1
|
Aghaee F, Abedinpour M, Anvari S, Saberi A, Fallah A, Bakhshi A. Natural killer cells in multiple sclerosis: foe or friends? Front Cell Neurosci 2025; 19:1500770. [PMID: 40255388 PMCID: PMC12006147 DOI: 10.3389/fncel.2025.1500770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disorder involving the central nervous system (CNS), in which demyelination is caused. The initiation and progression of MS is thought to depend largely on CD4+ T lymphocytes, yet new data has emphasized the involvement of the innate immune system in the MS disease responses. Generally, several types of immune cells play a part, with natural killer (NK) cells being essential. Different subsets of natural killer cells function differently within the course of an autoimmune disease, such as MS. There are mainly two types of natural killers in humans: immature CD56 bright CD16- and mature CD56 dim CD16+ natural killers, together with their respective subtypes. Factors from natural killers expand the T cell population and control the process by which native CD4+ T cells differentiate into Th1 or Th2 lymphocytes, which affect autoimmune responses. Natural killer subsets CD56 bright and CD56 dim may have differing roles in MS development. The impact of these NK cell subsets is influenced by factors such as Granzymes, genetics, infections, TLR, and HSP. We reviewed and evaluated the relationship between natural killer cells and MS.
Collapse
Affiliation(s)
- Fatemeh Aghaee
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadreza Abedinpour
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeid Anvari
- Department of Neurology, Neurosciences Research Center, Poursina Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Alia Saberi
- Department of Internal Medicine, Regenerative Medicine Research Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amir Fallah
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arash Bakhshi
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
2
|
Bergner CG, van der Meer F, Franz J, Vakrakou A, Würfel T, Nessler S, Schäfer L, Nau-Gietz C, Winkler A, Lagumersindez-Denis N, Wrzos C, Damkou IA, Sergiou C, Schultz V, Knauer C, Metz I, Bahn E, Garea Rodriguez E, Merkler D, Simons M, Stadelmann C. BCAS1-positive oligodendrocytes enable efficient cortical remyelination in multiple sclerosis. Brain 2025; 148:908-920. [PMID: 39319704 PMCID: PMC11884765 DOI: 10.1093/brain/awae293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 09/26/2024] Open
Abstract
Remyelination is a crucial regenerative process in demyelinating diseases, limiting persisting damage to the CNS. It restores saltatory nerve conduction and ensures trophic support of axons. In patients with multiple sclerosis, remyelination has been observed in both white and grey matter and found to be more efficient in the cortex. Brain-enriched myelin-associated protein 1 (BCAS1) identifies oligodendrocyte lineage cells in the stage of active myelin formation in development and regeneration. Other than in the white matter, BCAS1+ oligodendrocytes are maintained at high densities in the cortex throughout life. Here, we investigated cortical lesions in human biopsy and autopsy tissue from patients with multiple sclerosis in direct comparison to demyelinating mouse models and demonstrate that following a demyelinating insult BCAS1+ oligodendrocytes in remyelinating cortical lesions shift from a quiescent to an activated, internode-forming morphology co-expressing myelin-associated glycoprotein (MAG), necessary for axonal contact formation. Of note, activated BCAS1+ oligodendrocytes are found at early time points of experimental demyelination amidst ongoing inflammation. In human tissue, activated BCAS1+ oligodendrocytes correlate with the density of myeloid cells, further supporting their involvement in an immediate regenerative response. Furthermore, studying the microscopically normal appearing non demyelinated cortex in patients with chronic multiple sclerosis, we find a shift from quiescent BCAS1+ oligodendrocytes to mature, myelin-maintaining oligodendrocytes, suggesting oligodendrocyte differentiation and limited replenishment of BCAS1+ oligodendrocytes in long-standing disease. We also demonstrate that part of perineuronal satellite oligodendrocytes are BCAS1+ and contribute to remyelination in human and experimental cortical demyelination. In summary, our results provide evidence from human tissue and experimental models that BCAS1+ cells in the adult cortex represent a population of pre-differentiated oligodendrocytes that rapidly react after a demyelinating insult thus enabling immediate myelin regeneration. In addition, our data suggest that limited replenishment of BCAS1+ oligodendrocytes may contribute to the remyelination failure observed in the cortex in chronic multiple sclerosis.
Collapse
Affiliation(s)
- Caroline Gertrud Bergner
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Franziska van der Meer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Jonas Franz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Aigli Vakrakou
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens 11528, Greece
| | - Thea Würfel
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Stefan Nessler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Lisa Schäfer
- Department of Neurology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Cora Nau-Gietz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Anne Winkler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | | | - Claudia Wrzos
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Ioanna Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, Munich 81377, Germany
| | - Christina Sergiou
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Verena Schultz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Carolin Knauer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Imke Metz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Erik Bahn
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | | | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva 4 1211, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva 1205, Switzerland
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich 81377, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich 81377, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Network of Excitable Cells”(MBExC), University of Göttingen, Göttingen 37073, Germany
| |
Collapse
|
3
|
Lohrberg M, Mortensen LS, Thomas C, Fries F, van der Meer F, Götz A, Landt C, Rhee HJ, Rhee J, Gómez-Varela D, Schmidt M, Möbius W, Ruhwedel T, Pardo LA, Remling L, Kramann N, Wrzos C, Bahn E, Stadelmann C, Barrantes-Freer A. Astroglial modulation of synaptic function in the non-demyelinated cerebellar cortex is dependent on MyD88 signaling in a model of toxic demyelination. J Neuroinflammation 2025; 22:47. [PMID: 39988657 PMCID: PMC11849172 DOI: 10.1186/s12974-025-03368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/03/2025] [Indexed: 02/25/2025] Open
Abstract
Progressive neurological decline in multiple sclerosis is associated with axonal loss and synaptic dysfunction in the non-demyelinated normal appearing gray matter (NAGM) and prominently in the cerebellum. In contrast to early disease stages, where synaptic and neuro-axonal pathology correlates with the extent of T cell infiltration, a prominent role of the innate immune system has been proposed for progressive MS. However, the specific contribution of microglia and astrocytes to synaptic cerebellar pathology in the NAGM- independent of an adaptive T cell response - remains largely unexplored. In the present study, we quantified synaptic changes in the cerebellar NAGM distant from demyelinated lesions in a mouse model of toxic demyelination. Proteomic analysis of the cerebellar cortex revealed differential regulation of synaptic and glutamate transport proteins in the absence of evident structural synaptic pathology or local gray matter demyelination. At the functional level, synaptic changes manifested as a reduction in frequency-dependent facilitation at the parallel fiber- Purkinje cell synapse. Further, deficiency of MyD88, an adaptor protein of the innate immune response, associated with a functional recovery in facilitation, reduced changes in the differential expression of synaptic and glutamate transport proteins, and reduced transcription levels of inflammatory cytokines. Nevertheless, the characteristics of demyelinating lesions and their associated cellular response were similar to wild type animals. Our work brings forward an experimental paradigm mimicking the diffuse synaptic pathology independent of demyelination in late stage MS and highlights the complex regulation of synaptic pathology in the cerebellar NAGM. Moreover, our findings suggest a role of astrocytes, in particular Bergmann glia, as key cellular determinants of cerebellar synaptic dysfunction.
Collapse
Affiliation(s)
- Melanie Lohrberg
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Campus Institute Data Science, Göttingen, Germany
| | - Lena Sünke Mortensen
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany
| | - Carolina Thomas
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany
| | - Franziska Fries
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Alexander Götz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Carolin Landt
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Hong Jun Rhee
- Department of Molecular Neurobiology, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - David Gómez-Varela
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Manuela Schmidt
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Wiebke Möbius
- Department of Neurogenetics, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Luis A Pardo
- Oncophysiology Group, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Linus Remling
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Nadine Kramann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Claudia Wrzos
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Erik Bahn
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Alonso Barrantes-Freer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany.
| |
Collapse
|
4
|
Silva BA, Leal MC, Farias MI, Nava A, Galván DI, Fernandez E, Pitossi FJ, Ferrari CC. Proteomic analysis reveals candidate molecules to mediate cortical pathology and identify possible biomarkers in an animal model of multiple sclerosis. Front Immunol 2025; 16:1505459. [PMID: 40018028 PMCID: PMC11864942 DOI: 10.3389/fimmu.2025.1505459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/15/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Multiple Sclerosis (MS) is a complex neurodegenerative disease marked by recurring inflammatory episodes, demyelination, axonal damage, and subsequent loss of function. MS presents a wide range of clinical courses, with the progressive forms leading to irreversible neurological disability. Cortical demyelinating lesions are central to the pathology of these progressive forms, gaining critical importance in recent decades due to their strong correlation with physical disability and cognitive decline. Despite this, the underlying mechanisms driving cortical lesion formation remain poorly understood, and no specific treatments are currently available. A significant challenge lies in the lack of animal models that accurately mirror the key characteristics of these lesions. Methods We developed a focal cortical animal model that replicates many features of cortical lesions, including cognitive impairment. This study focuses on conducting proteomic analyses of both the cortical lesions and cerebrospinal fluid (CSF) from these animals, aiming to identify key proteins and biomarkers that could be validated in MS patients. Results Proteomic differences between frontal cortex tissue and CSF were observed when comparing experimental animals with controls. Among the identified proteins, some have been previously described in MS patients and animal models, while others represent novel discoveries. Notably, we identified two proteins, S100A8 and orosomucoid-1, that were highly expressed in both regions. Conclusions These findings suggest that the prognostic molecules identified in this model could facilitate the discovery of new biomarkers or key molecules relevant to MS, particularly in the cortical lesion that mainly characterized the progressive forms of the disease.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Fundación Instituto Leloir (FIL), Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Celeste Leal
- Fundación Instituto Leloir (FIL), Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Isabel Farias
- Fundación Instituto Leloir (FIL), Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Agustín Nava
- Fundación Instituto Leloir (FIL), Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Fundación Huésped, Buenos Aires, Argentina
| | - Daniela Inés Galván
- Fundación Instituto Leloir (FIL), Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Elmer Fernandez
- ScireLab, Fundación para el Progreso de la Medicina, CONICET, Córdoba, Argentina
- Facultad de Ciencias Exactas, Físicas y Naturales (FCEFyN), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Fernando Juan Pitossi
- Fundación Instituto Leloir (FIL), Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Fundación Instituto Leloir (FIL), Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
5
|
Harrison DM, Sati P, Klawiter EC, Narayanan S, Bagnato F, Beck ES, Barker P, Calvi A, Cagol A, Donadieu M, Duyn J, Granziera C, Henry RG, Huang SY, Hoff MN, Mainero C, Ontaneda D, Reich DS, Rudko DA, Smith SA, Trattnig S, Zurawski J, Bakshi R, Gauthier S, Laule C. The use of 7T MRI in multiple sclerosis: review and consensus statement from the North American Imaging in Multiple Sclerosis Cooperative. Brain Commun 2024; 6:fcae359. [PMID: 39445084 PMCID: PMC11497623 DOI: 10.1093/braincomms/fcae359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/28/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
The use of ultra-high-field 7-Tesla (7T) MRI in multiple sclerosis (MS) research has grown significantly over the past two decades. With recent regulatory approvals of 7T scanners for clinical use in 2017 and 2020, the use of this technology for routine care is poised to continue to increase in the coming years. In this context, the North American Imaging in MS Cooperative (NAIMS) convened a workshop in February 2023 to review the previous and current use of 7T technology for MS research and potential future research and clinical applications. In this workshop, experts were tasked with reviewing the current literature and proposing a series of consensus statements, which were reviewed and approved by the NAIMS. In this review and consensus paper, we provide background on the use of 7T MRI in MS research, highlighting this technology's promise for identification and quantification of aspects of MS pathology that are more difficult to visualize with lower-field MRI, such as grey matter lesions, paramagnetic rim lesions, leptomeningeal enhancement and the central vein sign. We also review the promise of 7T MRI to study metabolic and functional changes to the brain in MS. The NAIMS provides a series of consensus statements regarding what is currently known about the use of 7T MRI in MS, and additional statements intended to provide guidance as to what work is necessary going forward to accelerate 7T MRI research in MS and translate this technology for use in clinical practice and clinical trials. This includes guidance on technical development, proposals for a universal acquisition protocol and suggestions for research geared towards assessing the utility of 7T MRI to improve MS diagnostics, prognostics and therapeutic efficacy monitoring. The NAIMS expects that this article will provide a roadmap for future use of 7T MRI in MS.
