome, Rome, Italy
| | | | - Pierluigi Navarra
- Institute of Pharmacology, Catholic University Medical School, Largo F. Vito 1, 00168, Rome, Italy
| | - Francesco Patti
- Department of Medical and Surgical Sciences, and Advanced Technologies, GF Ingrassia, Multiple Sclerosis Center, University of Catania, Catania, Italy
| | - Maria Pia Sormani
- Department of Health Sciences, Section of Biostatistics, University of Genova, Genoa, Italy
| | - Angelo Ghezzi
- Multiple Sclerosis Study Center, ASST Valle Olona, Gallarate, VA, Italy
| |
Collapse
|
12
|
Patejdl R, Zettl UK. Spasticity in multiple sclerosis: Contribution of inflammation, autoimmune mediated neuronal damage and therapeutic interventions. Autoimmun Rev 2017; 16:925-936. [PMID: 28698092 DOI: 10.1016/j.autrev.2017.07.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 12/19/2022]
Abstract
In contrast to other diseases that go along with spasticity (e.g. spinal cord injury), spasticity in chronic autoimmune diseases involving the CNS is complicated by the ongoing damage of neuronal networks that leads to permanent changes in the clinical picture of spasticity. Multiple sclerosis (MS) is the most frequent autoimmune disease of the central nervous system (CNS) and spasticity is one of the most disabling symptoms. It occurs in more than 80% MS patients at some point of the disease and is associated with impaired ambulation, pain and the development of contractures. Besides causing cumulative structural damage, neuroinflammation occurring in MS leads to dynamic changes in motor circuit function and muscle tone that are caused by cytokines, prostaglandins, reactive oxygen species and stress hormones that affect neuronal circuits and thereby spasticity. The situation is complicated further by the fact that therapeutics used for the immunotherapy of MS may worsen spasticity and drugs used for the symptomatic treatment of spasticity have been shown to have the potential to alter immune cell function and CNS autoimmunity itself. This review summarizes the current knowledge on the immunologic pathways that are involved in the development, maintenance, dynamic changes and pharmacological modulation of spasticity in MS.
Collapse
Affiliation(s)
- Robert Patejdl
- University of Rostock, Department of Physiology, Germany.
| | - Uwe K Zettl
- University of Rostock, Department of Neurology, Division of Neuroimmunology, Germany
| |
Collapse
|
13
|
Levite M. Glutamate, T cells and multiple sclerosis. J Neural Transm (Vienna) 2017; 124:775-798. [PMID: 28236206 DOI: 10.1007/s00702-016-1661-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/25/2016] [Indexed: 12/18/2022]
Abstract
Glutamate is the major excitatory neurotransmitter in the nervous system, where it induces multiple beneficial and essential effects. Yet, excess glutamate, evident in a kaleidoscope of acute and chronic pathologies, is absolutely catastrophic, since it induces excitotoxicity and massive loss of brain function. Both the beneficial and the detrimental effects of glutamate are mediated by a large family of glutamate receptors (GluRs): the ionotropic glutamate receptors (iGluRs) and the metabotropic glutamate receptors (mGluRs), expressed by most/all cells of the nervous system, and also by many non-neural cells in various peripheral organs and tissues. T cells express on their cell surface several types of functional GluRs, and so do few other immune cells. Furthermore, glutamate by itself activates resting normal human T cells, and induces/elevates key T cell functions, among them: T cell adhesion, chemotactic migration, cytokine secretion, gene expression and more. Glutamate has also potent effects on antigen/mitogen/cytokine-activated T cells. Furthermore, T cells can even produce and release glutamate, and affect other cells and themselves via their own glutamate. Multiple sclerosis (MS) and its animal model Experimental Autoimmune