1
|
Amatruda M, Marechal D, Gacias M, Wentling M, Turpin-Nolan S, Morstein J, Moniruzzaman M, Brüning JC, Haughey NJ, Trauner DH, Casaccia P. Neuroprotective effect of neuron-specific deletion of the C16 ceramide synthetic enzymes in an animal model of multiple sclerosis. Glia 2025; 73:271-290. [PMID: 39489703 DOI: 10.1002/glia.24631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/05/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
Ceramide C16 is a sphingolipid detected at high levels in several neurodegenerative disorders, including multiple sclerosis (MS). It can be generated de novo or from the hydrolysis of other sphingolipids, such as sphingomyelin or through the recycling of sphingosine, in what is known as the salvage pathway. While the myelin damage occurring in MS suggests the importance of the hydrolytic and salvage pathways, the growing interest on the importance of diet in demyelinating disorders, prompted us to investigate the involvement of de novo ceramide C16 synthesis on disease severity. A diet rich in saturated fats such as palmitic acid, as found in many highly processed foods, provides substrates for the ceramide C16 synthetic enzymes ceramide synthase 6 (CERS6) and 5 (CERS5), which are expressed in the central nervous system. Using the experimental autoimmune encephalomyelitis (EAE) model of inflammatory demyelination, we show here that mice with CamK2a+ neuronal specific deletion of both CerS6 and CerS5 show a milder course of EAE than wild type mice, even when fed a diet enriched in palmitic acid. At a cellular level, neurons lacking both CerS6 and CerS5 are protected from the mitochondrial dysfunction arising from exposure to oxidative stress and palmitic acid in the medium. These data underscore the importance of a healthy diet avoiding processed foods for demyelinating disorders and identifies endogenous neuronal synthesis of ceramide C16 as an important determinant of disease severity.
Collapse
Affiliation(s)
- Mario Amatruda
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
| | - Damien Marechal
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
| | - Mar Gacias
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maureen Wentling
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
| | - Sarah Turpin-Nolan
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Cellular and Molecular Metabolism Laboratory, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Johannes Morstein
- Department of Chemistry, New York University, Silver Center, New York, New York, USA
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, California, USA
| | - Mohammed Moniruzzaman
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dirk H Trauner
- Department of Chemistry, New York University, Silver Center, New York, New York, USA
- Chemistry and Translational Therapeutics and Systems Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patrizia Casaccia
- Neuroscience Initiative, Advanced Science Research Center, CUNY, New York, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Program in Biochemistry, The Graduate Center of The City University of New York, New York, New York, USA
- Program in Biology, The Graduate Center of The City University of New York, New York, New York, USA
| |
Collapse
|
2
|
Maktabi B, Collins A, Safee R, Bouyer J, Wisner AS, Williams FE, Schiefer IT. Zebrafish as a Model for Multiple Sclerosis. Biomedicines 2024; 12:2354. [PMID: 39457666 PMCID: PMC11504653 DOI: 10.3390/biomedicines12102354] [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: 09/13/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Zebrafish have become a key model organism in neuroscience research because of their unique advantages. Their genetic, anatomical, and physiological similarities to humans, coupled with their rapid development and transparent embryos, make them an excellent tool for investigating various aspects of neurobiology. They have specifically emerged as a valuable and versatile model organism in biomedical research, including the study of neurological disorders such as multiple sclerosis. Multiple sclerosis is a chronic autoimmune disease known to cause damage to the myelin sheath that protects the nerves in the brain and spinal cord. Objective: This review emphasizes the importance of continued research in both in vitro and in vivo models to advance our understanding of MS and develop effective treatments, ultimately improving the quality of life for those affected by this debilitating disease. Conclusions: Recent studies show the significance of zebrafish as a model organism for investigating demyelination and remyelination processes, providing new insights into MS pathology and potential therapies.
Collapse
Affiliation(s)
- Briana Maktabi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Abigail Collins
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Raihaanah Safee
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
| | - Jada Bouyer
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Alexander S. Wisner
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Frederick E. Williams
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Isaac T. Schiefer
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
3
|
Wang M, Caryotakis SE, Smith GG, Nguyen AV, Pleasure DE, Soulika AM. CSF1R antagonism results in increased supraspinal infiltration in EAE. J Neuroinflammation 2024; 21:103. [PMID: 38643194 PMCID: PMC11031888 DOI: 10.1186/s12974-024-03063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 03/11/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Colony stimulating factor 1 receptor (CSF1R) signaling is crucial for the maintenance and function of various myeloid subsets. CSF1R antagonism was previously shown to mitigate clinical severity in experimental autoimmune encephalomyelitis (EAE). The associated mechanisms are still not well delineated. METHODS To assess the effect of CSF1R signaling, we employed the CSF1R antagonist PLX5622 formulated in chow (PLX5622 diet, PD) and its control chow (control diet, CD). We examined the effect of PD in steady state and EAE by analyzing cells isolated from peripheral immune organs and from the CNS via flow cytometry. We determined CNS infiltration sites and assessed the extent of demyelination using immunohistochemistry of cerebella and spinal cords. Transcripts of genes associated with neuroinflammation were also analyzed in these tissues. RESULTS In addition to microglial depletion, PD treatment reduced dendritic cells and macrophages in peripheral immune organs, both during steady state and during EAE. Furthermore, CSF1R antagonism modulated numbers and relative frequencies of T effector cells both in the periphery and in the CNS during the early stages of the disease. Classical neurological symptoms were milder in PD compared to CD mice. Interestingly, a subset of PD mice developed atypical EAE symptoms. Unlike previous studies, we observed that the CNS of PD mice was infiltrated by increased numbers of peripheral immune cells compared to that of CD mice. Immunohistochemical analysis showed that CNS infiltrates in PD mice were mainly localized in the cerebellum while in CD mice infiltrates were primarily localized in the spinal cords during the onset of neurological deficits. Accordingly, during the same timepoint, cerebella of PD but not of CD mice had extensive demyelinating lesions, while spinal cords of CD but not of PD mice were heavily demyelinated. CONCLUSIONS Our findings suggest that CSF1R activity modulates the cellular composition of immune cells both in the periphery and within the CNS, and affects lesion localization during the early EAE stages.
Collapse
Affiliation(s)
- Marilyn Wang
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Sofia E Caryotakis
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
- University of California, San Francisco, San Francisco, CA, USA
| | - Glendalyn G Smith
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
| | - Alan V Nguyen
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Sutro Biosciences, South San Francisco, CA, USA
| | - David E Pleasure
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Athena M Soulika
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA.
- Shriners Hospitals for Children, Northern California, Sacramento, CA, USA.
| |
Collapse
|
4
|
Peck T, Davis C, Lenihan-Geels G, Griffiths M, Spijkers-Shaw S, Zubkova OV, La Flamme AC. The novel HS-mimetic, Tet-29, regulates immune cell trafficking across barriers of the CNS during inflammation. J Neuroinflammation 2023; 20:251. [PMID: 37915090 PMCID: PMC10619265 DOI: 10.1186/s12974-023-02925-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Disruption of the extracellular matrix at the blood-brain barrier (BBB) underpins neuroinflammation in multiple sclerosis (MS). The degradation of extracellular matrix components, such as heparan sulfate (HS) proteoglycans, can be prevented by treatment with HS-mimetics through their ability to inhibit the enzyme heparanase. The heparanase-inhibiting ability of our small dendrimer HS-mimetics has been investigated in various cancers but their efficacy in neuroinflammatory models has not been evaluated. This study investigates the use of a novel HS-mimetic, Tet-29, in an animal model of MS. METHODS Neuroinflammation was induced in mice by experimental autoimmune encephalomyelitis, a murine model of MS. In addition, the BBB and choroid plexus were modelled in vitro using transmigration assays, and migration of immune cells in vivo and in vitro was quantified by flow cytometry. RESULTS We found that Tet-29 significantly reduced lymphocyte accumulation in the central nervous system which, in turn, decreased disease severity in experimental autoimmune encephalomyelitis. The disease-modifying effect of Tet-29 was associated with a rescue of BBB integrity, as well as inhibition of activated lymphocyte migration across the BBB and choroid plexus in transwell models. In contrast, Tet-29 did not significantly impair in vivo or in vitro steady state-trafficking under homeostatic conditions. CONCLUSIONS Together these results suggest that Tet-29 modulates, rather than abolishes, trafficking across central nervous system barriers.
Collapse
Affiliation(s)
- Tessa Peck
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand
| | - Connor Davis
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand
| | - Georgia Lenihan-Geels
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand
| | - Maddie Griffiths
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
- Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand
| | - Sam Spijkers-Shaw
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Olga V Zubkova
- Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Anne Camille La Flamme
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.