Collapse
Affiliation(s)
- Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Neurology, Baltimore VA Medical Center, Baltimore, MD 21201, USA
| | - Pascal Sati
- Neuroimaging Program, Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Eric C Klawiter
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Montreal Neurological Institute-Hospital, Montreal, QC, Canada, H3A 2B4
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada, H3A 2B4
| | - Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Department of Neurology, Nashville VA Medical Center, TN Valley Healthcare System, Nashville, TN 37212, USA
| | - Erin S Beck
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter Barker
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alberto Calvi
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Alessandro Cagol
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, 4001 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, 4001 Basel, Switzerland
- Department of Health Sciences, University of Genova, 16132 Genova, Italy
| | - Maxime Donadieu
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeff Duyn
- Advanced MRI Section, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cristina Granziera
- Translational Imaging in Neurology (ThINk) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel, University of Basel, 4001 Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, 4001 Basel, Switzerland
- Department of Neurology, University Hospital Basel, 4001 Basel, Switzerland
| | - Roland G Henry
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Susie Y Huang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - Michael N Hoff
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA
| | - Caterina Mainero
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02114, USA
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A Rudko
- McConnell Brain Imaging Centre, Montreal Neurological Institute-Hospital, Montreal, QC, Canada, H3A 2B4
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada, H3A 2B4
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Sciences, Vanderbilt University, Nashville, TN 37212, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN 37212, USA
| | - Siegfried Trattnig
- Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Jonathan Zurawski
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rohit Bakshi
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Susan Gauthier
- Department of Neurology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Cornelia Laule
- Radiology, Pathology and Laboratory Medicine, Physics and Astronomy, International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada, BC V6T 1Z4
| |
Collapse
|
6
|
Baraibar AM, Colomer T, Moreno-García A, Bernal-Chico A, Sánchez-Martín E, Utrilla C, Serrat R, Soria-Gómez E, Rodríguez-Antigüedad A, Araque A, Matute C, Marsicano G, Mato S. Autoimmune inflammation triggers aberrant astrocytic calcium signaling to impair synaptic plasticity. Brain Behav Immun 2024; 121:192-210. [PMID: 39032542 PMCID: PMC11415231 DOI: 10.1016/j.bbi.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
Cortical pathology involving inflammatory and neurodegenerative mechanisms is a hallmark of multiple sclerosis and a correlate of disease progression and cognitive decline. Astrocytes play a pivotal role in multiple sclerosis initiation and progression but astrocyte-neuronal network alterations contributing to gray matter pathology remain undefined. Here we unveil deregulation of astrocytic calcium signaling and astrocyte-to-neuron communication as key pathophysiological mechanisms of cortical dysfunction in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis. Using two-photon imaging ex vivo and fiber photometry in freely behaving mice, we found that acute EAE was associated with the emergence of spontaneously hyperactive cortical astrocytes exhibiting dysfunctional responses to cannabinoid, glutamate and purinoreceptor agonists. Abnormal astrocyte signaling by Gi and Gq protein coupled receptors was observed in the inflamed cortex. This was mirrored by treatments with pro-inflammatory factors both in vitro and ex vivo, suggesting cell-autonomous effects of the cortical neuroinflammatory environment. Finally, deregulated astrocyte calcium activity was associated with an enhancement of glutamatergic gliotransmission and a shift of astrocyte-mediated short-term and long-term plasticity mechanisms towards synaptic potentiation. Overall, our data identify astrocyte-neuronal network dysfunctions as key pathological features of gray matter inflammation in multiple sclerosis and potentially additional neuroimmunological disorders.
Collapse
Affiliation(s)
- A M Baraibar
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - T Colomer
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - A Moreno-García
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - A Bernal-Chico
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - E Sánchez-Martín
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - C Utrilla
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - R Serrat
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - E Soria-Gómez
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
| | - A Rodríguez-Antigüedad
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - A Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, 55455 MN, USA
| | - C Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - G Marsicano
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France.
| | - S Mato
- Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Neuroinmunology Group, Biobizkaia Health Research Institute, 48903 Barakaldo, Spain.
| |
Collapse
|
7
|
Ding J, Yan X, Zhao C, Zhao D, Jia Y, Ren K, Wang Y, Lu J, Sun T, Zhao S, Li H, Guo J. The ratio of circulating CD56 dim NK cells to follicular T helper cells as a promising predictor for disease activity of relapsing-remitting multiple sclerosis. Heliyon 2024; 10:e31533. [PMID: 38803865 PMCID: PMC11128518 DOI: 10.1016/j.heliyon.2024.e31533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system primarily mediated by CD4+ T helper cells. This study investigated the dynamic changes of natural killer (NK) cells and follicular T helper (Tfh) cells and their associations in relapsing-remitting MS patients. The findings revealed inverse relationships between NK cells and CD4+ T cells or Tfh cells. Specifically, CD56dim NK cells, not CD56bright NK cells, were negatively correlated with CD4+ T cells and Tfh cells. However, no significant correlations were found between NK cells and sNfL levels or EDSS scores. The ratio of CD56dim NK cells to circulating Tfh (cTfh) cells demonstrated superior discriminatory ability in distinguishing relapsing MS patients from healthy controls (HCs) and remitting patients, as determined by receiver operating characteristic (ROC) analysis. Following treatment with immunosuppressants or disease-modifying therapies (DMTs), a significant increase in the CD56dim NK/cTfh ratio was observed. These findings suggest that the CD56dim NK/cTfh ratio holds promise as a prognostic indicator for clinical relapse and treatment response in MS.
Collapse
Affiliation(s)
- Jiaqi Ding
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Xu Yan
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Cong Zhao
- Department of Neurology, Air Force Medical Center of PLA, Beijing, 100142, China
| | - Daidi Zhao
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yan Jia
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Kaixi Ren
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yao Wang
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jiarui Lu
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Tangna Sun
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Sijia Zhao
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Hongzeng Li
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Jun Guo
- Department of Neurology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| |
Collapse
|
8
|
Barakovic M, Weigel M, Cagol A, Schaedelin S, Galbusera R, Lu PJ, Chen X, Melie-Garcia L, Ocampo-Pineda M, Bahn E, Stadelmann C, Palombo M, Kappos L, Kuhle J, Magon S, Granziera C. A novel imaging marker of cortical "cellularity" in multiple sclerosis patients. Sci Rep 2024; 14:9848. [PMID: 38684744 PMCID: PMC11059177 DOI: 10.1038/s41598-024-60497-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
Pathological data showed focal inflammation and regions of diffuse neuronal loss in the cortex of people with multiple sclerosis (MS). In this work, we applied a novel model ("soma and neurite density imaging (SANDI)") to multishell diffusion-weighted MRI data acquired in healthy subjects and people with multiple sclerosis (pwMS), in order to investigate inflammation and degeneration-related changes in the cortical tissue of pwMS. We aimed to (i) establish whether SANDI is applicable in vivo clinical data; (ii) investigate inflammatory and degenerative changes using SANDI soma fraction (fsoma)-a marker of cellularity-in both cortical lesions and in the normal-appearing-cortex and (iii) correlate SANDI fsoma with clinical and biological measures in pwMS. We applied a simplified version of SANDI to a clinical scanners. We then provided evidence that pwMS exhibited an overall decrease in cortical SANDI fsoma compared to healthy subjects, suggesting global degenerative processes compatible with neuronal loss. On the other hand, we have found that progressive pwMS showed a higher SANDI fsoma in the outer part of the cortex compared to relapsing-remitting pwMS, possibly supporting current pathological knowledge of increased innate inflammatory cells in these regions. A similar finding was obtained in subpial lesions in relapsing-remitting patients, reflecting existing pathological data in these lesion types. A significant correlation was found between SANDI fsoma and serum neurofilament light chain-a biomarker of inflammatory axonal damage-suggesting a relationship between SANDI soma fraction and inflammatory processes in pwMS again. Overall, our data show that SANDI fsoma is a promising biomarker to monitor changes in cellularity compatible with neurodegeneration and neuroinflammation in the cortex of MS patients.
Collapse
Affiliation(s)
- Muhamed Barakovic
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Matthias Weigel
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Alessandro Cagol
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Sabine Schaedelin
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Riccardo Galbusera
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Po-Jui Lu
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Xinjie Chen
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Lester Melie-Garcia
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Mario Ocampo-Pineda
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Erik Bahn
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | | | - Marco Palombo
- School of Psychology, Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK
- School of Computer Science and Informatics, Cardiff University, Cardiff, UK
| | - Ludwig Kappos
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Stefano Magon
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Cristina Granziera
- Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, Faculty of Medicine, University Hospital Basel and University of Basel, Basel, Switzerland.
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland.
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
9
|
Yandamuri SS, Filipek B, Lele N, Cohen I, Bennett JL, Nowak RJ, Sotirchos ES, Longbrake EE, Mace EM, O’Connor KC. A Noncanonical CD56dimCD16dim/- NK Cell Subset Indicative of Prior Cytotoxic Activity Is Elevated in Patients with Autoantibody-Mediated Neurologic Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:785-800. [PMID: 38251887 PMCID: PMC10932911 DOI: 10.4049/jimmunol.2300015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein Ab disease, and autoimmune myasthenia gravis (MG) are autoantibody-mediated neurologic conditions where autoantibodies can induce Ab-dependent cellular cytotoxicity (ADCC), a NK cell-mediated effector function. However, whether ADCC is a pathogenic mechanism in patients with these conditions has not been confirmed. We sought to characterize circulatory NK cells using functional assays, phenotyping, and transcriptomics to elucidate their role in pathology. NK cells from NMOSD patients and MG patients with elevated disease burden exhibited reduced ADCC and CD56dimCD16hi NK cells, along with an elevated frequency of CD56dimCD16dim/- NK cells. We determined that ADCC induces a similar phenotypic shift in vitro. Bulk RNA sequencing distinguished the CD56dimCD16dim/- population from the canonical CD56dimCD16hi cytotoxic and CD56hiCD16- immunomodulatory subsets, as well as CD56hiCD16+ NK cells. Multiparameter immunophenotyping of NK cell markers, functional proteins, and receptors similarly showed that the CD56dimCD16dim/- subset exhibits a unique profile while still maintaining expression of characteristic NK markers CD56, CD94, and NKp44. Notably, expression of perforin and granzyme is reduced in comparison with CD56dimCD16hi NK cells. Moreover, they exhibit elevated trogocytosis capability, HLA-DR expression, and many chemokine receptors, including CCR7. In contrast with NMOSD and MG, myelin oligodendrocyte glycoprotein Ab disease NK cells did not exhibit functional, phenotypic, or transcriptomic perturbations. In summary, CD56dimCD16dim/- NK cells are a distinct peripheral blood immune cell population in humans elevated upon prior cytotoxic activity by the CD56dimCD16hi NK cell subset. The elevation of this subset in NMOSD and MG patients suggests prior ADCC activity.
Collapse
Affiliation(s)
- Soumya S. Yandamuri
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, United States
| | - Beata Filipek
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, United States
- Department of Pharmaceutical Microbiology and Biochemistry, Medical University of Lodz; Lodz, Poland
| | - Nikhil Lele
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Inessa Cohen
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Jeffrey L. Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado School of Medicine, Anschutz Medical Campus; Aurora, CO, United States
| | - Richard J. Nowak
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Elias S. Sotirchos
- Department of Neurology, Johns Hopkins University; Baltimore, MD, United States
| | - Erin E. Longbrake
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
| | - Emily M. Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center; New York, NY, United States
| | - Kevin C. O’Connor
- Department of Neurology, Yale School of Medicine; New Haven, CT, United States
- Department of Immunobiology, Yale School of Medicine; New Haven, CT, United States
| |
Collapse
|
10
|
Sarkar SK, Willson AML, Jordan MA. The Plasticity of Immune Cell Response Complicates Dissecting the Underlying Pathology of Multiple Sclerosis. J Immunol Res 2024; 2024:5383099. [PMID: 38213874 PMCID: PMC10783990 DOI: 10.1155/2024/5383099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative autoimmune disease characterized by the destruction of the myelin sheath of the neuronal axon in the central nervous system. Many risk factors, including environmental, epigenetic, genetic, and lifestyle factors, are responsible for the development of MS. It has long been thought that only adaptive immune cells, especially autoreactive T cells, are responsible for the pathophysiology; however, recent evidence has indicated that innate immune cells are also highly involved in disease initiation and progression. Here, we compile the available data regarding the role immune cells play in MS, drawn from both human and animal research. While T and B lymphocytes, chiefly enhance MS pathology, regulatory T cells (Tregs) may serve a more protective role, as can B cells, depending on context and location. Cells chiefly involved in innate immunity, including macrophages, microglia, astrocytes, dendritic cells, natural killer (NK) cells, eosinophils, and mast cells, play varied roles. In addition, there is evidence regarding the involvement of innate-like immune cells, such as γδ T cells, NKT cells, MAIT cells, and innate-like B cells as crucial contributors to MS pathophysiology. It is unclear which of these cell subsets are involved in the onset or progression of disease or in protective mechanisms due to their plastic nature, which can change their properties and functions depending on microenvironmental exposure and the response of neural networks in damage control. This highlights the need for a multipronged approach, combining stringently designed clinical data with carefully controlled in vitro and in vivo research findings, to identify the underlying mechanisms so that more effective therapeutics can be developed.