Encephalomyelitis (EAE) are mediated by autoimmune T cells. In MS and EAE, there are excess glutamate levels, and multiple abnormalities in glutamate degrading enzymes, glutamate transporters, glutamate receptors and glutamate signaling. Some GluR antagonists block EAE. Enhancer of mGluR4 protects from EAE via regulatory T cells (Tregs), while mGluR4 deficiency exacerbates EAE. The protective effect of mGluR4 on EAE calls for testing GluR4 enhancers in MS patients. Oral MS therapeutics, namely Fingolimod, dimethyl fumarate and their respective metabolites Fingolimod-phosphate and monomethyl fumarate, can protect neurons against acute glutamatergic excitotoxic damage. Furthermore, Fingolimod reduce glutamate-mediated intracortical excitability in relapsing-remitting MS. Glatiramer acetate -COPAXONE®, an immunomodulator drug for MS, reverses TNF-α-induced alterations of striatal glutamate-mediated excitatory postsynaptic currents in EAE-afflicted mice. With regard to T cells of MS patients: (1) The cell surface expression of a specific GluR: the AMPA GluR3 is elevated in T cells of MS patients during relapse and with active disease, (2) Glutamate and AMPA (a selective agonist for glutamate/AMPA iGluRs) augment chemotactic migration of T cells of MS patients, (3) Glutamate augments proliferation of T cells of MS patients in response to myelin-derived proteins: MBP and MOG, (4) T cells of MS patients respond abnormally to glutamate, (5) Significantly higher proliferation values in response to glutamate were found in MS patients assessed during relapse, and in those with gadolinium (Gd)+ enhancing lesions on MRI. Furthermore, glutamate released from autoreactive T cells induces excitotoxic cell death of neurons. Taken together, the evidences accumulated thus far indicate that abnormal glutamate levels and signaling in the nervous system, direct activation of T cells by glutamate, and glutamate release by T cells, can all contribute to MS. This may be true also to other neurological diseases. It is postulated herein that the detrimental activation of autoimmune T cells by glutamate in MS could lead to: (1) Cytotoxicity in the CNS: T cell-mediated killing of neurons and glia cells, which would subsequently increase the extracellular glutamate levels, and by doing so increase the excitotoxicity mediated by excess glutamate, (2) Release of proinflammatory cytokines, e.g., TNFα and IFNγ that increase neuroinflammation. Finally, if excess glutamate, abnormal neuronal signaling, glutamate-induced activation of T cells, and glutamate release by T cells are indeed all playing a key detrimental role in MS, then optional therapeutic tolls include GluR antagonists, although these may have various side effects. In addition, an especially attractive therapeutic strategy is the novel and entirely different therapeutic approach to minimize excess glutamate and excitotoxicity, titled: 'brain to blood glutamate scavenging', designed to lower excess glutamate levels in the CNS by 'pumping it out' from the brain to the blood. The glutamate scavanging is achieved by lowering glutamate levels in the blood by intravenous injection of the blood enzyme glutamate oxaloacetate transaminase (GOT). The glutamate-scavenging technology, which is still experimental, validated so far for other brain pathologies, but not tested on MS or EAE yet, may be beneficial for MS too, since it could decrease both the deleterious effects of excess glutamate on neural cells, and the activation of autoimmune T cells by glutamate in the brain. The topic of glutamate scavenging, and also its potential benefit for MS, are discussed towards the end of the review, and call for research in this direction.
Collapse
Affiliation(s)
- Mia Levite
- Faculty of Medicine, School of Pharmacy, The Hebrew University, Jerusalem, Israel. .,Institute of Gene Therapy, Hadassah Medical Center, 91120, Ein Karem, Jerusalem, Israel.