- Centre for Biodiscovery Wellington, Victoria University of Wellington, Wellington, New Zealand.
- Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
5
|
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: 1] [Impact Index Per Article: 1.0] [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
|
6
|
Packer D, Fresenko EE, Harrington EP. Remyelination in animal models of multiple sclerosis: finding the elusive grail of regeneration. Front Mol Neurosci 2023; 16:1207007. [PMID: 37448959 PMCID: PMC10338073 DOI: 10.3389/fnmol.2023.1207007] [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: 04/16/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Remyelination biology and the therapeutic potential of restoring myelin sheaths to prevent neurodegeneration and disability in multiple sclerosis (MS) has made considerable gains over the past decade with many regeneration strategies undergoing tested in MS clinical trials. Animal models used to investigate oligodendroglial responses and regeneration of myelin vary considerably in the mechanism of demyelination, involvement of inflammatory cells, neurodegeneration and capacity for remyelination. The investigation of remyelination in the context of aging and an inflammatory environment are of considerable interest for the potential translation to progressive multiple sclerosis. Here we review how remyelination is assessed in mouse models of demyelination, differences and advantages of these models, therapeutic strategies that have emerged and current pro-remyelination clinical trials.
Collapse
|
7
|
Makar TK, Guda PR, Ray S, Andhavarapu S, Keledjian K, Gerzanich V, Simard JM, Nimmagadda VKC, Bever CT. Immunomodulatory therapy with glatiramer acetate reduces endoplasmic reticulum stress and mitochondrial dysfunction in experimental autoimmune encephalomyelitis. Sci Rep 2023; 13:5635. [PMID: 37024509 PMCID: PMC10079956 DOI: 10.1038/s41598-023-29852-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 02/11/2023] [Indexed: 04/08/2023] Open
Abstract
Endoplasmic reticulum (ER) stress and mitochondrial dysfunction are found in lesions of multiple sclerosis (MS) and animal models of MS such as experimental autoimmune encephalomyelitis (EAE), and may contribute to the neuronal loss that underlies permanent impairment. We investigated whether glatiramer acetate (GA) can reduce these changes in the spinal cords of chronic EAE mice by using routine histology, immunostaining, and electron microscopy. EAE spinal cord tissue exhibited increased inflammation, demyelination, mitochondrial dysfunction, ER stress, downregulation of NAD+ dependent pathways, and increased neuronal death. GA reversed these pathological changes, suggesting that immunomodulating therapy can indirectly induce neuroprotective effects in the CNS by mediating ER stress.
Collapse
Affiliation(s)
- Tapas K Makar
- Department of Neurology, School of Medicine, University of Maryland, College Park, USA.
- Research Service, Institute of Human Virology, VA Maryland Health Care System, 725 W Lombard St, Baltimore, MD, 21201, USA.
| | - Poornachander R Guda
- Department of Neurology, School of Medicine, University of Maryland, College Park, USA
| | - Sugata Ray
- Department of Neurology, School of Medicine, University of Maryland, College Park, USA
| | - Sanketh Andhavarapu
- Department of Neurology, School of Medicine, University of Maryland, College Park, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, School of Medicine, University of Maryland, College Park, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, School of Medicine, University of Maryland, College Park, USA
| | - J Marc Simard
- Department of Neurosurgery, School of Medicine, University of Maryland, College Park, USA
| | - Vamshi K C Nimmagadda
- Department of Neurology, School of Medicine, University of Maryland, College Park, USA
| | - Christopher T Bever
- Department of Neurology, School of Medicine, University of Maryland, College Park, USA
- Research Service, Institute of Human Virology, VA Maryland Health Care System, 725 W Lombard St, Baltimore, MD, 21201, USA
- Department of Veterans Affairs, Office of Research and Development, Washington, USA
| |
Collapse
|
8
|
Jahanbazi Jahan-Abad A, Salapa HE, Libner CD, Thibault PA, Levin MC. hnRNP A1 dysfunction in oligodendrocytes contributes to the pathogenesis of multiple sclerosis. Glia 2023; 71:633-647. [PMID: 36382566 DOI: 10.1002/glia.24300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/17/2022]
Abstract
Oligodendrocyte (OL) damage and death are prominent features of multiple sclerosis (MS) pathology, yet mechanisms contributing to OL loss are incompletely understood. Dysfunctional RNA binding proteins (RBPs), hallmarked by nucleocytoplasmic mislocalization and altered expression, have been shown to result in cell loss in neurologic diseases, including in MS. Since we previously observed that the RBP heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) was dysfunctional in neurons in MS, we hypothesized that it might also contribute to OL pathology in MS and relevant models. We discovered that hnRNP A1 dysfunction is characteristic of OLs in MS brains. These findings were recapitulated in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS, where hnRNP A1 dysfunction was characteristic of OLs, including oligodendrocyte precursor cells and mature OLs in which hnRNP A1 dysfunction correlated with demyelination. We also found that hnRNP A1 dysfunction was induced by IFNγ, indicating that inflammation influences hnRNP A1 function. To fully understand the effects of hnRNP A1 dysfunction on OLs, we performed siRNA knockdown of hnRNP A1, followed by RNA sequencing. RNA sequencing detected over 4000 differentially expressed transcripts revealing alterations to RNA metabolism, cell morphology, and programmed cell death pathways. We confirmed that hnRNP A1 knockdown was detrimental to OLs and induced apoptosis and necroptosis. Together, these data demonstrate a critical role for hnRNP A1 in proper OL functioning and survival and suggest a potential mechanism of OL damage and death in MS that involves hnRNP A1 dysfunction.
Collapse
Affiliation(s)
- Ali Jahanbazi Jahan-Abad
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hannah E Salapa
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cole D Libner
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Patricia A Thibault
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Michael C Levin
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
9
|
Di Mauro G, Amoriello R, Lozano N, Carnasciali A, Guasti D, Becucci M, Cellot G, Kostarelos K, Ballerini C, Ballerini L. Graphene Oxide Nanosheets Reduce Astrocyte Reactivity to Inflammation and Ameliorate Experimental Autoimmune Encephalomyelitis. ACS NANO 2023; 17:1965-1978. [PMID: 36692902 PMCID: PMC9933621 DOI: 10.1021/acsnano.2c06609] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/10/2023] [Indexed: 06/17/2023]
Abstract
In neuroinflammation, astrocytes play multifaceted roles that regulate the neuronal environment. Astrocytes sense and respond to pro-inflammatory cytokines (CKs) and, by a repertoire of intracellular Ca2+ signaling, contribute to disease progression. Therapeutic approaches wish to reduce the overactivation in Ca2+ signaling in inflammatory-reactive astrocytes to restore dysregulated cellular changes. Cell-targeting therapeutics might take advantage by the use of nanomaterial-multifunctional platforms such as graphene oxide (GO). GO biomedical applications in the nervous system involve therapeutic delivery and sensing, and GO flakes were shown to enable interfacing of neuronal and glial membrane dynamics. We exploit organotypic spinal cord cultures and optical imaging to explore Ca2+ changes in astrocytes, and we report, when spinal tissue is exposed to CKs, neuroinflammatory-associated modulation of resident glia. We show the efficacy of GO to revert these dynamic changes in astrocytic reactivity to CKs, and we translate this potential in an animal model of immune-mediated neuroinflammatory disease.