Collapse
Affiliation(s)
- Sujan Kumar Sarkar
- Department of Anatomy, Histology and Physiology, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University, Dhaka, Bangladesh
| | - Annie M. L. Willson
- Biomedical Sciences and Molecular Biology, CPHMVS, James Cook University, Townsville, Queensland 4811, Australia
| | - Margaret A. Jordan
- Biomedical Sciences and Molecular Biology, CPHMVS, James Cook University, Townsville, Queensland 4811, Australia
| |
Collapse
|
11
|
Ning Z, Liu Y, Guo D, Lin WJ, Tang Y. Natural killer cells in the central nervous system. Cell Commun Signal 2023; 21:341. [PMID: 38031097 PMCID: PMC10685650 DOI: 10.1186/s12964-023-01324-9] [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: 07/18/2023] [Accepted: 09/17/2023] [Indexed: 12/01/2023] Open
Abstract
Natural killer (NK) cells are essential components of the innate lymphoid cell family that work as both cytotoxic effectors and immune regulators. Accumulating evidence points to interactions between NK cells and the central nervous system (CNS). Here, we review the basic knowledge of NK cell biology and recent advances in their roles in the healthy CNS and pathological conditions, with a focus on normal aging, CNS autoimmune diseases, neurodegenerative diseases, cerebrovascular diseases, and CNS infections. We highlight the crosstalk between NK cells and diverse cell types in the CNS and the potential value of NK cells as novel therapeutic targets for CNS diseases. Video Abstract.
Collapse
Affiliation(s)
- Zhiyuan Ning
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ying Liu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Daji Guo
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
12
|
Diebold M, Fehrenbacher L, Frosch M, Prinz M. How myeloid cells shape experimental autoimmune encephalomyelitis: At the crossroads of outside-in immunity. Eur J Immunol 2023; 53:e2250234. [PMID: 37505465 DOI: 10.1002/eji.202250234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/21/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of central nervous system (CNS) autoimmunity. It is most commonly used to mimic aspects of multiple sclerosis (MS), a demyelinating disorder of the human brain and spinal cord. The innate immune response displays one of the core pathophysiological features linked to both the acute and chronic stages of MS. Hence, understanding and targeting the innate immune response is essential. Microglia and other CNS resident MUs, as well as infiltrating myeloid cells, diverge substantially in terms of both their biology and their roles in EAE. Recent advances in the field show that antigen presentation, as well as disease-propagating and regulatory interactions with lymphocytes, can be attributed to specific myeloid cell types and cell states in EAE lesions, following a distinct temporal pattern during disease initiation, propagation and recovery. Furthermore, single-cell techniques enable the assessment of characteristic proinflammatory as well as beneficial cell states, and identification of potential treatment targets. Here, we discuss the principles of EAE induction and protocols for varying experimental paradigms, the composition of the myeloid compartment of the CNS during health and disease, and systematically review effects on myeloid cells for therapeutic approaches in EAE.
Collapse
Affiliation(s)
- Martin Diebold
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Luca Fehrenbacher
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Maximilian Frosch
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Enz LS, Winkler A, Wrzos C, Dasen B, Nessler S, Stadelmann C, Schaeren-Wiemers N. An Animal Model for Chronic Meningeal Inflammation and Inflammatory Demyelination of the Cerebral Cortex. Int J Mol Sci 2023; 24:13893. [PMID: 37762198 PMCID: PMC10531364 DOI: 10.3390/ijms241813893] [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: 06/30/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Modeling chronic cortical demyelination allows the study of long-lasting pathological changes observed in multiple sclerosis such as failure of remyelination, chronically disturbed functions of oligodendrocytes, neurons and astrocytes, brain atrophy and cognitive impairments. We aimed at generating an animal model for studying the consequences of chronic cortical demyelination and meningeal inflammation. To induce long-lasting cortical demyelination and chronic meningeal inflammation, we immunized female Lewis rats against myelin oligodendrocyte glycoprotein (MOG) and injected lentiviruses for continuing overexpression of the cytokines TNFα and IFNγ in the cortical brain parenchyma. Immunization with MOG and overexpression of TNFα and IFNγ led to widespread subpial demyelination and meningeal inflammation that were stable for at least 10 weeks. We demonstrate here that immunization with MOG is necessary for acute as well as chronic cortical demyelination. In addition, long-lasting overexpression of TNFα and IFNγ in the brain parenchyma is sufficient to induce chronic meningeal inflammation. Our model simulates key features of chronic cortical demyelination and inflammation, reminiscent of human multiple sclerosis pathology. This will allow molecular, cellular and functional investigations for a better understanding of the adaptation mechanisms of the cerebral cortex in multiple sclerosis.
Collapse
Affiliation(s)
- Lukas Simon Enz
- Neurobiology, Department of Biomedicine, University Hospital Basel, University Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland;
| | - Anne Winkler
- Institute of Neuropathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (A.W.); (S.N.); (C.S.)
| | - Claudia Wrzos
- Institute of Neuropathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (A.W.); (S.N.); (C.S.)
| | - Boris Dasen
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University Basel, CH-4031 Basel, Switzerland;
| | - Stefan Nessler
- Institute of Neuropathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (A.W.); (S.N.); (C.S.)
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (A.W.); (S.N.); (C.S.)
| | - Nicole Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland;
| |
Collapse
|
14
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
15
|
Magliozzi R, Howell OW, Calabrese M, Reynolds R. Meningeal inflammation as a driver of cortical grey matter pathology and clinical progression in multiple sclerosis. Nat Rev Neurol 2023:10.1038/s41582-023-00838-7. [PMID: 37400550 DOI: 10.1038/s41582-023-00838-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/05/2023]
Abstract
Growing evidence from cerebrospinal fluid samples and post-mortem brain tissue from individuals with multiple sclerosis (MS) and rodent models indicates that the meninges have a key role in the inflammatory and neurodegenerative mechanisms underlying progressive MS pathology. The subarachnoid space and associated perivascular spaces between the membranes of the meninges are the access points for entry of lymphocytes, monocytes and macrophages into the brain parenchyma, and the main route for diffusion of inflammatory and cytotoxic molecules from the cerebrospinal fluid into the brain tissue. In addition, the meningeal spaces act as an exit route for CNS-derived antigens, immune cells and metabolites. A number of studies have demonstrated an association between chronic meningeal inflammation and a more severe clinical course of MS, suggesting that the build-up of immune cell aggregates in the meninges represents a rational target for therapeutic intervention. Therefore, understanding the precise cell and molecular mechanisms, timing and anatomical features involved in the compartmentalization of inflammation within the meningeal spaces in MS is vital. Here, we present a detailed review and discussion of the cellular, molecular and radiological evidence for a role of meningeal inflammation in MS, alongside the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy.
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Owain W Howell
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
- Institute of Life Sciences, Swansea University, Swansea, UK
| | - Massimiliano Calabrese
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
16
|
Yandamuri SS, Filipek B, Obaid AH, Lele N, Thurman JM, Makhani N, Nowak RJ, Guo Y, Lucchinetti CF, Flanagan EP, Longbrake EE, O'Connor KC. MOGAD patient autoantibodies induce complement, phagocytosis, and cellular cytotoxicity. JCI Insight 2023; 8:e165373. [PMID: 37097758 PMCID: PMC10393237 DOI: 10.1172/jci.insight.165373] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 04/20/2023] [Indexed: 04/26/2023] Open
Abstract
Myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD) is an inflammatory demyelinating CNS condition characterized by the presence of MOG autoantibodies. We sought to investigate whether human MOG autoantibodies are capable of mediating damage to MOG-expressing cells through multiple mechanisms. We developed high-throughput assays to measure complement activity (CA), complement-dependent cytotoxicity (CDC), antibody-dependent cellular phagocytosis (ADCP), and antibody-dependent cellular cytotoxicity (ADCC) of live MOG-expressing cells. MOGAD patient sera effectively mediate all of these effector functions. Our collective analyses reveal that (a) cytotoxicity is not incumbent on MOG autoantibody quantity alone; (b) engagement of effector functions by MOGAD patient serum is bimodal, with some sera exhibiting cytotoxic capacity while others did not; (c) the magnitude of CDC and ADCP is elevated closer to relapse, while MOG-IgG binding is not; and (d) all IgG subclasses can damage MOG-expressing cells. Histopathology from a representative MOGAD case revealed congruence between lesion histology and serum CDC and ADCP, and we identified NK cells, mediators of ADCC, in the cerebrospinal fluid of relapsing patients with MOGAD. Thus, MOGAD-derived autoantibodies are cytotoxic to MOG-expressing cells through multiple mechanisms, and assays quantifying CDC and ADCP may prove to be effective tools for predicting risk of future relapses.
Collapse
Affiliation(s)
- Soumya S Yandamuri
- Department of Neurology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Beata Filipek
- Department of Neurology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pharmaceutical Microbiology and Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Abeer H Obaid
- Department of Neurology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Institute of Biomedical Studies, Baylor University, Waco, Texas, USA
| | | | - Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Naila Makhani
- Department of Neurology and
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Yong Guo
- Department of Neurology and Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Claudia F Lucchinetti
- Department of Neurology and Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eoin P Flanagan
- Department of Neurology and Center for MS and Autoimmune Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kevin C O'Connor
- Department of Neurology and
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
17
|
Villar J, Cros A, De Juan A, Alaoui L, Bonte PE, Lau CM, Tiniakou I, Reizis B, Segura E. ETV3 and ETV6 enable monocyte differentiation into dendritic cells by repressing macrophage fate commitment. Nat Immunol 2023; 24:84-95. [PMID: 36543959 PMCID: PMC9810530 DOI: 10.1038/s41590-022-01374-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
In inflamed tissues, monocytes differentiate into macrophages (mo-Macs) or dendritic cells (mo-DCs). In chronic nonresolving inflammation, mo-DCs are major drivers of pathogenic events. Manipulating monocyte differentiation would therefore be an attractive therapeutic strategy. However, how the balance of mo-DC versus mo-Mac fate commitment is regulated is not clear. In the present study, we show that the transcriptional repressors ETV3 and ETV6 control human monocyte differentiation into mo-DCs. ETV3 and ETV6 inhibit interferon (IFN)-stimulated genes; however, their action on monocyte differentiation is independent of IFN signaling. Instead, we find that ETV3 and ETV6 directly repress mo-Mac development by controlling MAFB expression. Mice deficient for Etv6 in monocytes have spontaneous expression of IFN-stimulated genes, confirming that Etv6 regulates IFN responses in vivo. Furthermore, these mice have impaired mo-DC differentiation during inflammation and reduced pathology in an experimental autoimmune encephalomyelitis model. These findings provide information about the molecular control of monocyte fate decision and identify ETV6 as a therapeutic target to redirect monocyte differentiation in inflammatory disorders.
Collapse
Affiliation(s)
- Javiera Villar
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | - Adeline Cros
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | - Alba De Juan
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | - Lamine Alaoui
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | | | - Colleen M Lau
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ioanna Tiniakou
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Elodie Segura
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France.
| |
Collapse
|
18
|
Kolb H, Al-Louzi O, Beck ES, Sati P, Absinta M, Reich DS. From pathology to MRI and back: Clinically relevant biomarkers of multiple sclerosis lesions. Neuroimage Clin 2022; 36:103194. [PMID: 36170753 PMCID: PMC9668624 DOI: 10.1016/j.nicl.2022.103194] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Focal lesions in both white and gray matter are characteristic of multiple sclerosis (MS). Histopathological studies have helped define the main underlying pathological processes involved in lesion formation and evolution, serving as a gold standard for many years. However, histopathology suffers from an intrinsic bias resulting from over-reliance on tissue samples from late stages of the disease or atypical cases and is inadequate for routine patient assessment. Pathological-radiological correlative studies have established advanced MRI's sensitivity to several relevant MS-pathological substrates and its practicality for assessing dynamic changes and following lesions over time. This review focuses on novel imaging techniques that serve as biomarkers of critical pathological substrates of MS lesions: the central vein, chronic inflammation, remyelination and repair, and cortical lesions. For each pathological process, we address the correlative value of MRI to MS pathology, its contribution in elucidating MS pathology in vivo, and the clinical utility of the imaging biomarker.