| |
Collapse
|
14
|
Expression of brain-derived neurotrophic factor in astrocytes - Beneficial effects of glatiramer acetate in the R6/2 and YAC128 mouse models of Huntington's disease. Exp Neurol 2016; 285:12-23. [PMID: 27587303 DOI: 10.1016/j.expneurol.2016.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 07/31/2016] [Accepted: 08/21/2016] [Indexed: 12/18/2022]
Abstract
Glatiramer acetate (GA) is a FDA-approved drug which is licensed for the treatment of relapsing-remitting multiple sclerosis and which may exert neuroprotective effects via brain-derived neurotrophic factor (BDNF). In this study, we investigate effects of GA on BDNF expression especially in astrocytes in vitro and in vivo in brains of R6/2 and YAC128 transgenic mouse models of Huntington's disease (HD) where a pathogenic role of astroglial cells has recently been shown. We show that GA increases the expression of functionally active BDNF in astrocyte culture and in astrocytes of GA treated HD mice. In the brains of these mice, GA decreases neurodegeneration and restores BDNF levels. The beneficial effect of GA in R6/2 mice also comprises reduced weight loss and prolonged life span and, for both models, also improved motor performance. Further studies with this safe and effective drug in HD are warranted.
Collapse
|
15
|
Gentile A, Musella A, Bullitta S, Fresegna D, De Vito F, Fantozzi R, Piras E, Gargano F, Borsellino G, Battistini L, Schubart A, Mandolesi G, Centonze D. Siponimod (BAF312) prevents synaptic neurodegeneration in experimental multiple sclerosis. J Neuroinflammation 2016; 13:207. [PMID: 27566665 PMCID: PMC5002118 DOI: 10.1186/s12974-016-0686-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/18/2016] [Indexed: 12/31/2022] Open
Abstract
Background Data from multiple sclerosis (MS) and the MS rodent model, experimental autoimmune encephalomyelitis (EAE), highlighted an inflammation-dependent synaptopathy at the basis of the neurodegenerative damage causing irreversible disability in these disorders. This synaptopathy is characterized by an imbalance between glutamatergic and GABAergic transmission and has been proposed to be a potential therapeutic target. Siponimod (BAF312), a selective sphingosine 1-phosphate1,5 receptor modulator, is currently under investigation in a clinical trial in secondary progressive MS patients. We investigated whether siponimod, in addition to its peripheral immune modulation, may exert direct neuroprotective effects in the central nervous system (CNS) of mice with chronic progressive EAE. Methods Minipumps allowing continuous intracerebroventricular (icv) infusion of siponimod for 4 weeks were implanted into C57BL/6 mice subjected to MOG35-55-induced EAE. Electrophysiology, immunohistochemistry, western blot, qPCR experiments, and peripheral lymphocyte counts were performed. In addition, the effect of siponimod on activated microglia was assessed in vitro to confirm the direct effect of the drug on CNS-resident immune cells. Results Siponimod administration (0.45 μg/day) induced a significant beneficial effect on EAE clinical scores with minimal effect on peripheral lymphocyte counts. Siponimod rescued defective GABAergic transmission in the striatum of EAE, without correcting the EAE-induced alterations of glutamatergic transmission. We observed a significant attenuation of astrogliosis and microgliosis together with reduced lymphocyte infiltration in the striatum of EAE mice treated with siponimod. Interestingly, siponimod reduced the release of IL-6 and RANTES from activated microglial cells in vitro, which might explain the reduced lymphocyte infiltration. Furthermore, the loss of parvalbumin-positive (PV+) GABAergic interneurons typical of EAE brains was rescued by siponimod treatment, providing a plausible explanation of the selective effects of this drug on inhibitory synaptic transmission. Conclusions Altogether, our results show that siponimod has neuroprotective effects in the CNS of EAE mice, which are likely independent of its peripheral immune effect, suggesting that this drug could be effective in limiting neurodegenerative pathological processes in MS.