Collapse
Affiliation(s)
- Giuseppe Di Mauro
- International
School for Advanced Studies (SISSA/ISAS), 34136Trieste, Italy
| | - Roberta Amoriello
- International
School for Advanced Studies (SISSA/ISAS), 34136Trieste, Italy
- Dipartimento
di Medicina Sperimentale e Clinica, University
of Florence, 50139Florence, Italy
| | - Neus Lozano
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), 08193Barcelona, Spain
| | - Alberto Carnasciali
- Dipartimento
di Medicina Sperimentale e Clinica, University
of Florence, 50139Florence, Italy
| | - Daniele Guasti
- Dipartimento
di Medicina Sperimentale e Clinica, University
of Florence, 50139Florence, Italy
| | - Maurizio Becucci
- Dipartimento
di Chimica “Ugo Schiff”, DICUS, University of Florence, 50139Florence, Italy
| | - Giada Cellot
- International
School for Advanced Studies (SISSA/ISAS), 34136Trieste, Italy
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), 08193Barcelona, Spain
- Nanomedicine
Lab, and Faculty of Biology, Medicine & Health, The National Graphene
Institute, University of Manchester, ManchesterM13 9PL, United Kingdom
| | - Clara Ballerini
- Dipartimento
di Medicina Sperimentale e Clinica, University
of Florence, 50139Florence, Italy
| | - Laura Ballerini
- International
School for Advanced Studies (SISSA/ISAS), 34136Trieste, Italy
| |
Collapse
|
10
|
Gharagozloo M, Mace JW, Calabresi PA. Animal models to investigate the effects of inflammation on remyelination in multiple sclerosis. Front Mol Neurosci 2022; 15:995477. [PMID: 36407761 PMCID: PMC9669474 DOI: 10.3389/fnmol.2022.995477] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/22/2022] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS). In people with MS, impaired remyelination and axonal loss lead to debilitating long-term neurologic deficits. Current MS disease-modifying drugs mainly target peripheral immune cells and have demonstrated little efficacy for neuroprotection or promoting repair. To elucidate the pathological mechanisms and test therapeutic interventions, multiple animal models have been developed to recapitulate specific aspects of MS pathology, particularly the acute inflammatory stage. However, there are few animal models that facilitate the study of remyelination in the presence of inflammation, and none fully replicate the biology of chronic demyelination in MS. In this review, we describe the animal models that have provided insight into the mechanisms underlying demyelination, myelin repair, and potential therapeutic targets for remyelination. We highlight the limitations of studying remyelination in toxin-based demyelination models and discuss the combinatorial models that recapitulate the inflammatory microenvironment, which is now recognized to be a major inhibitor of remyelination mechanisms. These models may be useful in identifying novel therapeutics that promote CNS remyelination in inflammatory diseases such as MS.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jackson W. Mace
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
11
|
Zha Z, Liu S, Liu Y, Li C, Wang L. Potential Utility of Natural Products against Oxidative Stress in Animal Models of Multiple Sclerosis. Antioxidants (Basel) 2022; 11:antiox11081495. [PMID: 36009214 PMCID: PMC9404913 DOI: 10.3390/antiox11081495] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune-mediated degenerative disease of the central nervous system (CNS) characterized by immune cell infiltration, demyelination and axonal injury. Oxidative stress-induced inflammatory response, especially the destructive effect of immune cell-derived free radicals on neurons and oligodendrocytes, is crucial in the onset and progression of MS. Therefore, targeting oxidative stress-related processes may be a promising preventive and therapeutic strategy for MS. Animal models, especially rodent models, can be used to explore the in vivo molecular mechanisms of MS considering their similarity to the pathological processes and clinical signs of MS in humans and the significant oxidative damage observed within their CNS. Consequently, these models have been used widely in pre-clinical studies of oxidative stress in MS. To date, many natural products have been shown to exert antioxidant effects to attenuate the CNS damage in animal models of MS. This review summarized several common rodent models of MS and their association with oxidative stress. In addition, this review provides a comprehensive and concise overview of previously reported natural antioxidant products in inhibiting the progression of MS.
Collapse
|
12
|
Wang Y, Pleasure D, Deng W, Guo F. Therapeutic Potentials of Poly (ADP-Ribose) Polymerase 1 (PARP1) Inhibition in Multiple Sclerosis and Animal Models: Concept Revisiting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102853. [PMID: 34935305 PMCID: PMC8844485 DOI: 10.1002/advs.202102853] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/12/2021] [Indexed: 05/05/2023]
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) plays a fundamental role in DNA repair and gene expression. Excessive PARP1 hyperactivation, however, has been associated with cell death. PARP1 and/or its activity are dysregulated in the immune and central nervous system of multiple sclerosis (MS) patients and animal models. Pharmacological PARP1 inhibition is shown to be protective against immune activation and disease severity in MS animal models while genetic PARP1 deficiency studies reported discrepant results. The inconsistency suggests that the function of PARP1 and PARP1-mediated PARylation may be complex and context-dependent. The article reviews PARP1 functions, discusses experimental findings and possible interpretations of PARP1 in inflammation, neuronal/axonal degeneration, and oligodendrogliopathy, three major pathological components cooperatively determining MS disease course and neurological progression, and points out future research directions. Cell type specific PARP1 manipulations are necessary for revisiting the role of PARP1 in the three pathological components prior to moving PARP1 inhibition into clinical trials for MS therapy.
Collapse
Affiliation(s)
- Yan Wang
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| | - David Pleasure
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityGuangzhou510006China
| | - Fuzheng Guo
- Department of NeurologySchool of MedicineUniversity of CaliforniaDavisCA95817USA
- Institute for Pediatric Regenerative MedicineUC Davis School of Medicine/Shriners Hospitals for ChildrenSacramentoCAUSA
| |
Collapse
|
13
|
Osipchuk NC, Soulika AM, Fomina AF. Modulation of Ryanodine Receptors Activity Alters the Course of Experimental Autoimmune Encephalomyelitis in Mice. Front Physiol 2022; 12:770820. [PMID: 35027891 PMCID: PMC8751758 DOI: 10.3389/fphys.2021.770820] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/31/2021] [Indexed: 11/22/2022] Open
Abstract
Ryanodine receptors (RyRs), the intracellular Ca2+ release channels, are expressed in T lymphocytes and other types of immune cells. Modulation of RyRs has been shown to affect T cell functions in vitro and immune responses in vivo. The effects of modulation of RyRs on the development of autoimmune diseases have not been investigated. Here we studied how modulation of RyRs through administration of RyR inhibitor dantrolene or introducing a gain-of-function RYR1-p.R163C mutation affects clinical progression of experimental autoimmune encephalomyelitis (EAE) in mice, a T cell-mediated autoimmune neuroinflammatory disease. We found that daily intraperitoneal administration of 5 or 10 mg/kg dantrolene beginning at the time of EAE induction significantly reduced the severity of EAE clinical symptoms and dampened inflammation in the spinal cord. The protective effect of dantrolene on EAE was reversible. Dantrolene administration elicited dose-dependent skeletal muscle weakness: mice that received 10 mg/kg dose developed a waddling gait, while 5 mg/kg dantrolene dose administration produced a reduction in four-limb holding impulse values. Mice bearing the gain-of-function RYR1-p.R163C mutation developed the EAE clinical symptoms faster and more severely than wild-type mice. This study demonstrates that RyRs play a significant role in EAE pathogenesis and suggests that inhibition of RyRs with low doses of dantrolene may have a protective effect against autoimmunity and inflammation in humans.
Collapse
Affiliation(s)
- Natalia C Osipchuk
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Athena M Soulika
- Shriners Hospitals for Children Northern California, Institute for Pediatric Regenerative Research, Sacramento, CA, United States.,Department of Dermatology, University of California, Davis, Davis, CA, United States
| | - Alla F Fomina
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
14
|
Li H, Liu S, Han J, Li S, Gao X, Wang M, Zhu J, Jin T. Role of Toll-Like Receptors in Neuroimmune Diseases: Therapeutic Targets and Problems. Front Immunol 2021; 12:777606. [PMID: 34790205 PMCID: PMC8591135 DOI: 10.3389/fimmu.2021.777606] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/15/2021] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptors (TLRs) are a class of proteins playing a key role in innate and adaptive immune responses. TLRs are involved in the development and progression of neuroimmune diseases via initiating inflammatory responses. Thus, targeting TLRs signaling pathway may be considered as a potential therapy for neuroimmune diseases. However, the role of TLRs is elusive and complex in neuroimmune diseases. In addition to the inadequate immune response of TLRs inhibitors in the experiments, the recent studies also demonstrated that partial activation of TLRs is conducive to the production of anti-inflammatory factors and nervous system repair. Exploring the mechanism of TLRs in neuroimmune diseases and combining with developing the emerging drug may conquer neuroimmune diseases in the future. Herein, we provide an overview of the role of TLRs in several neuroimmune diseases, including multiple sclerosis, neuromyelitis optica spectrum disorder, Guillain-Barré syndrome and myasthenia gravis. Emerging difficulties and potential solutions in clinical application of TLRs inhibitors will also be discussed.