Collapse
Affiliation(s)
- Hadar Kolb
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv-Yaffo, Israel,Corresponding author at: Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv-Yaffo, Israel.
| | - Omar Al-Louzi
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Erin S. Beck
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pascal Sati
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Martina Absinta
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA,Institute of Experimental Neurology (INSPE), IRCSS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy,Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Daniel S. Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
19
|
Sadeghi Hassanabadi N, Broux B, Marinović S, Gotthardt D. Innate Lymphoid Cells - Neglected Players in Multiple Sclerosis. Front Immunol 2022; 13:909275. [PMID: 35784374 PMCID: PMC9247827 DOI: 10.3389/fimmu.2022.909275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a highly debilitating autoimmune disease affecting millions of individuals worldwide. Although classically viewed as T-cell mediated disease, the role of innate lymphoid cells (ILC) such as natural killer (NK) cells and ILC 1-3s has become a focal point as several findings implicate them in the disease pathology. The role of ILCs in MS is still not completely understood as controversial findings have been reported assigning them either a protective or disease-accelerating role. Recent findings in experimental autoimmune encephalomyelitis (EAE) suggest that ILCs infiltrate the central nervous system (CNS), mediate inflammation, and have a disease exacerbating role by influencing the recruitment of autoreactive T-cells. Elucidating the detailed role of ILCs and altered signaling pathways in MS is essential for a more complete picture of the disease pathology and novel therapeutic targets. We here review the current knowledge about ILCs in the development and progression of MS and preclinical models of MS and discuss their potential for therapeutic applications.
Collapse
Affiliation(s)
| | - Bieke Broux
- University MSCenter; Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Sonja Marinović
- Division of Molecular Medicine, Laboratory of Personalized Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Dagmar Gotthardt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- *Correspondence: Dagmar Gotthardt,
| |
Collapse
|
20
|
Rodríguez-Lorenzo S, van Olst L, Rodriguez-Mogeda C, Kamermans A, van der Pol SMA, Rodríguez E, Kooij G, de Vries HE. Single-cell profiling reveals periventricular CD56 bright NK cell accumulation in multiple sclerosis. eLife 2022; 11:e73849. [PMID: 35536009 PMCID: PMC9135404 DOI: 10.7554/elife.73849] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 04/29/2022] [Indexed: 11/21/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease characterised by immune cell infiltration resulting in lesions that preferentially affect periventricular areas of the brain. Despite research efforts to define the role of various immune cells in MS pathogenesis, the focus has been on a few immune cell populations while full-spectrum analysis, encompassing others such as natural killer (NK) cells, has not been performed. Here, we used single-cell mass cytometry (CyTOF) to profile the immune landscape of brain periventricular areas - septum and choroid plexus - and of the circulation from donors with MS, dementia and controls without neurological disease. Using a 37-marker panel, we revealed the infiltration of T cells and antibody-secreting cells in periventricular brain regions and identified a novel NK cell signature specific to MS. CD56bright NK cells were accumulated in the septum of MS donors and displayed an activated and migratory phenotype, similar to that of CD56bright NK cells in the circulation. We validated this signature by multiplex immunohistochemistry and found that the number of NK cells with high expression of granzyme K, typical of the CD56bright subset, was increased in both periventricular lesions and the choroid plexus of donors with MS. Together, our multi-tissue single-cell data shows that CD56bright NK cells accumulate in the periventricular brain regions of MS patients, bringing NK cells back to the spotlight of MS pathology.
Collapse
Affiliation(s)
- Sabela Rodríguez-Lorenzo
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Lynn van Olst
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Carla Rodriguez-Mogeda
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Alwin Kamermans
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Susanne MA van der Pol
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Ernesto Rodríguez
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity InstituteAmsterdamNetherlands
| | - Gijs Kooij
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Helga E de Vries
- MS Center Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| |
Collapse
|
21
|
Beliën J, Goris A, Matthys P. Natural Killer Cells in Multiple Sclerosis: Entering the Stage. Front Immunol 2022; 13:869447. [PMID: 35464427 PMCID: PMC9019710 DOI: 10.3389/fimmu.2022.869447] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/14/2022] [Indexed: 11/14/2022] Open
Abstract
Studies investigating the immunopathology of multiple sclerosis (MS) have largely focused on adaptive T and B lymphocytes. However, in recent years there has been an increased interest in the contribution of innate immune cells, amongst which the natural killer (NK) cells. Apart from their canonical role of controlling viral infections, cell stress and malignancies, NK cells are increasingly being recognized for their modulating effect on the adaptive immune system, both in health and autoimmune disease. From different lines of research there is now evidence that NK cells contribute to MS immunopathology. In this review, we provide an overview of studies that have investigated the role of NK cells in the pathogenesis of MS by use of the experimental autoimmune encephalomyelitis (EAE) animal model, MS genetics or through ex vivo and in vitro work into the immunology of MS patients. With the advent of modern hypothesis-free technologies such as single-cell transcriptomics, we are exposing an unexpected NK cell heterogeneity, increasingly blurring the boundaries between adaptive and innate immunity. We conclude that unravelling this heterogeneity, as well as the mechanistic link between innate and adaptive immune cell functions will lay the foundation for the use of NK cells as prognostic tools and therapeutic targets in MS and a myriad of other currently uncurable autoimmune disorders.
Collapse
Affiliation(s)
- Jarne Beliën
- Department of Neurosciences, Laboratory for Neuroimmunology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - An Goris
- Department of Neurosciences, Laboratory for Neuroimmunology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Ruder J, Rex J, Obahor S, Docampo MJ, Müller AMS, Schanz U, Jelcic I, Martin R. NK Cells and Innate-Like T Cells After Autologous Hematopoietic Stem Cell Transplantation in Multiple Sclerosis. Front Immunol 2022; 12:794077. [PMID: 34975899 PMCID: PMC8716406 DOI: 10.3389/fimmu.2021.794077] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/29/2021] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system, in which autoreactive T and B cells play important roles. Other lymphocytes such as NK cells and innate-like T cells appear to be involved as well. To name a few examples, CD56bright NK cells were described as an immunoregulatory NK cell subset in MS while innate-like T cells in MS were described in brain lesions and with proinflammatory signatures. Autologous hematopoietic stem cell transplantation (aHSCT) is a procedure used to treat MS. This procedure includes hematopoietic stem/progenitor cell (HSPC) mobilization, then high-dose chemotherapy combined with anti-thymocyte globulin (ATG) and subsequent infusion of the patients own HSPCs to reconstitute a functional immune system. aHSCT inhibits MS disease activity very effectively and for long time, presumably due to elimination of autoreactive T cells. Here, we performed multidimensional flow cytometry experiments in peripheral blood lymphocytes of 27 MS patients before and after aHSCT to address its potential influence on NK and innate-like T cells. After aHSCT, the relative frequency and absolute numbers of CD56bright NK cells rise above pre-aHSCT levels while all studied innate-like T cell populations decrease. Hence, our data support an enhanced immune regulation by CD56bright NK cells and the efficient reduction of proinflammatory innate-like T cells by aHSCT in MS. These observations contribute to our current understanding of the immunological effects of aHSCT in MS.
Collapse
Affiliation(s)
- Josefine Ruder
- Neuroimmunology and Multiple Sclerosis (MS) Research Section (NIMS), Department of Neurology, University and University Hospital Zurich, Zurich, Switzerland
| | - Jordan Rex
- Neuroimmunology and Multiple Sclerosis (MS) Research Section (NIMS), Department of Neurology, University and University Hospital Zurich, Zurich, Switzerland
| | - Simon Obahor
- Neuroimmunology and Multiple Sclerosis (MS) Research Section (NIMS), Department of Neurology, University and University Hospital Zurich, Zurich, Switzerland
| | - María José Docampo
- Neuroimmunology and Multiple Sclerosis (MS) Research Section (NIMS), Department of Neurology, University and University Hospital Zurich, Zurich, Switzerland
| | - Antonia M S Müller
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - Urs Schanz
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis (MS) Research Section (NIMS), Department of Neurology, University and University Hospital Zurich, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis (MS) Research Section (NIMS), Department of Neurology, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Age-related changes in multiple sclerosis and experimental autoimmune encephalomyelitis. Semin Immunol 2022; 59:101631. [PMID: 35752572 DOI: 10.1016/j.smim.2022.101631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
A better understanding of the pathological mechanisms that drive neurodegeneration in people living with multiple sclerosis (MS) is needed to design effective therapies to treat and/or prevent disease progression. We propose that CNS-intrinsic inflammation and re-modelling of the sub-arachnoid space of the leptomeninges sets the stage for neurodegeneration from the earliest stages of MS. While neurodegenerative processes are clinically silent early in disease, ageing results in neurodegenerative changes that become clinically manifest as progressive disability. Here we review pathological correlates of MS disease progression, highlight emerging mouse models that mimic key progressive changes in MS, and provide new perspectives on therapeutic approaches to protect against MS-associated neurodegeneration.
Collapse
|
24
|
Mi Y, Han J, Zhu J, Jin T. Role of the PD-1/PD-L1 Signaling in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis: Recent Insights and Future Directions. Mol Neurobiol 2021; 58:6249-6271. [PMID: 34480337 PMCID: PMC8639577 DOI: 10.1007/s12035-021-02495-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmunity-related chronic demyelination disease of the central nervous system (CNS), causing young disability. Currently, highly specific immunotherapies for MS are still lacking. Programmed cell death 1 (PD-1) is an immunosuppressive co-stimulatory molecule, which is expressed on activated T lymphocytes, B lymphocytes, natural killer cells, and other immune cells. PD-L1, the ligand of PD-1, is expressed on T lymphocytes, B lymphocytes, dendritic cells, and macrophages. PD-1/PD-L1 delivers negative regulatory signals to immune cells, maintaining immune tolerance and inhibiting autoimmunity. This review comprehensively summarizes current insights into the role of PD-1/PD-L1 signaling in MS and its animal model experimental autoimmune encephalomyelitis (EAE). The potentiality of PD-1/PD-L1 as biomarkers or therapeutic targets for MS will also be discussed.
Collapse
Affiliation(s)
- Yan Mi
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Present Address: Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| |
Collapse
|
25
|
Schwichtenberg SC, Wisgalla A, Schroeder-Castagno M, Alvarez-González C, Schlickeiser S, Siebert N, Bellmann-Strobl J, Wernecke KD, Paul F, Dörr J, Infante-Duarte C. Fingolimod Therapy in Multiple Sclerosis Leads to the Enrichment of a Subpopulation of Aged NK Cells. Neurotherapeutics 2021; 18:1783-1797. [PMID: 34244929 PMCID: PMC8608997 DOI: 10.1007/s13311-021-01078-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/04/2023] Open
Abstract
Fingolimod is an approved oral treatment for relapsing-remitting multiple sclerosis (RRMS) that modulates agonistically the sphingosin-1-phosphate receptor (S1PR), inhibiting thereby the egress of lymphocytes from the lymph nodes. In this interventional prospective clinical phase IV trial, we longitudinally investigated the impact of fingolimod on frequencies of NK cell subpopulations by flow cytometry in 17 RRMS patients at baseline and 1, 3, 6, and 12 months after treatment initiation. Clinical outcome was assessed by the Expanded Disability Status Scale (EDSS) and annualized relapse rates (ARR). Over the study period, median EDSS remained stable from month 3 to month 12, and ARR decreased compared to ARR in the 24 months prior treatment. Treatment was paralleled by an increased frequency of circulating NK cells, due primarily to an increase in CD56dimCD94low mature NK cells, while the CD56bright fraction and CD127+ innate lymphoid cells (ILCs) decreased over time. An unsupervised clustering algorithm further revealed that a particular fraction of NK cells defined by the expression of CD56dimCD16++KIR+/-NKG2A-CD94-CCR7+/-CX3CR1+/-NKG2C-NKG2D+NKp46-DNAM1++CD127+ increased during treatment. This specific phenotype might reflect a status of aged, fully differentiated, and less functional NK cells. Our study confirms that fingolimod treatment affects both NK cells and ILC. In addition, our study suggests that treatment leads to the enrichment of a specific NK cell subset characterized by an aged phenotype. This might limit the anti-microbial and anti-tumour NK cell activity in fingolimod-treated patients.