Collapse
Affiliation(s)
- Antonietta Gentile
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Alessandra Musella
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy
| | - Silvia Bullitta
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy
| | - Diego Fresegna
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Francesca De Vito
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.,Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Roberta Fantozzi
- Unit of Neurology and Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli, IS, Italy
| | - Eleonora Piras
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia-CERC, 00143, Rome, Italy
| | - Francesca Gargano
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia-CERC, 00143, Rome, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia-CERC, 00143, Rome, Italy
| | - Anna Schubart
- Novartis Institutes of Biomedical Research, Basel, Switzerland
| | - Georgia Mandolesi
- Laboratory of Neuroimmunology and Synaptic Transmission, IRCCS Fondazione Santa Lucia, Centro Europeo di Ricerca sul Cervello (CERC), 00143, Rome, Italy.
| | - Diego Centonze
- Multiple Sclerosis Research Unit, Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy.,Unit of Neurology and Neurorehabilitation, IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077, Pozzilli, IS, Italy
| |
Collapse
|
16
|
Comi G, Amato MP, Bertolotto A, Centonze D, De Stefano N, Farina C, Gallo P, Ghezzi A, Grimaldi LM, Mancardi G, Marrosu MG, Montanari E, Patti F, Pozzilli C, Provinciali L, Salvetti M, Tedeschi G, Trojano M. The heritage of glatiramer acetate and its use in multiple sclerosis. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s40893-016-0010-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
17
|
Wen J, Ribeiro R, Tanaka M, Zhang Y. Activation of CB2 receptor is required for the therapeutic effect of ABHD6 inhibition in experimental autoimmune encephalomyelitis. Neuropharmacology 2015; 99:196-209. [DOI: 10.1016/j.neuropharm.2015.07.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/29/2015] [Accepted: 07/10/2015] [Indexed: 01/01/2023]
|
18
|
Gentile A, De Vito F, Fresegna D, Musella A, Buttari F, Bullitta S, Mandolesi G, Centonze D. Exploring the role of microglia in mood disorders associated with experimental multiple sclerosis. Front Cell Neurosci 2015; 9:243. [PMID: 26161070 PMCID: PMC4479791 DOI: 10.3389/fncel.2015.00243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
Abstract
Microglia is increasingly recognized to play a crucial role in the pathogenesis of psychiatric diseases. In particular, microglia may be the cellular link between inflammation and behavioral alterations: by releasing a number of soluble factors, among which pro-inflammatory cytokines, that can regulate synaptic activity, thereby leading to perturbation of behavior. In multiple sclerosis (MS), the most common neuroinflammatory disorder affecting young adults, microglia activation and dysfunction may account for mood symptoms, like depression and anxiety, that are often diagnosed in patients even in the absence of motor disability. Behavioral studies in experimental autoimmune encephalomyelitis (EAE), the animal model of MS, have shown that emotional changes occur early in the disease and in correlation to inflammatory mediator and neurotransmitter level alterations. However, such studies lack a full and comprehensive analysis of the role played by microglia in EAE-behavioral syndrome. We review the experimental studies addressing behavioral symptoms in EAE, and propose the study of neuron-glia interaction as a powerful but still poorly explored tool to investigate the burden of microglia in mood alterations associated to MS.