Collapse
Affiliation(s)
- Haixia Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Shan Liu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Shengxian Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyan Gao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meng Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital, Solna, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Chitu V, Biundo F, Stanley ER. Colony stimulating factors in the nervous system. Semin Immunol 2021; 54:101511. [PMID: 34743926 DOI: 10.1016/j.smim.2021.101511] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Although traditionally seen as regulators of hematopoiesis, colony-stimulating factors (CSFs) have emerged as important players in the nervous system, both in health and disease. This review summarizes the cellular sources, patterns of expression and physiological roles of the macrophage (CSF-1, IL-34), granulocyte-macrophage (GM-CSF) and granulocyte (G-CSF) colony stimulating factors within the nervous system, with a particular focus on their actions on microglia. CSF-1 and IL-34, via the CSF-1R, are required for the development, proliferation and maintenance of essentially all CNS microglia in a temporal and regional specific manner. In contrast, in steady state, GM-CSF and G-CSF are mainly involved in regulation of microglial function. The alterations in expression of these growth factors and their receptors, that have been reported in several neurological diseases, are described and the outcomes of their therapeutic targeting in mouse models and humans are discussed.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
16
|
Mei Y, Yang J, Yuan Y, Liu Y, Liu X, Li M, Fan S, Li L, Jiang C, Xu Y. Systemic Inflammation Index Values Are Associated With Worsened Disease Severity and Poor Response to Autoimmune Encephalitis Treatment. Front Neurol 2021; 12:709553. [PMID: 34675864 PMCID: PMC8523674 DOI: 10.3389/fneur.2021.709553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/03/2021] [Indexed: 01/15/2023] Open
Abstract
Both specific and innate immune responses play important roles in autoimmune encephalitis (AE). We aimed to explore the predictive value of the systemic inflammation index (SII) at admission as a peripheral biomarker of treatment response of AE. A total of 146 patients diagnosed with AE in the First Affiliated Hospital of Zhengzhou University from January 1, 2018 to September 22, 2020 were retrospectively and consecutively analyzed as per the inclusion criteria and divided into two groups according to their response to immunotherapy after 30 days. The predictive value of the SII as a peripheral biomarker for AE treatment response was calculated using the receiver operating characteristic curve analysis, which showed that the best SII cut-off value for predicting poor response to AE treatment was 863.3; the area under the curve was 0.75, with 83.0% sensitivity and 72.0% specificity. The risk factors for poor response to AE treatment were analyzed; univariable analysis showed that the rate of decreased level of consciousness, rate of cognitive or mental behavior abnormality, cerebrospinal fluid pressure, blood neutrophils, platelets, time until treatment initiation, neutrophil to lymphocyte ratio, platelet to lymphocyte ratio, and SII were significantly higher in patients with poor response to AE immunotherapy after 30 days than in patients with good response. Meanwhile, the blood lymphocyte counts and Glasgow Coma Scale (GCS) scores in patients with poor response were significantly lower than those in patients with good response (all p < 0.05), and multivariable binary logistic regression with backward stepwise method showed that decreased levels of consciousness, time until treatment initiation and SII were associated with poor response to immunotherapy. Moreover, the SII ≤ 863.3 group had lower rates of decreased consciousness levels, admission to the intensive care unit, and mechanical ventilation; lower cerebrospinal fluid pressure, blood neutrophil count, and platelet count; and higher blood lymphocyte count and GCS scores. The SII was associated with worsened disease severity and poor response to treatment after 30 days of the initially diagnosed AE, and patients with an SII > 863.3 were more likely to have poor response to immunotherapy.
Collapse
Affiliation(s)
- Yanliang Mei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanpeng Yuan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,National Health Commission Key Laboratory of Cerebrovascular Disease, Zhengzhou University, Zhengzhou, China
| | - Yutao Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingli Li
- National Health Commission Key Laboratory of Cerebrovascular Disease, Zhengzhou University, Zhengzhou, China
| | - Shiheng Fan
- National Health Commission Key Laboratory of Cerebrovascular Disease, Zhengzhou University, Zhengzhou, China
| | - Lanjun Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chenyang Jiang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,National Health Commission Key Laboratory of Cerebrovascular Disease, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Dias DO, Kalkitsas J, Kelahmetoglu Y, Estrada CP, Tatarishvili J, Holl D, Jansson L, Banitalebi S, Amiry-Moghaddam M, Ernst A, Huttner HB, Kokaia Z, Lindvall O, Brundin L, Frisén J, Göritz C. Pericyte-derived fibrotic scarring is conserved across diverse central nervous system lesions. Nat Commun 2021; 12:5501. [PMID: 34535655 PMCID: PMC8448846 DOI: 10.1038/s41467-021-25585-5] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/17/2021] [Indexed: 12/04/2022] Open
Abstract
Fibrotic scar tissue limits central nervous system regeneration in adult mammals. The extent of fibrotic tissue generation and distribution of stromal cells across different lesions in the brain and spinal cord has not been systematically investigated in mice and humans. Furthermore, it is unknown whether scar-forming stromal cells have the same origin throughout the central nervous system and in different types of lesions. In the current study, we compared fibrotic scarring in human pathological tissue and corresponding mouse models of penetrating and non-penetrating spinal cord injury, traumatic brain injury, ischemic stroke, multiple sclerosis and glioblastoma. We show that the extent and distribution of stromal cells are specific to the type of lesion and, in most cases, similar between mice and humans. Employing in vivo lineage tracing, we report that in all mouse models that develop fibrotic tissue, the primary source of scar-forming fibroblasts is a discrete subset of perivascular cells, termed type A pericytes. Perivascular cells with a type A pericyte marker profile also exist in the human brain and spinal cord. We uncover type A pericyte-derived fibrosis as a conserved mechanism that may be explored as a therapeutic target to improve recovery after central nervous system lesions.
Collapse
Affiliation(s)
- David O Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jannis Kalkitsas
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yildiz Kelahmetoglu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Cynthia P Estrada
- Department of Clinical Neuroscience, Karolinska University Hospital, Solna, Sweden
| | | | - Daniel Holl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Linda Jansson
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Shervin Banitalebi
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Aurélie Ernst
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Group Genome Instability in Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Hagen B Huttner
- Department of Neurology, University Hospital Erlangen, Erlangen, Germany
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Olle Lindvall
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Lou Brundin
- Department of Clinical Neuroscience, Karolinska University Hospital, Solna, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
- Ming Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
Li Q, Fu N, Han Y, Qin J. Pediatric Autoimmune Encephalitis and Its Relationship With Infection. Pediatr Neurol 2021; 120:27-32. [PMID: 33964702 DOI: 10.1016/j.pediatrneurol.2021.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/31/2022]
Abstract
Autoimmune encephalitis (AE) is an increasingly recognized inflammatory disorder of the central nervous system and is most often characterized by antibodies against intracellular and neuronal surface antigens. AE is a devastating disease that may result in developmental delay or regression in children. However, the pathogenesis of AE is not clear, and immune system disorders after infection likely play an important role in AE. Many studies have reported that patients with herpes simplex virus encephalitis develop anti-N-methyl-d-aspartate receptor encephalitis after antiviral treatment. It is critical to recognize pediatric AE early and to distinguish it from infectious forms because AE is treatable and responsive to immunotherapies. In this review, we discuss the clinical features of pediatric AE and focus on the relationship between AE and postinfection status. In addition, we review the probable mechanisms underlying infection-triggered AE, which include molecular mimicry, bystander activation, epitope spreading, immune system disorder, and genetic susceptibility.
Collapse
Affiliation(s)
- Qinrui Li
- Department of Pediatrics, Peking University People's Hospital, Beijing, P.R. China
| | - Na Fu
- Department of Pediatrics, Peking University People's Hospital, Beijing, P.R. China
| | - Ying Han
- Department of Pediatrics, Peking University First Hospital, Beijing, P.R. China.
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, P.R. China.
| |
Collapse
|
19
|
Deerhake ME, Danzaki K, Inoue M, Cardakli ED, Nonaka T, Aggarwal N, Barclay WE, Ji RR, Shinohara ML. Dectin-1 limits autoimmune neuroinflammation and promotes myeloid cell-astrocyte crosstalk via Card9-independent expression of Oncostatin M. Immunity 2021; 54:484-498.e8. [PMID: 33581044 PMCID: PMC7956124 DOI: 10.1016/j.immuni.2021.01.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 11/20/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
Pathologic roles of innate immunity in neurologic disorders are well described, but their beneficial aspects are less understood. Dectin-1, a C-type lectin receptor (CLR), is largely known to induce inflammation. Here, we report that Dectin-1 limited experimental autoimmune encephalomyelitis (EAE), while its downstream signaling molecule, Card9, promoted the disease. Myeloid cells mediated the pro-resolution function of Dectin-1 in EAE with enhanced gene expression of the neuroprotective molecule, Oncostatin M (Osm), through a Card9-independent pathway, mediated by the transcription factor NFAT. Furthermore, we find that the Osm receptor (OsmR) functioned specifically in astrocytes to reduce EAE severity. Notably, Dectin-1 did not respond to heat-killed Mycobacteria, an adjuvant to induce EAE. Instead, endogenous Dectin-1 ligands, including galectin-9, in the central nervous system (CNS) were involved to limit EAE. Our study reveals a mechanism of beneficial myeloid cell-astrocyte crosstalk regulated by a Dectin-1 pathway and identifies potential therapeutic targets for autoimmune neuroinflammation.