Collapse
Affiliation(s)
- Svenja C Schwichtenberg
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
| | - Anne Wisgalla
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for "Psychiatrie Und Medizinische Klinik M.S. Psychosomatik,", Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Maria Schroeder-Castagno
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
| | - Cesar Alvarez-González
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Föhrer Str. 15, 13353, Berlin, Germany
| | - Nadja Siebert
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Judith Bellmann-Strobl
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Klaus-Dieter Wernecke
- Charité - Universitätsmedizin Berlin and CRO SOSTANA GmbH, Wildensteiner Straße 27, 10318, Berlin, Germany
| | - Friedemann Paul
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Jan Dörr
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Campus Mitte, Sauerbruchweg 5, 10117, Berlin, Germany
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany
- Current Affiliation: Multiple Sclerosis Center, Oberhavel Kliniken, Marwitzer Straße 91, 16761, Hennigsdorf, Germany
| | - Carmen Infante-Duarte
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Campus Virchow Klinikum, Augustenburger Platz 1 (Südstr. 2/Föhrer Str. 15), 13353, Berlin, Germany.
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine & Charité - Universitätsmedizin Berlin, Robert-Rössle-Straße 10, 13125, Berlin, Germany.
| |
Collapse
|
26
|
Elser H, Parks RM, Moghavem N, Kiang MV, Bozinov N, Henderson VW, Rehkopf DH, Casey JA. Anomalously warm weather and acute care visits in patients with multiple sclerosis: A retrospective study of privately insured individuals in the US. PLoS Med 2021; 18:e1003580. [PMID: 33901187 PMCID: PMC8109782 DOI: 10.1371/journal.pmed.1003580] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 05/10/2021] [Accepted: 03/08/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND As the global climate changes in response to anthropogenic greenhouse gas emissions, weather and temperature are expected to become increasingly variable. Although heat sensitivity is a recognized clinical feature of multiple sclerosis (MS), a chronic demyelinating disorder of the central nervous system, few studies have examined the implications of climate change for patients with this disease. METHODS AND FINDINGS We conducted a retrospective cohort study of individuals with MS ages 18-64 years in a nationwide United States patient-level commercial and Medicare Advantage claims database from 2003 to 2017. We defined anomalously warm weather as any month in which local average temperatures exceeded the long-term average by ≥1.5°C. We estimated the association between anomalously warm weather and MS-related inpatient, outpatient, and emergency department visits using generalized log-linear models. From 75,395,334 individuals, we identified 106,225 with MS. The majority were women (76.6%) aged 36-55 years (59.0%). Anomalously warm weather was associated with increased risk for emergency department visits (risk ratio [RR] = 1.043, 95% CI: 1.025-1.063) and inpatient visits (RR = 1.032, 95% CI: 1.010-1.054). There was limited evidence of an association between anomalously warm weather and MS-related outpatient visits (RR = 1.010, 95% CI: 1.005-1.015). Estimates were similar for men and women, strongest among older individuals, and exhibited substantial variation by season, region, and climate zone. Limitations of the present study include the absence of key individual-level measures of socioeconomic position (i.e., race/ethnicity, occupational status, and housing quality) that may determine where individuals live-and therefore the extent of their exposure to anomalously warm weather-as well as their propensity to seek treatment for neurologic symptoms. CONCLUSIONS Our findings suggest that as global temperatures rise, individuals with MS may represent a particularly susceptible subpopulation, a finding with implications for both healthcare providers and systems.
Collapse
Affiliation(s)
- Holly Elser
- Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| | - Robbie M. Parks
- Earth Institute, Columbia University, New York, New York, United States of America
- Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - Nuriel Moghavem
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Mathew V. Kiang
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, United States of America
| | - Nina Bozinov
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Victor W. Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - David H. Rehkopf
- Center for Population Health Sciences, Stanford, California, United States of America
| | - Joan A. Casey
- Mailman School of Public Health, Columbia University, New York, New York, United States of America
| |
Collapse
|
27
|
Jafari M, Schumacher AM, Snaidero N, Ullrich Gavilanes EM, Neziraj T, Kocsis-Jutka V, Engels D, Jürgens T, Wagner I, Weidinger JDF, Schmidt SS, Beltrán E, Hagan N, Woodworth L, Ofengeim D, Gans J, Wolf F, Kreutzfeldt M, Portugues R, Merkler D, Misgeld T, Kerschensteiner M. Phagocyte-mediated synapse removal in cortical neuroinflammation is promoted by local calcium accumulation. Nat Neurosci 2021; 24:355-367. [PMID: 33495636 DOI: 10.1038/s41593-020-00780-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/16/2020] [Indexed: 01/30/2023]
Abstract
Cortical pathology contributes to chronic cognitive impairment of patients suffering from the neuroinflammatory disease multiple sclerosis (MS). How such gray matter inflammation affects neuronal structure and function is not well understood. In the present study, we use functional and structural in vivo imaging in a mouse model of cortical MS to demonstrate that bouts of cortical inflammation disrupt cortical circuit activity coincident with a widespread, but transient, loss of dendritic spines. Spines destined for removal show local calcium accumulations and are subsequently removed by invading macrophages or activated microglia. Targeting phagocyte activation with a new antagonist of the colony-stimulating factor 1 receptor prevents cortical synapse loss. Overall, our study identifies synapse loss as a key pathological feature of inflammatory gray matter lesions that is amenable to immunomodulatory therapy.
Collapse
Affiliation(s)
- Mehrnoosh Jafari
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Adrian-Minh Schumacher
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nicolas Snaidero
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | - Emily M Ullrich Gavilanes
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Tradite Neziraj
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Virág Kocsis-Jutka
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Daniel Engels
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Tanja Jürgens
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Juan Daniel Flórez Weidinger
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Stephanie S Schmidt
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nellwyn Hagan
- Rare and Neurological Disease Research, Sanofi, Framingham, MA, USA
| | - Lisa Woodworth
- Rare and Neurological Disease Research, Sanofi, Framingham, MA, USA
| | - Dimitry Ofengeim
- Rare and Neurological Disease Research, Sanofi, Framingham, MA, USA
| | - Joseph Gans
- Translational Sciences Genomics, Sanofi, Framingham, MA, USA
| | - Fred Wolf
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany.,Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Ruben Portugues
- Sensorimotor Control, Max Planck Institute of Neurobiology, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. .,Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland.
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany. .,German Center for Neurodegenerative Diseases, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany. .,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
28
|
Ramaglia V, Rojas O, Naouar I, Gommerman JL. The Ins and Outs of Central Nervous System Inflammation-Lessons Learned from Multiple Sclerosis. Annu Rev Immunol 2021; 39:199-226. [PMID: 33524273 DOI: 10.1146/annurev-immunol-093019-124155] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by the inappropriate invasion of lymphocytes and monocytes into the central nervous system (CNS), where they orchestrate the demyelination of axons, leading to physical and cognitive disability. There are many reasons immunologists should be interested in MS. Aside from the fact that there is still significant unmet need for patients living with the progressive form of the disease, MS is a case study for how immune cells cross CNS barriers and subsequently interact with specialized tissue parenchymal cells. In this review, we describe the types of immune cells that infiltrate the CNS and then describe interactions between immune cells and glial cells in different types of lesions. Lastly, we provide evidence for CNS-compartmentalized immune cells and speculate on how this impacts disease progression for MS patients.
Collapse
Affiliation(s)
- Valeria Ramaglia
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Olga Rojas
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | | |
Collapse
|
29
|
Trend S, Leffler J, Teige I, Frendéus B, Kermode AG, French MA, Hart PH. FcγRIIb Expression Is Decreased on Naive and Marginal Zone-Like B Cells From Females With Multiple Sclerosis. Front Immunol 2021; 11:614492. [PMID: 33505402 PMCID: PMC7832177 DOI: 10.3389/fimmu.2020.614492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
B cells are critical to the development of multiple sclerosis (MS), but the mechanisms by which they contribute to the disease are poorly defined. We hypothesised that the expression of CD32b (FcγRIIb), a receptor for the Fc region of IgG with inhibitory activities in B cells, is lower on B cell subsets from people with clinically isolated syndrome (CIS) or MS. CD32b expression was highest on post-naive IgM+ B cell subsets in healthy controls. For females with MS or CIS, significantly lower CD32b expression was identified on IgM+ B cell subsets, including naive and IgMhi MZ-like B cells, when compared with control females. Lower CD32b expression on these B cell subsets was associated with detectable anti-Epstein Barr Virus viral capsid antigen IgM antibodies, and higher serum levels of B cell activating factor. To investigate the effects of lower CD32b expression, B cells were polyclonally activated in the presence of IgG immune complexes, with or without a CD32b blocking antibody, and the expression of TNF and IL-10 in B cell subsets was assessed. The reduction of TNF but not IL-10 expression in controls mediated by IgG immune complexes was reversed by CD32b blockade in naive and IgMhi MZ-like B cells only. However, no consequence of lower CD32b expression on these cells from females with CIS or MS was detected. Our findings highlight a potential role for naive and marginal zone-like B cells in the immunopathogenesis of MS in females, which requires further investigation.
Collapse
Affiliation(s)
- Stephanie Trend
- Inflammation Laboratory, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,Perron Institute for Neurological and Translational Science, University of Western Australia, Perth, WA, Australia
| | - Jonatan Leffler
- Inflammation Laboratory, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Ingrid Teige
- Demyelinating Diseases Research Group, BioInvent International AB, Lund, Sweden
| | - Björn Frendéus
- Demyelinating Diseases Research Group, BioInvent International AB, Lund, Sweden
| | - Allan G Kermode
- Perron Institute for Neurological and Translational Science, University of Western Australia, Perth, WA, Australia.,Institute for Immunology and Infectious Disease, Murdoch University, Perth, WA, Australia
| | - Martyn A French
- Medical School and School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Prue H Hart
- Inflammation Laboratory, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
30
|
Morgan BP, Gommerman JL, Ramaglia V. An "Outside-In" and "Inside-Out" Consideration of Complement in the Multiple Sclerosis Brain: Lessons From Development and Neurodegenerative Diseases. Front Cell Neurosci 2021; 14:600656. [PMID: 33488361 PMCID: PMC7817777 DOI: 10.3389/fncel.2020.600656] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The last 15 years have seen an explosion of new findings on the role of complement, a major arm of the immune system, in the central nervous system (CNS) compartment including contributions to cell migration, elimination of synapse during development, aberrant synapse pruning in neurologic disorders, damage to nerve cells in autoimmune diseases, and traumatic injury. Activation of the complement system in multiple sclerosis (MS) is typically thought to occur as part of a primary (auto)immune response from the periphery (the outside) against CNS antigens (the inside). However, evidence of local complement production from CNS-resident cells, intracellular complement functions, and the more recently discovered role of early complement components in shaping synaptic circuits in the absence of inflammation opens up the possibility that complement-related sequelae may start and finish within the brain itself. In this review, the complement system will be introduced, followed by evidence that implicates complement in shaping the developing, adult, and normal aging CNS as well as its contribution to pathology in neurodegenerative conditions. Discussion of data supporting "outside-in" vs. "inside-out" roles of complement in MS will be presented, concluded by thoughts on potential approaches to therapies targeting specific elements of the complement system.
Collapse
Affiliation(s)
- B. Paul Morgan
- UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| | | | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Ramaglia V, Florescu A, Zuo M, Sheikh-Mohamed S, Gommerman JL. Stromal Cell–Mediated Coordination of Immune Cell Recruitment, Retention, and Function in Brain-Adjacent Regions. THE JOURNAL OF IMMUNOLOGY 2021; 206:282-291. [DOI: 10.4049/jimmunol.2000833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022]
|
32
|
Delbridge ARD, Huh D, Brickelmaier M, Burns JC, Roberts C, Challa R, Raymond N, Cullen P, Carlile TM, Ennis KA, Liu M, Sun C, Allaire NE, Foos M, Tsai HH, Franchimont N, Ransohoff RM, Butts C, Mingueneau M. Organotypic Brain Slice Culture Microglia Exhibit Molecular Similarity to Acutely-Isolated Adult Microglia and Provide a Platform to Study Neuroinflammation. Front Cell Neurosci 2020; 14:592005. [PMID: 33473245 PMCID: PMC7812919 DOI: 10.3389/fncel.2020.592005] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Microglia are central nervous system (CNS) resident immune cells that have been implicated in neuroinflammatory pathogenesis of a variety of neurological conditions. Their manifold context-dependent contributions to neuroinflammation are only beginning to be elucidated, which can be attributed in part to the challenges of studying microglia in vivo and the lack of tractable in vitro systems to study microglia function. Organotypic brain slice cultures offer a tissue-relevant context that enables the study of CNS resident cells and the analysis of brain slice microglial phenotypes has provided important insights, in particular into neuroprotective functions. Here we use RNA sequencing, direct digital quantification of gene expression with nCounter® technology and targeted analysis of individual microglial signature genes, to characterize brain slice microglia relative to acutely-isolated counterparts and 2-dimensional (2D) primary microglia cultures, a widely used in vitro surrogate. Analysis using single cell and population-based methods found brain slice microglia exhibited better preservation of canonical microglia markers and overall gene expression with stronger fidelity to acutely-isolated adult microglia, relative to in vitro cells. We characterized the dynamic phenotypic changes of brain slice microglia over time, after plating in culture. Mechanical damage associated with slice preparation prompted an initial period of inflammation, which resolved over time. Based on flow cytometry and gene expression profiling we identified the 2-week timepoint as optimal for investigation of microglia responses to exogenously-applied stimuli as exemplified by treatment-induced neuroinflammatory changes observed in microglia following LPS, TNF and GM-CSF addition to the culture medium. Altogether these findings indicate that brain slice cultures provide an experimental system superior to in vitro culture of microglia as a surrogate to investigate microglia functions, and the impact of soluble factors and cellular context on their physiology.