Collapse
Affiliation(s)
- Antonietta Gentile
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy ; Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy
| | - Francesca De Vito
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy ; Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy
| | - Diego Fresegna
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy ; Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy
| | - Alessandra Musella
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy
| | - Fabio Buttari
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy ; IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed Pozzilli, Italy
| | - Silvia Bullitta
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy
| | - Georgia Mandolesi
- Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC) Rome, Italy
| | - Diego Centonze
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata Rome, Italy ; IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed Pozzilli, Italy
| |
Collapse
|
19
|
LoPresti P. Glatiramer acetate guards against rapid memory decline during relapsing-remitting experimental autoimmune encephalomyelitis. Neurochem Res 2015; 40:473-9. [PMID: 25481047 DOI: 10.1007/s11064-014-1491-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/18/2014] [Accepted: 11/26/2014] [Indexed: 01/09/2023]
Abstract
Cognitive decline presents a therapeutic challenge for patients with multiple sclerosis (MS), a disease characterized by recurrent autoimmune demyelination and by progressive CNS degeneration. Glatiramer acetate (GA, also known as Copolymer 1, Cop-1, or Copaxone), commonly used to treat MS, reduces the frequency of relapses; it has both anti-inflammatory and neuroprotective properties. However, clinical trials have not definitively shown that GA improves cognitive impairment during MS. Using an in vivo animal model of autoimmune demyelination, i.e., relapsing-remitting experimental autoimmune encephalomyelitis (EAE), we tested short-term memory in EAE mice (EAE), in EAE mice treated with GA for 10 days starting at the time of immunization (EAE + GA), and in age-matched healthy, naïve mice (Naïve). Short-term memory was assessed using the cross-maze test at 10, 20, and 30 days post-immunization (d.p.i.); data were analyzed at each time point and over time. At 10 d.p.i., EAE and EAE + GA mice had better memory function than Naïve mice. However, at the later time points, EAE mice had a steep negative slope of memory function (indicating decline), whereas EAE + GA mice had a flatter, less-negative slope of memory function. Notably, the memory function of EAE mice significantly decreased over time compared with that of Naïve mice, indicating that EAE had a negative impact on cognitive ability. In contrast, there was no statistically significant difference between the slopes of memory function in mice with EAE treated with GA versus Naïve mice, which revealed effective, albeit partial, protection by GA treatment against progressive memory decline during EAE disease. Of particular interest, although EAE mice had memory decline over 30 d.p.i., their clinical disease scores improved during that time. Thus, our results suggest that EAE mice had a significant progressive memory decline and that GA, administered at the time of immunization, partially guards against rapid memory decline.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA,
| |
Collapse
|
20
|
Liu S, Zwinger P, Black J, Waxman S. Tapered withdrawal of phenytoin removes protective effect in EAE without inflammatory rebound and mortality. J Neurol Sci 2014; 341:8-12. [DOI: 10.1016/j.jns.2014.03.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 11/29/2022]
|
21
|
Giunti D, Parodi B, Cordano C, Uccelli A, Kerlero de Rosbo N. Can we switch microglia's phenotype to foster neuroprotection? Focus on multiple sclerosis. Immunology 2014; 141:328-39. [PMID: 24116890 DOI: 10.1111/imm.12177] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 09/30/2013] [Indexed: 12/17/2022] Open
Abstract
Microglia cells, the resident innate immune cells in the brain, are highly active, extending and retracting highly motile processes through which they continuously survey their microenvironment for 'danger signals' and interact dynamically with surrounding cells. Upon sensing changes in their central nervous system microenvironment, microglia become activated, undergoing morphological and functional changes. Microglia activation is not an 'all-or-none' process, but rather a continuum depending on encountered stimuli, which is expressed through a spectrum of molecular and functional phenotypes ranging from so-called 'classically activated', with a highly pro-inflammatory profile, to 'alternatively activated' associated with a beneficial, less inflammatory, neuroprotective profile. Microglia activation has been demonstrated in most neurological diseases of diverse aetiology and has been implicated as a contributor to neurodegeneration. The possibility to promote microglia's neuroprotective phenotype has therefore become a therapeutic goal. We have focused our discussion on the role of microglia in multiple sclerosis, a prototype of inflammatory, demyelinating, neurodegenerative disease, and on the effect of currently approved or on-trial anti-inflammatory therapeutic strategies that might mediate neuroprotection at least in part through their effect on microglia by modifying their behaviour via a switch of their functional phenotype from a detrimental to a protective one. In addition to pharmaceutical approaches, such as treatment with glatiramer acetate, interferon-β, fingolimod or dimethyl fumarate, we address the alternative therapeutic approach of treatment with mesenchymal stem cells and their potential role in neuroprotection through their 'calming' effect on microglia.
Collapse
Affiliation(s)
- Debora Giunti
- Department of Neurosciences, Ophthalmology, Genetics, Rehabilitation and Child Health, University of Genoa, Genoa, Italy
| | | | | | | | | |
Collapse
|