Collapse
MESH Headings
- Animals
- Astrocytes/immunology
- Brain/pathology
- CARD Signaling Adaptor Proteins/metabolism
- Cell Communication
- Cells, Cultured
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Galectins/metabolism
- Gene Expression Regulation
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Multiple Sclerosis/immunology
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Myeloid Cells/immunology
- Neurogenic Inflammation/immunology
- Oncostatin M/genetics
- Oncostatin M/metabolism
- Oncostatin M Receptor beta Subunit/metabolism
- Peptide Fragments/immunology
- Receptors, Mitogen/genetics
- Receptors, Mitogen/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- M Elizabeth Deerhake
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Keiko Danzaki
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Makoto Inoue
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - Emre D Cardakli
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Toshiaki Nonaka
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nupur Aggarwal
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - William E Barclay
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
20
|
Duncan GJ, Simkins TJ, Emery B. Neuron-Oligodendrocyte Interactions in the Structure and Integrity of Axons. Front Cell Dev Biol 2021; 9:653101. [PMID: 33763430 PMCID: PMC7982542 DOI: 10.3389/fcell.2021.653101] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
The myelination of axons by oligodendrocytes is a highly complex cell-to-cell interaction. Oligodendrocytes and axons have a reciprocal signaling relationship in which oligodendrocytes receive cues from axons that direct their myelination, and oligodendrocytes subsequently shape axonal structure and conduction. Oligodendrocytes are necessary for the maturation of excitatory domains on the axon including nodes of Ranvier, help buffer potassium, and support neuronal energy metabolism. Disruption of the oligodendrocyte-axon unit in traumatic injuries, Alzheimer's disease and demyelinating diseases such as multiple sclerosis results in axonal dysfunction and can culminate in neurodegeneration. In this review, we discuss the mechanisms by which demyelination and loss of oligodendrocytes compromise axons. We highlight the intra-axonal cascades initiated by demyelination that can result in irreversible axonal damage. Both the restoration of oligodendrocyte myelination or neuroprotective therapies targeting these intra-axonal cascades are likely to have therapeutic potential in disorders in which oligodendrocyte support of axons is disrupted.
Collapse
Affiliation(s)
- Greg J. Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Tyrell J. Simkins
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, VA Portland Health Care System, Portland, OR, United States
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
21
|
Chen Y, Li Y, Guo L, Hong J, Zhao W, Hu X, Chang C, Liu W, Xiong K. Bibliometric Analysis of the Inflammasome and Pyroptosis in Brain. Front Pharmacol 2021; 11:626502. [PMID: 33551822 PMCID: PMC7854385 DOI: 10.3389/fphar.2020.626502] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Considering the pivotal role of inflammasome/pyroptosis in biological function, we visually analyzed the research hotspots of inflammasome/pyroptosis related to the brain in this work through the method of bibliometrics from the Web of Science (WOS) Core database over the past two decades. Methods: Documents were retrieved from WOS Core Collection on October 16, 2020. The search terms and strategies used for the WOS database are as follow: # 1, “pyroptosis”; # 2, “pyroptotic”; # 3, “inflammasome”; # 4, “pyroptosome”; # 5 “brain”; # 6, “# 1” OR “# 2” OR “# 3” OR “# 4”; # 7, “# 5” AND “# 6”. We selected articles and reviews published in English from 2000 to 2020. Visualization analysis and statistical analysis were performed by VOSviewer 1.6.15 and CiteSpace 5.7. R2. Results: 1,222 documents were selected for analysis. In the approximately 20 years since the pyroptosis was first presented, the publications regarding the inflammasome and pyroptosis in brain were presented since 2005. The number of annual publications increased gradually over a decade, which are involved in this work, and will continue to increase in 2020. The most prolific country was China with 523 documents but the United States was with 16,328 citations. The most influential author was Juan Pablo de Rivero Vaccari with 27 documents who worked at the University of Miami. The bibliometric analysis showed that inflammasome/pyroptosis involved a variety of brain cell types (microglia, astrocyte, neuron, etc.), physiological processes, ER stress, mitochondrial function, oxidative stress, and disease (traumatic brain injuries, stroke, Alzheimer’s disease, and Parkinson’s disease). Conclusion: The research of inflammasome/pyroptosis in brain will continue to be the hotspot. We recommend investigating the mechanism of mitochondrial molecules involved in the complex crosstalk of pyroptosis and regulated cell deaths (RCDs) in brain glial cells, which will facilitate the development of effective therapeutic strategies targeting inflammasome/pyroptosis and large-scale clinical trials. Thus, this study presents the trend and characteristic of inflammasome/pyroptosis in brain, which provided a helpful bibliometric analysis for researchers to further studies.
Collapse
Affiliation(s)
- Yuhua Chen
- Central Laboratory of Medicine School, Xi'an Peihua University, Xi'an, China.,Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China.,Department of Neurosurgery, First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yan Li
- Department of Histology and Embryology, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Limin Guo
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jun Hong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Wenjuan Zhao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Ximin Hu
- Clinical Medicine Eight-year Program, 02 Class, 17 Grade, Xiangya School of Medicine, Central South University, Changsha, China
| | - Cuicui Chang
- Central Laboratory of Medicine School, Xi'an Peihua University, Xi'an, China
| | - Wei Liu
- Department of Neurosurgery, First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China.,Hunan Key Laboratory of Ophthalmology, Changsha, China
| |
Collapse
|
22
|
Timmerman R, Burm SM, Bajramovic JJ. Tissue-specific features of microglial innate immune responses. Neurochem Int 2020; 142:104924. [PMID: 33248205 DOI: 10.1016/j.neuint.2020.104924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023]
Abstract
As tissue-resident macrophages of the brain, microglia are increasingly considered as cellular targets for therapeutical intervention. Innate immune responses in particular have been implicated in central nervous system (CNS) infections, neuro-oncology, neuroinflammatory and neurodegenerative diseases. We here review the impact of 'nature and nurture' on microglial innate immune responses and summarize documented tissue-specific adaptations. Overall, such adaptations are associated with regulatory processes rather than with overt differences in the expressed repertoire of activating receptors of different tissue-resident macrophages. Microglial responses are characterized by slower kinetics, by a more persistent nature and by a differential usage of downstream enzymes and accessory receptors. We further consider factors like aging, previous exposure to inflammatory stimuli, and differences in the microenvironment that can modulate innate immune responses. The long-life span of microglia in the metabolically active CNS renders them susceptible to the phenomenon of 'inflammaging', and major challenges lie in the unraveling of the factors that underlie age-related alterations in microglial behavior.
Collapse
Affiliation(s)
- R Timmerman
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - S M Burm
- Genmab, Utrecht, the Netherlands
| | - J J Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands.
| |
Collapse
|
23
|
HIFα Regulates Developmental Myelination Independent of Autocrine Wnt Signaling. J Neurosci 2020; 41:251-268. [PMID: 33208471 DOI: 10.1523/jneurosci.0731-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/15/2020] [Accepted: 11/11/2020] [Indexed: 01/17/2023] Open
Abstract
The developing CNS is exposed to physiological hypoxia, under which hypoxia-inducible factor α (HIFα) is stabilized and plays a crucial role in regulating neural development. The cellular and molecular mechanisms of HIFα in developmental myelination remain incompletely understood. A previous concept proposes that HIFα regulates CNS developmental myelination by activating the autocrine Wnt/β-catenin signaling in oligodendrocyte progenitor cells (OPCs). Here, by analyzing a battery of genetic mice of both sexes, we presented in vivo evidence supporting an alternative understanding of oligodendroglial HIFα-regulated developmental myelination. At the cellular level, we found that HIFα was required for developmental myelination by transiently controlling upstream OPC differentiation but not downstream oligodendrocyte maturation and that HIFα dysregulation in OPCs but not oligodendrocytes disturbed normal developmental myelination. We demonstrated that HIFα played a minor, if any, role in regulating canonical Wnt signaling in the oligodendroglial lineage or in the CNS. At the molecular level, blocking autocrine Wnt signaling did not affect HIFα-regulated OPC differentiation and myelination. We further identified HIFα-Sox9 regulatory axis as an underlying molecular mechanism in HIFα-regulated OPC differentiation. Our findings support a concept shift in our mechanistic understanding of HIFα-regulated CNS myelination from the previous Wnt-dependent view to a Wnt-independent one and unveil a previously unappreciated HIFα-Sox9 pathway in regulating OPC differentiation.SIGNIFICANCE STATEMENT Promoting disturbed developmental myelination is a promising option in treating diffuse white matter injury, previously called periventricular leukomalacia, a major form of brain injury affecting premature infants. In the developing CNS, hypoxia-inducible factor α (HIFα) is a key regulator that adapts neural cells to physiological and pathologic hypoxic cues. The role and mechanism of HIFα in oligodendroglial myelination, which is severely disturbed in preterm infants affected with diffuse white matter injury, is incompletely understood. Our findings presented here represent a concept shift in our mechanistic understanding of HIFα-regulated developmental myelination and suggest the potential of intervening with an oligodendroglial HIFα-mediated signaling pathway to mitigate disturbed myelination in premature white matter injury.