Collapse
Affiliation(s)
- Alex R D Delbridge
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States.,Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Dann Huh
- Translational Biology, Biogen, Cambridge, MA, United States
| | - Margot Brickelmaier
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Jeremy C Burns
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Chris Roberts
- Translational Biology, Biogen, Cambridge, MA, United States
| | - Ravi Challa
- Translational Biology, Biogen, Cambridge, MA, United States
| | - Naideline Raymond
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Patrick Cullen
- Translational Biology, Biogen, Cambridge, MA, United States
| | | | - Katelin A Ennis
- Genetic and Neurodevelopmental Disorders, Biogen, Cambridge, MA, United States
| | - Mei Liu
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Chao Sun
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Normand E Allaire
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Marianna Foos
- Biogen Postdoctoral Scientist Program, Biogen, Cambridge, MA, United States
| | - Hui-Hsin Tsai
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | | | - Richard M Ransohoff
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| | - Cherie Butts
- Digital & Quantitative Medicine, Biogen, Cambridge, MA, United States
| | - Michael Mingueneau
- Multiple Sclerosis and Neuroimmunology Research Unit, Biogen, Cambridge, MA, United States
| |
Collapse
|
33
|
Silva BA, Miglietta EA, Ferrari CC. Training the brain: could it improve multiple sclerosis treatment? Rev Neurosci 2020; 31:779-792. [PMID: 32712593 DOI: 10.1515/revneuro-2020-0014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS) is a neurological disease characterized by neuroinflammation, demyelination and axonal degeneration along with loss of function in the central nervous system. For many years, research in MS has focused on the efficacy of pharmacological treatments. However, during the last years, many publications have been dedicated to the study of the efficacy of non-pharmacological strategies, such as physical exercise and cognitive training. Beneficial effects of the combination of both strategies on cognitive function have been described in both ageing adults and patients with neurodegenerative diseases, such as MS. The analysis of combining both physical and cognitive stimulation can be summarized by the environmental enrichment (EE) experiments, which are more suitable for animal models. EE refers to housing conditions consisting of exercise and cognitive and social stimulation. In this review, we will summarize the available studies that describe the influence of EE in both MS patients and MS animal models.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET, Potosí 4240, Buenos Aires, C1181ACH, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA-CONICET, Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina
| | - Esteban Alberto Miglietta
- Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA-CONICET, Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET, Potosí 4240, Buenos Aires, C1181ACH, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA-CONICET, Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina
| |
Collapse
|
34
|
Zelek WM, Menzies GE, Brancale A, Stockinger B, Morgan BP. Characterizing the original anti-C5 function-blocking antibody, BB5.1, for species specificity, mode of action and interactions with C5. Immunology 2020; 161:103-113. [PMID: 32557571 PMCID: PMC7496778 DOI: 10.1111/imm.13228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/05/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022] Open
Abstract
The implication of complement in multiple diseases over the last 20 years has fuelled interest in developing anti-complement drugs. To date, the focus has been on C5; blocking cleavage of C5 prevents formation of two pro-inflammatory activities, C5a anaphylatoxin and membrane attack complex. The concept of C5 blockade to inhibit inflammation dates back 30 years to the description of BB5.1, an anti-C5 blocking monoclonal antibody raised in C5-deficient mice. This antibody proved an invaluable tool to demonstrate complement involvement in mouse disease models and catalysed enthusiasm for anti-complement drug development, culminating in the anti-human C5 monoclonal antibody eculizumab, the most successful anti-complement drug to date, already in clinical use for several rare diseases. Despite its key role in providing proof-of-concept for C5 blockade, the mechanism of BB5.1 inhibition remains poorly understood. Here, we characterized BB5.1 cross-species inhibition, C5 binding affinity and chain specificity. BB5.1 efficiently inhibited C5 in mouse serum but not in human or other rodent sera; it prevented C5 cleavage and C5a generation. BB5.1 bound the C5 α-chain with high affinity and slow off-rate. BB5.1 complementarity-determining regions were obtained and docking algorithms were used to predict the likely binding interface on mouse C5.
Collapse
Affiliation(s)
- Wioleta M. Zelek
- Systems Immunity University Research InstituteSchool of MedicineCardiff UniversityCardiffUK
| | | | - Andrea Brancale
- School of Pharmacy and Pharmaceutical SciencesCardiff UniversityCardiffUK
| | | | - Bryan Paul Morgan
- Systems Immunity University Research InstituteSchool of MedicineCardiff UniversityCardiffUK
- Dementia Research InstituteCardiff UniversityCardiffUK
| |
Collapse
|
35
|
Cui LY, Chu SF, Chen NH. The role of chemokines and chemokine receptors in multiple sclerosis. Int Immunopharmacol 2020; 83:106314. [PMID: 32197226 PMCID: PMC7156228 DOI: 10.1016/j.intimp.2020.106314] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/31/2020] [Accepted: 02/11/2020] [Indexed: 01/13/2023]
Abstract
Summarize the study of the role of chemokines and their receptors in multiple sclerosis (MS) patients and MS animal models. Discuss their potential significance in inflammatory injury and repair of MS. Summarize the progress in the research of MS antagonists in recent years with chemokine receptors as targets.
Multiple sclerosis (MS) is a chronic inflammatory disease that is characterized by leukocyte infiltration and subsequent axonal damage, demyelinating inflammation, and formation of sclerosing plaques in brain tissue. The results of various studies in patients indicate that autoimmunity and inflammation make an important impact on the pathogenesis of MS. Chemokines are key mediators of inflammation development and cell migration, mediating various immune cell responses, including chemotaxis and immune activation, and are important in immunity and inflammation, therefore we focus on chemokines and their receptors in multiple sclerosis. In this article, we summarize the study of the role of prominent chemokines and their receptors in MS patients and MS animal modelsand discuss their potential significance in inflammatory injury and repair of MS. We have also summarized the progress in the treatment of multiple sclerosis antagonists in recent years with chemokine receptors as targets.
Collapse
Affiliation(s)
- Li-Yuan Cui
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
36
|
Mimpen M, Smolders J, Hupperts R, Damoiseaux J. Natural killer cells in multiple sclerosis: A review. Immunol Lett 2020; 222:1-11. [PMID: 32113900 DOI: 10.1016/j.imlet.2020.02.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
As the most common non-traumatic disabling disease among adolescents, multiple sclerosis (MS) is a devastating neurological inflammatory disease of the central nervous system. Research has not yet fully elucidated its pathogenesis, but it has shown MS to be a complex, multifactorial disease with many interplaying factors. One of these factors, natural killer (NK) cells, lymphocytes of the innate immune system, have recently gained attention due to the effects of daclizumab therapy, causing an expansion of the immunoregulatory subset of NK cells. Since then, NK cells and their relation to MS have been the focus of research, with many new findings being published in the last decade. In this review, NK cells are pictured as potent cytotoxic killers, as well as unique immune-regulators. Additionally, an overview of our current knowledge regarding NK cells in MS is given. The role of NK cells in MS is reviewed in the context of well-established environmental factors and current disease modifying therapies to gain further understanding of the pathogenesis and treatment options in MS.
Collapse
Affiliation(s)
- Max Mimpen
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands
| | - Joost Smolders
- Department of Neurology, Erasmus University Medical Center, Rotterdam The Netherlands; Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam The Netherlands
| | - Raymond Hupperts
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands; Department of Neurology, Zuyderland Medical Center, Sittard The Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht The Netherlands.
| |
Collapse
|
37
|
Junker A, Wozniak J, Voigt D, Scheidt U, Antel J, Wegner C, Brück W, Stadelmann C. Extensive subpial cortical demyelination is specific to multiple sclerosis. Brain Pathol 2020; 30:641-652. [PMID: 31916298 PMCID: PMC8018087 DOI: 10.1111/bpa.12813] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
Cortical demyelinated lesions are frequent and widespread in chronic multiple sclerosis (MS) patients, and may contribute to disease progression. Inflammation and related oxidative stress have been proposed as central mediators of cortical damage, yet meningeal and cortical inflammation is not specific to MS, but also occurs in other diseases. The first aim of this study was to test whether cortical demyelination was specific for demyelinating CNS diseases compared to other CNS disorders with prominent meningeal and cortical inflammation. The second aim was to assess whether oxidative tissue damage was associated with the extent of neuroaxonal damage. We studied a large cohort of patients diagnosed with demyelinating CNS diseases and non‐demyelinating diseases of autoimmune, infectious, neoplastic or metabolic origin affecting the meninges and the cortex. Included were patients with MS, acute disseminated encephalomyelitis (ADEM), neuromyelitis optica (NMO), viral and bacterial meningoencephalitis, progressive multifocal leukoencephalopathy (PML), subacute sclerosing panencephalitis (SSPE), carcinomatous and lymphomatous meningitis and metabolic disorders such as extrapontine myelinolysis, thus encompassing a wide range of adaptive and innate cytokine signatures. Using myelin protein immunohistochemistry, we found cortical demyelination in MS, ADEM, PML and extrapontine myelinolysis, whereby each condition showed a disease‐specific histopathological pattern. Remarkably, extensive ribbon‐like subpial demyelination was only observed in MS, thus providing an important pathogenetic and diagnostic cue. Cortical oxidative injury was detected in both demyelinating and non‐demyelinating CNS disorders. Our data demonstrate that meningeal and cortical inflammation alone accompanied by oxidative stress are not sufficient to generate the extensive subpial cortical demyelination found in MS, but require other MS‐specific factors.
Collapse
Affiliation(s)
- Andreas Junker
- Institute of Neuropathology, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Department of Neuropathology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Jadwiga Wozniak
- Institute of Neuropathology, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - David Voigt
- Institute of Neuropathology, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Uta Scheidt
- Institute of Neuropathology, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Jack Antel
- Montreal Neurological Institute, McGill University Health Centre, 2155 Guy Street, Montreal, Canada
| | - Christiane Wegner
- Institute of Neuropathology, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.,Department of Child and Adolescent Psychiatry/Psychotherapy, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Georg August University Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| |
Collapse
|
38
|
Ribes S, Arcilla C, Ott M, Schütze S, Hanisch UK, Nessler S, Nau R. Pre-treatment with the viral Toll-like receptor 3 agonist poly(I:C) modulates innate immunity and protects neutropenic mice infected intracerebrally with Escherichia coli. J Neuroinflammation 2020; 17:24. [PMID: 31952519 PMCID: PMC6969464 DOI: 10.1186/s12974-020-1700-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/03/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Individuals with impaired immunity are more susceptible to infections than immunocompetent subjects. No vaccines are currently available to induce protection against E. coli meningoencephalitis. This study evaluated the potential of poly(I:C) pre-treatment to induce trained immunity. Poly(I:C) was administered as a non-specific stimulus of innate immune responses to protect immunocompetent and neutropenic wild-type mice from a subsequent challenge by the intracranial injection of E. coli K1. METHODS Three days prior to infection, mice received an intraperitoneal injection of poly(I:C) or vehicle. Kaplan-Meier survival curves were analyzed. In short-term experiments, bacterial titers and the inflammatory response were characterized in the blood, cerebellum, and spleen homogenates. NK cell subpopulations in the brain and spleen were analyzed by flow cytometry. Numbers of microglia and activation scores were evaluated by histopathology. RESULTS Pre-treatment with 200 μg poly(I:C) increased survival time, reduced mortality, and enhanced bacterial clearance in the blood, cerebellum, and spleen at early infection in neutropenic mice. Poly(I:C)-mediated protection correlated with an augmented number of NK cells (CD45+NK1.1+CD3-) and Iba-1+ microglial cells and a higher production of IFN-γ in the brain. In the spleen, levels of CCL5/RANTES and IFN-γ were increased and sustained in surviving poly(I:C)-treated animals for 14 days after infection. In immunocompetent animals, survival time was not significantly prolonged in poly(I:C)-treated animals although poly(I:C) priming reduced brain bacterial concentrations compared with vehicle-injected animals at early infection. CONCLUSIONS Pre-treatment with the viral TLR3 agonist poly(I:C) modulated innate immune responses and strengthened the resistance of neutropenic mice against E. coli K1 meningoencephalitis.