Collapse
|
24
|
Le CT, Khuat LT, Caryotakis SE, Wang M, Dunai C, Nguyen AV, Vick LV, Stoffel KM, Blazar BR, Monjazeb AM, Murphy WJ, Soulika AM. PD-1 Blockade Reverses Obesity-Mediated T Cell Priming Impairment. Front Immunol 2020; 11:590568. [PMID: 33193426 PMCID: PMC7658608 DOI: 10.3389/fimmu.2020.590568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/22/2020] [Indexed: 01/22/2023] Open
Abstract
Despite obesity reaching pandemic proportions, its impact on antigen-specific T cell responses is still unclear. We have recently demonstrated that obesity results in increased expression of PD-1 on T cells, and checkpoint blockade targeting PD-1/PD-L1 surprisingly resulted in greater clinical efficacy in cancer therapy. Adverse events associated with this therapy center around autoimmune reactions. In this study, we examined the impact of obesity on T cell priming and on autoimmune pathogenesis using the mouse model experimental autoimmune encephalomyelitis (EAE), which is mediated by autoreactive myelin-specific T cells generated after immunization. We observed that diet-induced obese (DIO) mice had a markedly delayed EAE onset and developed milder clinical symptoms compared to mice on control diet (CD). This delay was associated with impaired generation of myelin-specific T cell numbers and concurrently correlated with increased PD-L1 upregulation on antigen-presenting cells in secondary lymphoid organs. PD-1 blockade during the priming stage of EAE restored disease onset and severity and increased numbers of pathogenic CD4+ T cells in the central nervous system (CNS) of DIO mice to similar levels to those of CD mice. Administration of anti-PD-1 after onset of clinical symptoms did not increase EAE pathogenesis demonstrating that initial priming is the critical juncture affected by obesity. These findings demonstrate that obesity impairs antigen-specific T cell priming, but this can be reversed with PD-1 blockade. Our results further suggest that PD-1 blockade may increase the risk of autoimmune toxicities, particularly in obesity.
Collapse
Affiliation(s)
- Catherine T Le
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Lam T Khuat
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Sofia E Caryotakis
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Marilyn Wang
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Cordelia Dunai
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Alan V Nguyen
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Logan V Vick
- Department of Radiation-Oncology, School of Medicine, Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, United States
| | - Kevin M Stoffel
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Bruce R Blazar
- Masonic Cancer Center, and Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Arta M Monjazeb
- Department of Radiation-Oncology, School of Medicine, Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, United States
| | - William J Murphy
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States.,Department of Internal Medicine, Division of Hematology and Oncology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Athena M Soulika
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| |
Collapse
|
25
|
Reali C, Magliozzi R, Roncaroli F, Nicholas R, Howell OW, Reynolds R. B cell rich meningeal inflammation associates with increased spinal cord pathology in multiple sclerosis. Brain Pathol 2020; 30:779-793. [PMID: 32243032 PMCID: PMC8018043 DOI: 10.1111/bpa.12841] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022] Open
Abstract
Increased inflammation in the cerebral meninges is associated with extensive subpial cortical grey matter pathology in the forebrain and a more severe disease course in a substantial proportion of secondary progressive multiple sclerosis (SPMS) cases. It is not known whether this relationship extends to spinal cord pathology. We assessed the contribution of meningeal and parenchymal immune infiltrates to spinal cord pathology in SPMS cases characterized in the presence (F+) or absence (F-) of lymphoid-like structures in the forebrain meninges. Transverse cryosections of cervical, thoracic and lumbar cord of 22 SPMS and five control cases were analyzed for CD20+ B cells, CD4+ and CD8+ T cells, microglia/macrophages (IBA-1+), demyelination (myelin oligodendrocyte glycoprotein+) and axon density (neurofilament-H+). Lymphoid-like structures containing follicular dendritic cell networks and dividing B cells were seen in the spinal meninges of 3 out of 11 F+ SPMS cases. CD4+ and CD20+ cell counts were increased in F+ SPMS compared to F- SPMS and controls, whilst axon loss was greatest in motor and sensory tracts of the F+ SPMS cases (P < 0.01). The density of CD20+ B cells of the spinal leptomeninges correlated with CD4+ T cells and total B and T cells of the meninges; with the density of white matter perivascular CD20+ and CD4+ lymphocytes (P < 0.05); with white matter lesion area (P < 0.05); and the extent of axon loss (P < 0.05) in F+ SPMS cases only. We show that the presence of lymphoid-like structures in the forebrain is associated with a profound spinal cord pathology and local B cell rich meningeal inflammation associates with the extent of cord pathology. Our work supports a principal role for B cells in sustaining inflammation and tissue injury throughout the CNS in the progressive disease stage.
Collapse
Affiliation(s)
- Camilla Reali
- Department of Brain SciencesFaculty of MedicineImperial CollegeLondonUK
- Merck Healthcare KGaADarmstadtGermany
| | - Roberta Magliozzi
- Department of Brain SciencesFaculty of MedicineImperial CollegeLondonUK
- Department of Neuroscience, Biomedicine and MovementUniversity of VeronaVeronaItaly
| | - Federico Roncaroli
- Department of Brain SciencesFaculty of MedicineImperial CollegeLondonUK
- Division of Neuroscience and Experimental PsychologyFaculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Manchester Academic Health Science CentreManchesterUK
| | - Richard Nicholas
- Department of Brain SciencesFaculty of MedicineImperial CollegeLondonUK
| | - Owain W. Howell
- Department of Brain SciencesFaculty of MedicineImperial CollegeLondonUK
- Institute for Life SciencesSwansea University Medical SchoolSwanseaUK
| | - Richard Reynolds
- Department of Brain SciencesFaculty of MedicineImperial CollegeLondonUK
| |
Collapse
|
26
|
Yavarpour-Bali H, Ghasemi-Kasman M. The role of inflammasomes in multiple sclerosis. Mult Scler 2020; 27:1323-1331. [PMID: 32539629 DOI: 10.1177/1352458520932776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Multiple sclerosis (MS) is considered as an inflammatory autoimmune disease of the central nervous system (CNS), with a complex and heterogenic etiology. However, the involvement of inflammation in its pathophysiology is well documented and current therapies for MS are mainly immunosuppressive drugs. Although the available drugs reduce new lesions and relapses, their long-term outcome is not completely satisfactory. Inflammasomes are multimeric protein complexes that play a critical role in the inflammatory process. Several lines of evidence suggest an association between inflammasome activation and MS. In this paper, we have reviewed current studies that demonstrate the involvement of inflammasomes in MS development, in both animal model and MS patients. Furthermore, prior studies about the effect of inflammasome inhibitor drugs on development and progression of MS are discussed.
Collapse
Affiliation(s)
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran/Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
27
|
Gatta V, Mengod G, Reale M, Tata AM. Possible Correlation between Cholinergic System Alterations and Neuro/Inflammation in Multiple Sclerosis. Biomedicines 2020; 8:E153. [PMID: 32521719 PMCID: PMC7345633 DOI: 10.3390/biomedicines8060153] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system. Although the etiology of MS is still unknown, both genetic and environmental factors contribute to the pathogenesis of the disease. Acetylcholine participates in the modulation of central and peripheral inflammation. The cells of the immune system, as well as microglia, astrocytes and oligodendrocytes express cholinergic markers and receptors of muscarinic and nicotinic type. The role played by acetylcholine in MS has been recently investigated. In the present review, we summarize the evidence indicating the cholinergic dysfunction in serum and cerebrospinal fluid of relapsing-remitting (RR)-MS patients and in the brains of the MS animal model experimental autoimmune encephalomyelitis (EAE). The correlation between the increased activity of the cholinergic hydrolyzing enzymes acetylcholinesterase and butyrylcholinesterase, the reduced levels of acetylcholine and the increase of pro-inflammatory cytokines production were recently described in immune cells of MS patients. Moreover, the genetic polymorphisms for both hydrolyzing enzymes and the possible correlation with the altered levels of their enzymatic activity have been also reported. Finally, the changes in cholinergic markers expression in the central nervous system of EAE mice in peak and chronic phases suggest the involvement of the acetylcholine also in neuro-inflammatory processes.