Collapse
Affiliation(s)
- Sandra Ribes
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
| | - Christa Arcilla
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Martina Ott
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Sandra Schütze
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Uwe-Karsten Hanisch
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Stefan Nessler
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Roland Nau
- Institute of Neuropathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.,Department of Geriatrics, Evangelisches Krankenhaus Göttingen-Weende, 37075, Göttingen, Germany
| |
Collapse
|
39
|
Ward LA, Lee DS, Sharma A, Wang A, Naouar I, Ma XI, Pikor N, Nuesslein-Hildesheim B, Ramaglia V, Gommerman JL. Siponimod therapy implicates Th17 cells in a preclinical model of subpial cortical injury. JCI Insight 2020; 5:132522. [PMID: 31821174 DOI: 10.1172/jci.insight.132522] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/25/2019] [Indexed: 12/27/2022] Open
Abstract
Subpial demyelination is a specific hallmark of multiple sclerosis and a correlate of disease progression. Although the mechanism(s) that mediate pathogenesis in the subpial compartment remain unclear, it has been speculated that inflammation in the overlying meninges may be associated with subpial injury. Here we show that adoptive transfer of proteolipid protein-primed Th17 cells into SJL/J recipient mice induces subpial demyelination associated with microglial/macrophage activation, disruption of the glial limitans, and evidence of an oxidative stress response. This pathology was topologically associated with foci of immune cells in the meninges and occurred in the absence of measurable anti-myelin oligodendrocyte glycoprotein IgM or IgG antibodies. To test the role of brain-infiltrating leukocytes on subpial injury, we modulated sphingosine 1-phosphate (S1P) receptor1,5 activity with BAF312 (siponimod) treatment. Administration of BAF312, even after adoptively transferred T cells had entered the brain, significantly ameliorated clinical experimental autoimmune encephalomyelitis and diminished subpial pathology, concomitant with a selective reduction in the capacity of transferred T cells to make Th17 cytokines. We conclude that sustained subpial cortical injury is associated with the capacity for brain-resident T cells to produce Th17 cytokines, and this pathological process occurs in an S1P receptor1,5-dependent manner.
Collapse
Affiliation(s)
- Lesley A Ward
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Dennis Sw Lee
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Anshu Sharma
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Angela Wang
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Xianjie I Ma
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Natalia Pikor
- Institute of Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
| | | | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
40
|
Silva BA, Leal MC, Farías MI, Erhardt B, Galeano P, Pitossi FJ, Ferrari CC. Environmental enrichment improves cognitive symptoms and pathological features in a focal model of cortical damage of multiple sclerosis. Brain Res 2020; 1727:146520. [PMID: 31669283 DOI: 10.1016/j.brainres.2019.146520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/27/2019] [Accepted: 10/20/2019] [Indexed: 10/25/2022]
Abstract
Multiple Sclerosis (MS) is a neuroinflammatory disease affecting white and grey matter, it is characterized by demyelination, axonal degeneration along with loss of motor, sensitive and cognitive functions. MS is a heterogeneous disease that displays different clinical courses: relapsing/remitting MS (RRMS), and MS progressive forms: primary progressive (PPMS) and secondary progressive (SPMS). Cortical damage in the progressive MS forms has considerable clinical relevance due to its association with cognitive impairment and disability progression in patients. One treatment is available for the progressive forms of the disease, but none are specific for cognitive deficits. We developed an animal model that reflects most of the characteristics of the cortical damage, such as cortical neuroinflammation, demyelination, neurodegeneration and meningeal inflammation, which was associated with cognitive impairment. Cognitive rehabilitation, exercise and social support have begun to be evaluated in patients and animal models of neurodegenerative diseases. Environmental enrichment (EE) provides exercise as well as cognitive and social stimulation. EE has been demonstrated to exert positive effects on cognitive domains, such as learning and memory, and improving anxiety-like symptoms. We proposed to study the effect of EE on peripherally stimulated cortical lesion induced by the long term expression of interleukin IL-1β (IL-1β) in adult rats. Here, we demonstrated that EE: 1) reduces the peripheral inflammatory response to the stimulus, 2) ameliorates cognitive deficits and anxiety-like symptoms, 3) modulates neurodegeneration, demyelination and glial activation, 4) regulates neuroinflammation by reducing the expression of pro-inflammatory cytokines and enhancing the expression of anti-inflammatory ones. Our findings correlate with the fact that EE housing could be considered an effective non- pharmacological therapeutic agent that can synergistically aid in the rehabilitation of the disease.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Institute of Translational Medicine and Biomedical Engineering of the Italian Hospital (IMTIB, CONICET), Potosí 4240, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - María Celeste Leal
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - María Isabel Farías
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Brenda Erhardt
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Pablo Galeano
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Fernando Juan Pitossi
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Institute of Translational Medicine and Biomedical Engineering of the Italian Hospital (IMTIB, CONICET), Potosí 4240, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina.
| |
Collapse
|
41
|
Fritsche L, Teuber-Hanselmann S, Soub D, Harnisch K, Mairinger F, Junker A. MicroRNA profiles of MS gray matter lesions identify modulators of the synaptic protein synaptotagmin-7. Brain Pathol 2019; 30:524-540. [PMID: 31663645 PMCID: PMC8018161 DOI: 10.1111/bpa.12800] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022] Open
Abstract
We established microRNA (miRNA) profiles in gray and white matter multiple sclerosis (MS) lesions and identified seven miRNAs which were significantly more upregulated in the gray matter lesions. Five of those seven miRNAs, miR‐330‐3p, miR‐4286, miR‐4488, let‐7e‐5p, miR‐432‐5p shared the common target synaptotagmin7 (Syt7). Immunohistochemistry and transcript analyses using nanostring technology revealed a maldistribution of Syt7, with Syt7 accumulation in neuronal soma and decreased expression in axonal structures. This maldistribution could be at least partially explained by an axonal Syt7 transport disturbance. Since Syt7 is a synapse‐associated molecule, this maldistribution could result in impairment of neuronal functions in MS patients. Thus, our results lead to the hypothesis that the overexpression of these five miRNAs in gray matter lesions is a cellular mechanism to reduce further endogenous neuronal Syt7 production. Therefore, miRNAs seem to play an important role as modulators of neuronal structures in MS.
Collapse
Affiliation(s)
- Lena Fritsche
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| | | | - Daniel Soub
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| | - Kim Harnisch
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, D-45147, Essen, Germany
| | - Andreas Junker
- Institute of Neuropathology, University Hospital Essen, D-45147, Essen, Germany
| |
Collapse
|
42
|
Banerjee PP, Pang L, Soldan SS, Miah SM, Eisenberg A, Maru S, Waldman A, Smith EA, Rosenberg-Hasson Y, Hirschberg D, Smith A, Ablashi DV, Campbell KS, Orange JS. KIR2DL4-HLAG interaction at human NK cell-oligodendrocyte interfaces regulates IFN-γ-mediated effects. Mol Immunol 2019; 115:39-55. [PMID: 30482463 PMCID: PMC6543535 DOI: 10.1016/j.molimm.2018.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/11/2018] [Accepted: 09/30/2018] [Indexed: 12/12/2022]
Abstract
Interactions between germline-encoded natural killer (NK) cell receptors and their respective ligands on tumorigenic or virus-infected cells determine NK cell cytotoxic activity and/or cytokine secretion. NK cell cytokine responses can be augmented in and can potentially contribute to multiple sclerosis (MS), an inflammatory disease of the central nervous system focused upon the oligodendrocytes (OLs). To investigate mechanisms by which NK cells may contribute to MS pathogenesis, we developed an in vitro human model of OL-NK cell interaction. We found that activated, but not resting human NK cells form conjugates with, and mediate cytotoxicity against, human oligodendrocytes. NK cells, when in conjugate with OLs, rapidly synthesize and polarize IFN-γ toward the OLs. IFN-γ is capable of reducing myelin oligodendrocyte and myelin associated glycoproteins (MOG and MAG) content. This activity is independent of MHC class-I mediated inhibition via KIR2DL1, but dependent upon the interaction between NK cell-expressed KIR2DL4 and its oligodendrocyte-expressed ligand, HLA-G. NK cells from patients with MS express higher levels of IFN-γ following conjugation to OLs, more actively promote in vitro reduction of MOG and MAG and have higher frequencies of the KIR2DL4 positive population. These data collectively suggest a mechanism by which NK cells can promote pathogenic effects upon OLs.
Collapse
Affiliation(s)
- P P Banerjee
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA.
| | - L Pang
- Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - S S Soldan
- The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA
| | - S M Miah
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - A Eisenberg
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - S Maru
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - A Waldman
- The Children's Hospital of Philadelphia Research Institute, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - E A Smith
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - Y Rosenberg-Hasson
- Human Immune Monitoring Center, Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| | - D Hirschberg
- Human Immune Monitoring Center, Stanford School of Medicine, 291 Campus Drive, Stanford, CA, 94305, USA
| | - A Smith
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| | - D V Ablashi
- Human Herpes Virus 6 Foundation, 1482 East Valley Road, Suite 619 Santa Barbara, CA 93108, USA
| | - K S Campbell
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - J S Orange
- Baylor College of Medicine, 1 Baylor Plaza, Houston, TX-77030, USA; Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates St, Houston, TX, 77030, USA
| |
Collapse
|
43
|
Eigel D, Zoupi L, Sekizar S, Welzel PB, Werner C, Williams A, Newland B. Cryogel scaffolds for regionally constrained delivery of lysophosphatidylcholine to central nervous system slice cultures: A model of focal demyelination for multiple sclerosis research. Acta Biomater 2019; 97:216-229. [PMID: 31425890 DOI: 10.1016/j.actbio.2019.08.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/26/2019] [Accepted: 08/14/2019] [Indexed: 12/11/2022]
Abstract
The pathology of multiple sclerosis (MS) is typified by focal demyelinated areas of the brain and spinal cord, which results in axonal degeneration and atrophy. Although the field has made much progress in developing immunomodulatory therapies to reduce the occurrence of these focal lesions, there is a conspicuous lack of licensed effective therapies to reduce axonal degeneration or promote repair. Remyelination, carried out by oligodendrocytes, does occur in MS, and is protective against axonal degeneration. Unfortunately, remyelination is not very efficient, and ultimately fails and so there is a research focus to generate new therapeutics to enhance remyelination leading to neuroprotection. To develop these therapies, we need preclinical models that well reflect remyelination in MS. We have previously characterized an ex vivo model that uses lysophosphatidylcholine (LPC) to cause acute and global demyelination of tissue slices, followed by spontaneous remyelination, which has been widely used as a surrogate for in vivo rodent models of demyelination. However, this ex vivo model lacks the focal demyelinated lesions seen in MS, surrounded by normal tissue from which the repairing oligodendrocytes are derived. Therefore, to improve the model, we have developed and characterized small macroporous cryogel scaffolds for controlled/regional delivery of LPC with diameters of either 0.5, 1 or 2 mm. Placement of LPC loaded scaffolds adjacent to ex vivo cultured mouse brain and spinal cord slices induced focal areas of demyelination in proximity to the scaffold. To the best of our knowledge, this is the first such report of spatial mimicry of the in vivo condition in ex vivo tissue culture. This will allow not only the investigation into focal lesions, but also provides a better platform technology with which to test remyelination-promoting therapeutics. STATEMENT OF SIGNIFICANCE: This manuscript is the first report of using macroporous hydrogels (cryogels) as a research tool for lysophosphatidylcholine (LPC) delivery, in order to create an ex vivo model of focal demyelination in the brain and spinal cord, which is of great relevance to multiple sclerosis research. Here, we transform an existing ex vivo model of demyelination by delivering LPC to focal regions of brain and spinal cord slice cultures. We have developed an easy-to-handle cylindrical and macroporous PEG-based sponge-like scaffold material (cryogel) that can deliver LPC only to a small area of the slice. Such cryogels are ideal as a delivery system in this culture model as they exhibit a soft but robust nature, with high mechanical deformability in their dry and swollen state, with no need to stay permanently hydrated. In addition, the synthesis of these cryogels is simple and easy to reproduce via photochemical cryopolymerisation using a PEG-diacrylate monomer and a photoinitiator, which are both commercially available. This more accurate model of demyelination will not only allow researchers to gain a better understanding of the CNS remyelination process in diseases such as MS, but also provides a platform technology, which could be utilized to screen and test pro-remyelination compounds which may help to find new therapeutics for progressive MS.
Collapse
Affiliation(s)
- Dimitri Eigel
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Lida Zoupi
- MRC-Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Sowmya Sekizar
- MRC-Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Petra B Welzel
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Carsten Werner
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| | - Anna Williams
- MRC-Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, EH16 4UU Edinburgh, UK.
| | - Ben Newland
- Leibniz-Institut für Polymerforschung Dresden e.V., Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany; School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
44
|
Stadelmann C, Timmler S, Barrantes-Freer A, Simons M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol Rev 2019; 99:1381-1431. [PMID: 31066630 DOI: 10.1152/physrev.00031.2018] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate multiple layers of myelin membrane around axons of the central nervous system to enable fast and efficient nerve conduction. Until recently, saltatory nerve conduction was considered the only purpose of myelin, but it is now clear that myelin has more functions. In fact, myelinating oligodendrocytes are embedded in a vast network of interconnected glial and neuronal cells, and increasing evidence supports an active role of oligodendrocytes within this assembly, for example, by providing metabolic support to neurons, by regulating ion and water homeostasis, and by adapting to activity-dependent neuronal signals. The molecular complexity governing these interactions requires an in-depth molecular understanding of how oligodendrocytes and axons interact and how they generate, maintain, and remodel their myelin sheaths. This review deals with the biology of myelin, the expanded relationship of myelin with its underlying axons and the neighboring cells, and its disturbances in various diseases such as multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Furthermore, we will highlight how specific interactions between astrocytes, oligodendrocytes, and microglia contribute to demyelination in hereditary white matter pathologies.
Collapse
Affiliation(s)
- Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Sebastian Timmler
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Mikael Simons
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| |
Collapse
|
45
|
When encephalitogenic T cells collaborate with microglia in multiple sclerosis. Nat Rev Neurol 2019; 15:704-717. [PMID: 31527807 DOI: 10.1038/s41582-019-0253-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2019] [Indexed: 01/07/2023]
Abstract
Immune cells mediate critical inflammatory and neurodegenerative processes in the CNS in individuals with multiple sclerosis (MS). In MS, activated microglia, border-associated macrophages and monocyte-derived macrophages in the CNS can encounter T cells that have infiltrated the brain parenchyma from the circulation. Although microglia and T cells both contribute to normal CNS development and homeostasis, evidence suggests that the meeting of activated microglia and macrophages with encephalitogenic T cells exacerbates their capacity to inflict injury. This crosstalk involves many cell-surface molecules, cytokines and neurotoxic factors. In this Review, we summarize the mechanisms and consequences of T cell-microglia interactions as identified with in vitro experiments and animal models, and discuss the challenges that arise when translating this preclinical knowledge to MS in humans. We also consider therapeutic approaches to MS of which the mechanisms involve prevention or modulation of T cell and microglia responses and their interactions.
Collapse
|
46
|
Singhal T, O'Connor K, Dubey S, Pan H, Chu R, Hurwitz S, Cicero S, Tauhid S, Silbersweig D, Stern E, Kijewski M, DiCarli M, Weiner HL, Bakshi R. Gray matter microglial activation in relapsing vs progressive MS: A [F-18]PBR06-PET study. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e587. [PMID: 31355321 PMCID: PMC6624145 DOI: 10.1212/nxi.0000000000000587] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/15/2019] [Indexed: 11/15/2022]
Abstract
Objective To determine the value of [F-18]PBR06-PET for assessment of microglial activation in the cerebral gray matter in patients with MS. Methods Twelve patients with MS (7 relapsing-remitting and 5 secondary progressive [SP]) and 5 healthy controls (HCs) had standardized uptake value (SUV) PET maps coregistered to 3T MRI and segmented into cortical and subcortical gray matter regions. SUV ratios (SUVRs) were global brain normalized. Voxel-by-voxel analysis was performed using statistical parametric mapping (SPM). Normalized brain parenchymal volumes (BPVs) were determined from MRI using SIENAX. Results Cortical SUVRs were higher in the hippocampus, amygdala, midcingulate, posterior cingulate, and rolandic operculum and lower in the medial-superior frontal gyrus and cuneus in the MS vs HC group (all p < 0.05). Subcortical gray matter SUVR was higher in SPMS vs RRMS (+10.8%, p = 0.002) and HC (+11.3%, p = 0.055) groups. In the MS group, subcortical gray matter SUVR correlated with the Expanded Disability Status Scale (EDSS) score (r = 0.75, p = 0.005) and timed 25-foot walk (T25FW) (r = 0.70, p = 0.01). Thalamic SUVRs increased with increasing EDSS scores (r = 0.83, p = 0.0008) and T25FW (r = 0.65, p = 0.02) and with decreasing BPV (r = -0.63, p = 0.03). Putaminal SUVRs increased with increasing EDSS scores (0.71, p = 0.009) and with decreasing BPV (r = -0.67, p = 0.01). On SPM analysis, peak correlations of thalamic voxels with BPV were seen in the pulvinar and with the EDSS score and T25FW in the dorsomedial thalamic nuclei. Conclusions This study suggests that [F-18]PBR06-PET detects widespread abnormal microglial activation in the cerebral gray matter in MS. Increased translocator protein binding in subcortical gray matter regions is associated with brain atrophy and may link to progressive MS.
Collapse
Affiliation(s)
- Tarun Singhal
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kelsey O'Connor
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shipra Dubey
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hong Pan
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Renxin Chu
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shelley Hurwitz
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Steven Cicero
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shahamat Tauhid
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David Silbersweig
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Emily Stern
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marie Kijewski
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marcelo DiCarli
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Howard L Weiner
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rohit Bakshi
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
47
|
Silva BA, Ferrari CC. Cortical and meningeal pathology in progressive multiple sclerosis: a new therapeutic target? Rev Neurosci 2019; 30:221-232. [PMID: 30048237 DOI: 10.1515/revneuro-2018-0017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/04/2018] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease that involves an intricate interaction between the central nervous system and the immune system. Nevertheless, its etiology is still unknown. MS exhibits different clinical courses: recurrent episodes with remission periods ('relapsing-remitting') that can evolve to a 'secondary progressive' form or persistent progression from the onset of the disease ('primary progressive'). The discovery of an effective treatment and cure has been hampered due to the pathological and clinical heterogeneity of the disease. Historically, MS has been considered as a disease exclusively of white matter. However, patients with progressive forms of MS present with cortical lesions associated with meningeal inflammation along with physical and cognitive disabilities. The pathogenesis of the cortical lesions has not yet been fully described. Animal models that represent both the cortical and meningeal pathologies will be critical in addressing MS pathogenesis as well as the design of specific treatments. In this review, we will address the state-of-the-art diagnostic and therapeutic alternatives and the development of strategies to discover new therapeutic approaches, especially for the progressive forms.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Potosi 4240 (C1199ABB), CABA, Buenos Aires, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435 (C1405BWE), Buenos Aires, Argentina, e-mail:
| | - Carina Cintia Ferrari
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Potosi 4240 (C1199ABB), CABA, Buenos Aires, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435 (C1405BWE), Buenos Aires, Argentina
| |
Collapse
|
48
|
Nally FK, De Santi C, McCoy CE. Nanomodulation of Macrophages in Multiple Sclerosis. Cells 2019; 8:cells8060543. [PMID: 31195710 PMCID: PMC6628349 DOI: 10.3390/cells8060543] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Multiple Sclerosis (MS) is a chronic demyelinating autoimmune disease primarily affecting young adults. Despite an unclear causal factor, symptoms and pathology arise from the infiltration of peripheral immune cells across the blood brain barrier. Accounting for the largest fraction of this infiltrate, macrophages are functionally heterogeneous innate immune cells capable of adopting either a pro or an anti-inflammatory phenotype, a phenomenon dependent upon cytokine milieu in the CNS. This functional plasticity is of key relevance in MS, where the pro-inflammatory state dominates the early stage, instructing demyelination and axonal loss while the later anti-inflammatory state holds a key role in promoting tissue repair and regeneration in later remission. This review highlights a potential therapeutic benefit of modulating macrophage polarisation to harness the anti-inflammatory and reparative state in MS. Here, we outline the role of macrophages in MS and look at the role of current FDA approved therapeutics in macrophage polarisation. Moreover, we explore the potential of particulate carriers as a novel strategy to manipulate polarisation states in macrophages, whilst examining how optimising macrophage uptake via nanoparticle size and functionalisation could offer a novel therapeutic approach for MS.
Collapse
Affiliation(s)
- Frances K Nally
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Chiara De Santi
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Claire E McCoy
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| |
Collapse
|
49
|
Ott M, Avendaño-Guzmán E, Ullrich E, Dreyer C, Strauss J, Harden M, Schön M, Schön MP, Bernhardt G, Stadelmann C, Wegner C, Brück W, Nessler S. Laquinimod, a prototypic quinoline-3-carboxamide and aryl hydrocarbon receptor agonist, utilizes a CD155-mediated natural killer/dendritic cell interaction to suppress CNS autoimmunity. J Neuroinflammation 2019; 16:49. [PMID: 30808363 PMCID: PMC6390632 DOI: 10.1186/s12974-019-1437-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/17/2019] [Indexed: 12/18/2022] Open
Abstract
Background Quinoline-3-carboxamides, such as laquinimod, ameliorate CNS autoimmunity in patients and reduce tumor cell metastasis experimentally. Previous studies have focused on the immunomodulatory effect of laquinimod on myeloid cells. The data contained herein suggest that quinoline-3-carboxamides improve the immunomodulatory and anti-tumor effects of NK cells by upregulating the adhesion molecule DNAX accessory molecule-1 (DNAM-1). Methods We explored how NK cell activation by laquinimod inhibits CNS autoimmunity in experimental autoimmune encephalomyelitis (EAE), the most utilized model of MS, and improves immunosurveillance of experimental lung melanoma metastasis. Functional manipulations included in vivo NK and DC depletion experiments and in vitro assays of NK cell function. Clinical, histological, and flow cytometric read-outs were assessed. Results We demonstrate that laquinimod activates natural killer (NK) cells via the aryl hydrocarbon receptor and increases their DNAM-1 cell surface expression. This activation improves the cytotoxicity of NK cells against B16F10 melanoma cells and augments their immunoregulatory functions in EAE by interacting with CD155+ dendritic cells (DC). Noteworthy, the immunosuppressive effect of laquinimod-activated NK cells was due to decreasing MHC class II antigen presentation by DC and not by increasing DC killing. Conclusions This study clarifies how DNAM-1 modifies the bidirectional crosstalk of NK cells with CD155+ DC, which can be exploited to suppress CNS autoimmunity and strengthen tumor surveillance. Electronic supplementary material The online version of this article (10.1186/s12974-019-1437-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martina Ott
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Erika Avendaño-Guzmán
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Evelyn Ullrich
- LOEWE Center for Cell and Gene Therapy, Goethe University, Frankfurt am Main, Germany.,Division of Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, Hospital of the Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Carolin Dreyer
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Judith Strauss
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Harden
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Margarete Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany.,Lower Saxony Institute of Occupational Dermatology, University Medical Center Göttingen and University of Osnabrück, Göttingen, Germany
| | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Straße 1, Gebäude I11 OE 5240, 30625, Hannover, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christiane Wegner
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Present Address: Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Nessler
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
50
|
Correale J, Marrodan M, Ysrraelit MC. Mechanisms of Neurodegeneration and Axonal Dysfunction in Progressive Multiple Sclerosis. Biomedicines 2019; 7:biomedicines7010014. [PMID: 30791637 PMCID: PMC6466454 DOI: 10.3390/biomedicines7010014] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/14/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Multiple Sclerosis (MS) is a major cause of neurological disability, which increases predominantly during disease progression as a result of cortical and grey matter structures involvement. The gradual accumulation of disability characteristic of the disease seems to also result from a different set of mechanisms, including in particular immune reactions confined to the Central Nervous System such as: (a) B-cell dysregulation, (b) CD8+ T cells causing demyelination or axonal/neuronal damage, and (c) microglial cell activation associated with neuritic transection found in cortical demyelinating lesions. Other potential drivers of neurodegeneration are generation of oxygen and nitrogen reactive species, and mitochondrial damage, inducing impaired energy production, and intra-axonal accumulation of Ca2+, which in turn activates a variety of catabolic enzymes ultimately leading to progressive proteolytic degradation of cytoskeleton proteins. Loss of axon energy provided by oligodendrocytes determines further axonal degeneration and neuronal loss. Clearly, these different mechanisms are not mutually exclusive and could act in combination. Given the multifactorial pathophysiology of progressive MS, many potential therapeutic targets could be investigated in the future. This remains however, an objective that has yet to be undertaken.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, FLENI, Buenos Aires 1428, Argentina.
| | | | | |
Collapse
|