Collapse
Affiliation(s)
- Valentina Gatta
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University, 66100 Chieti, Italy;
| | | | - Marcella Reale
- Department of Medical, Oral and Biotechnological Science, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy;
| | - Ada Maria Tata
- Department of Biology and Biotechnologies C. Darwin, “Sapienza” University of Rome, 00185 Rome, Italy
- Research Center of Neurobiology Daniel Bovet, “Sapienza” University of Rome, 00185 Rome, Italy
| |
Collapse
|
28
|
Khaw YM, Cunningham C, Tierney A, Sivaguru M, Inoue M. Neutrophil-selective deletion of Cxcr2 protects against CNS neurodegeneration in a mouse model of multiple sclerosis. J Neuroinflammation 2020; 17:49. [PMID: 32019585 PMCID: PMC7001284 DOI: 10.1186/s12974-020-1730-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/30/2020] [Indexed: 11/10/2022] Open
Abstract
Background Multiple sclerosis (MS) is a chronic debilitating immune-mediated disease of the central nervous system (CNS) driven by demyelination and gray matter neurodegeneration. We previously reported an experimental autoimmune encephalomyelitis (EAE) MS mouse model with elevated serum CXCL1 that developed severe and prolonged neuron damage. Our findings suggested that CXCR2 signaling may be important in neuronal damage, thus implicating neutrophils, which express CXCR2 in abundance, as a potential cell type involved. The goals of this study were to determine if CXCR2 signaling in neutrophils mediate neuronal damage and to identify potential mechanisms of damage. Methods EAE was induced in wild-type control and neutrophil-specific Cxcr2 knockout (Cxcr2 cKO) mice by repeated high-dose injections of heat-killed Mycobacterium tuberculosis and MOG35–55 peptide. Mice were examined daily for motor deficit. Serum CXCL1 level was determined at different time points throughout disease development. Neuronal morphology in Golgi-Cox stained lumbar spinal cord ventral horn was assessed using recently developed confocal reflection super-resolution technique. Immune cells from CNS and lymphoid organs were quantified by flow cytometry. CNS-derived neutrophils were co-cultured with neuronal crest cells and neuronal cell death was measured. Neutrophils isolated from lymphoid organs were examined for expression of reactive oxygen species (ROS) and ROS-related genes. Thioglycolate-activated neutrophils were isolated, treated with recombinant CXCL1, and measured for ROS production. Results Cxcr2 cKO mice had less severe disease symptoms at peak and late phase when compared to control mice with similar levels of CNS-infiltrating neutrophils and other immune cells despite high levels of circulating CXCL1. Additionally, Cxcr2 cKO mice had significantly reduced CNS neuronal damage in the ventral horn of the spinal cord. Neutrophils isolated from control EAE mice induced vast neuronal cell death in vitro when compared with neutrophils isolated from Cxcr2 cKO EAE mice. Neutrophils isolated from control EAE mice, but not Cxcr2 cKO mice, exhibited elevated ROS generation, in addition to heightened Ncf1 and Il1b transcription. Furthermore, recombinant CXCL1 was sufficient to significantly increase neutrophils ROS production. Conclusions CXCR2 signal in neutrophils is critical in triggering CNS neuronal damage via ROS generation, which leads to prolonged EAE disease. These findings emphasize that CXCR2 signaling in neutrophils may be a viable target for therapeutic intervention against CNS neuronal damage.
Collapse
Affiliation(s)
- Yee Ming Khaw
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Claire Cunningham
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,The School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Abigail Tierney
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,The School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Mayandi Sivaguru
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Makoto Inoue
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA. .,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
29
|
Zheng C, Chen J, Chu F, Zhu J, Jin T. Inflammatory Role of TLR-MyD88 Signaling in Multiple Sclerosis. Front Mol Neurosci 2020; 12:314. [PMID: 31998072 PMCID: PMC6965019 DOI: 10.3389/fnmol.2019.00314] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a neuro-autoimmune and neurodegenerative disorder leading to chronic inflammation, demyelination, axonal, and neuronal loss in the central nervous system (CNS). Despite intense research efforts, the pathogenesis of MS still remains unclear. Toll-like receptors (TLRs) are a family of type I transmembrane receptors that play a crucial role in the innate immune response. Myeloid differentiation factor 88 (MyD88) is the adaptor of major TLRs. It has been widely considered that the TLR-MyD88 signaling pathway plays an important role in the occurrence and development of autoimmune disease. Data have revealed that the TLR-MyD88 signaling may be involved in the pathogenesis of MS and experimental autoimmune encephalomyelitis (EAE), an animal model for MS, by regulating the antigen presentation of dendritic cells, the integrity of blood-brain barrier (BBB), and the activation of T cells and B cells. Here, we summarize the role of TLRs and MyD88 in MS and discuss the possible therapies that are based on these molecules.
Collapse
Affiliation(s)
- Chao Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fengna Chu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Rossi B, Constantin G, Zenaro E. The emerging role of neutrophils in neurodegeneration. Immunobiology 2020; 225:151865. [DOI: 10.1016/j.imbio.2019.10.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022]
|
31
|
Di Pinto G, Di Bari M, Martin-Alvarez R, Sperduti S, Serrano-Acedo S, Gatta V, Tata AM, Mengod G. Comparative study of the expression of cholinergic system components in the CNS of experimental autoimmune encephalomyelitis mice: Acute vs remitting phase. Eur J Neurosci 2019; 48:2165-2181. [PMID: 30144326 DOI: 10.1111/ejn.14125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 02/02/2023]
Abstract
Acetylcholine (ACh) is involved in the modulation of the inflammatory response. ACh levels are regulated by its synthesizing enzyme, choline acetyltransferase (ChAT), and by its hydrolyzing enzymes, mainly acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). A more comprehensive understanding of the cholinergic system in experimental autoimmune encephalomyelitis (EAE) disease progression could pave the path for the development of therapies to ameliorate multiple sclerosis (MS). In this work, we analyzed possible alterations of the CNS cholinergic system in the neuroinflammation process by using a MOG-induced EAE mice model. MOG- and vehicle-treated animals were studied at acute and remitting phases. We examined neuropathology and analyzed mRNA expression of ChAT, AChE and the α7 subunit of the nicotinic acetylcholine receptor (α7nAChR), as well as AChE and BuChE enzyme activities, in brain and spinal cord sections during disease progression. The mRNA expression and enzyme activities of these cholinergic markers were up- or down-regulated in many cholinergic areas and other brain areas of EAE mice in the acute and remitting phases of the disease. BuChE was present in a higher proportion of astroglia and microglia/macrophage cells in the EAE remitting group. The observed changes in cholinergic markers expression and cellular localization in the CNS during EAE disease progression suggests their potential involvement in the development of the neuroinflammatory process and may lay the ground to consider cholinergic system components as putative anti-inflammatory therapeutic targets for MS.
Collapse
Affiliation(s)
- Giovanni Di Pinto
- IIBB-CSIC, IDIBAPS, CIBERNED, Barcelona, Spain.,Department of Biology and Biotechnologies C. Darwin, "Sapienza" University of Rome, Rome, Italy
| | - Maria Di Bari
- IIBB-CSIC, IDIBAPS, CIBERNED, Barcelona, Spain.,Department of Biology and Biotechnologies C. Darwin, "Sapienza" University of Rome, Rome, Italy
| | | | - Samantha Sperduti
- IIBB-CSIC, IDIBAPS, CIBERNED, Barcelona, Spain.,Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Chieti, Italy.,Molecular Genetics Unit, CeSI-Met, Chieti, Italy
| | | | - Valentina Gatta
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, "G.d'Annunzio" University, Chieti, Italy.,Molecular Genetics Unit, CeSI-Met, Chieti, Italy
| | - Ada M Tata
- Department of Biology and Biotechnologies C. Darwin, "Sapienza" University of Rome, Rome, Italy.,Research Center of Neurobiology Daniel Bovet, "Sapienza" University of Rome, Rome, Italy
| | | |
Collapse
|
32
|
Clark K, Zhang S, Barthe S, Kumar P, Pivetti C, Kreutzberg N, Reed C, Wang Y, Paxton Z, Farmer D, Guo F, Wang A. Placental Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Myelin Regeneration in an Animal Model of Multiple Sclerosis. Cells 2019; 8:cells8121497. [PMID: 31771176 PMCID: PMC6952942 DOI: 10.3390/cells8121497] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) display potent immunomodulatory and regenerative capabilities through the secretion of bioactive factors, such as proteins, cytokines, chemokines as well as the release of extracellular vesicles (EVs). These functional properties of MSCs make them ideal candidates for the treatment of degenerative and inflammatory diseases, including multiple sclerosis (MS). MS is a heterogenous disease that is typically characterized by inflammation, demyelination, gliosis and axonal loss. In the current study, an induced experimental autoimmune encephalomyelitis (EAE) murine model of MS was utilized. At peak disease onset, animals were treated with saline, placenta-derived MSCs (PMSCs), as well as low and high doses of PMSC-EVs. Animals treated with PMSCs and high-dose PMSC-EVs displayed improved motor function outcomes as compared to animals treated with saline. Symptom improvement by PMSCs and PMSC-EVs led to reduced DNA damage in oligodendroglia populations and increased myelination within the spinal cord of treated mice. In vitro data demonstrate that PMSC-EVs promote myelin regeneration by inducing endogenous oligodendrocyte precursor cells to differentiate into mature myelinating oligodendrocytes. These findings support that PMSCs’ mechanism of action is mediated by the secretion of EVs. Therefore, PMSC-derived EVs are a feasible alternative to cellular based therapies for MS, as demonstrated in an animal model of the disease.
Collapse
Affiliation(s)
- Kaitlin Clark
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
| | - Sheng Zhang
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
| | - Sylvain Barthe
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
| | - Priyadarsini Kumar
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
| | - Christopher Pivetti
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
| | - Nicole Kreutzberg
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
| | - Camille Reed
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
| | - Yan Wang
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
| | - Zachary Paxton
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
| | - Diana Farmer
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
| | - Fuzheng Guo
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (K.C.); (S.B.); (P.K.); (C.P.); (N.K.); (C.R.); (Z.P.); (D.F.)
- Shriner’s Hospitals for Children, Northern California, Sacramento, CA 95817, USA; (S.Z.); (Y.W.); (F.G.)
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
- Correspondence: ; Tel.: +1-916-703-0422
| |
Collapse
|
33
|
Deerhake ME, Biswas DD, Barclay WE, Shinohara ML. Pattern Recognition Receptors in Multiple Sclerosis and Its Animal Models. Front Immunol 2019; 10:2644. [PMID: 31781124 PMCID: PMC6861384 DOI: 10.3389/fimmu.2019.02644] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022] Open
Abstract
Pattern recognition receptors (PRRs) coordinate the innate immune response and have a significant role in the development of multiple sclerosis (MS). Accumulating evidence has identified both pathogenic and protective functions of PRR signaling in MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Additionally, evidence for PRR signaling in non-immune cells and PRR responses to host-derived endogenous ligands has also revealed new pathways controlling the development of CNS autoimmunity. Many PRRs remain uncharacterized in MS and EAE, and understanding the distinct triggers and functions of PRR signaling in CNS autoimmunity requires further investigation. In this brief review, we discuss the diverse pathogenic and protective functions of PRRs in MS and EAE, and highlight major avenues for future research.
Collapse
Affiliation(s)
- M Elizabeth Deerhake
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Debolina D Biswas
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| | - William E Barclay
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
34
|
Aged hind-limb clasping experimental autoimmune encephalomyelitis models aspects of the neurodegenerative process seen in multiple sclerosis. Proc Natl Acad Sci U S A 2019; 116:22710-22720. [PMID: 31641069 DOI: 10.1073/pnas.1915141116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most common model of multiple sclerosis (MS). This model has been instrumental in understanding the events that lead to the initiation of central nervous system (CNS) autoimmunity. Though EAE has been an effective screening tool for identifying novel therapies for relapsing-remitting MS, it has proven to be less successful in identifying therapies for progressive forms of this disease. Though axon injury occurs in EAE, it is rapid and acute, making it difficult to intervene for the purpose of evaluating neuroprotective therapies. Here, we describe a variant of spontaneous EAE in the 2D2 T cell receptor transgenic mouse (2D2+ mouse) that presents with hind-limb clasping upon tail suspension and is associated with T cell-mediated inflammation in the posterior spinal cord and spinal nerve roots. Due to the mild nature of clinical signs in this model, we were able to maintain cohorts of mice into middle age. Over 9 mo, these mice exhibited a relapsing-remitting course of hind-limb clasping with the development of progressive motor deficits. Using a combined approach of ex vivo magnetic resonance (MR) imaging and histopathological analysis, we observed neurological progression to associate with spinal cord atrophy, synapse degradation, and neuron loss in the gray matter, as well as ongoing axon injury in the white matter of the spinal cord. These findings suggest that mild EAE coupled with natural aging may be a solution to better modeling the neurodegenerative processes seen in MS.
Collapse
|
35
|
Thomas AM, Xu J, Calabresi PA, van Zijl PCM, Bulte JWM. Monitoring diffuse injury during disease progression in experimental autoimmune encephalomyelitis with on resonance variable delay multiple pulse (onVDMP) CEST MRI. Neuroimage 2019; 204:116245. [PMID: 31605825 DOI: 10.1016/j.neuroimage.2019.116245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 09/16/2019] [Accepted: 10/03/2019] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder that targets myelin proteins and results in extensive damage in the central nervous system in the form of focal lesions as well as diffuse molecular changes. Lesions are currently detected using T1-weighted, T2-weighted, and gadolinium-enhanced magnetic resonance imaging (MRI); however, monitoring such lesions has been shown to be a poor predictor of disease progression. Chemical exchange saturation transfer (CEST) MRI is sensitive to many of the biomolecules in the central nervous system altered in MS that cannot be detected using conventional MRI. We monitored disease progression in an experimental autoimmune encephalomyelitis (EAE) model of MS using on resonance variable delay multiple pulse (onVDMP) CEST MRI. Alterations in onVDMP signal were observed in regions responsible for hindlimb function throughout the central nervous system. Histological analysis revealed glial activation in areas highlighted in onVDMP CEST MRI. onVDMP signal changes in the 3rd ventricle preceded paralysis onset that could not be observed with conventional MRI techniques. Hence, the onVDMP CEST MRI signal has potential as a novel imaging biomarker and predictor of disease progression in MS.
Collapse
Affiliation(s)
- Aline M Thomas
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiadi Xu
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Peter A Calabresi
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C M van Zijl
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jeff W M Bulte
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
36
|
Gharagozloo M, Mahmoud S, Simard C, Yamamoto K, Bobbala D, Ilangumaran S, Smith MD, Lamontagne A, Jarjoura S, Denault JB, Blais V, Gendron L, Vilariño-Güell C, Sadovnick AD, Ting JP, Calabresi PA, Amrani A, Gris D. NLRX1 inhibits the early stages of CNS inflammation and prevents the onset of spontaneous autoimmunity. PLoS Biol 2019; 17:e3000451. [PMID: 31525189 PMCID: PMC6762215 DOI: 10.1371/journal.pbio.3000451] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 09/26/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022] Open
Abstract
Nucleotide-binding, leucine-rich repeat containing X1 (NLRX1) is a mitochondria-located innate immune sensor that inhibits major pro-inflammatory pathways such as type I interferon and nuclear factor-κB signaling. We generated a novel, spontaneous, and rapidly progressing mouse model of multiple sclerosis (MS) by crossing myelin-specific T-cell receptor (TCR) transgenic mice with Nlrx1−/− mice. About half of the resulting progeny developed spontaneous experimental autoimmune encephalomyelitis (spEAE), which was associated with severe demyelination and inflammation in the central nervous system (CNS). Using lymphocyte-deficient mice and a series of adoptive transfer experiments, we demonstrate that genetic susceptibility to EAE lies within the innate immune compartment. We show that NLRX1 inhibits the subclinical stages of microglial activation and prevents the generation of neurotoxic astrocytes that induce neuronal and oligodendrocyte death in vitro. Moreover, we discovered several mutations within NLRX1 that run in MS-affected families. In summary, our findings highlight the importance of NLRX1 in controlling the early stages of CNS inflammation and preventing the onset of spontaneous autoimmunity. NLRX1 is a guardian protein that inhibits the inflammatory response of glial cells within the central nervous system and prevents the onset of a spontaneous multiple sclerosis–like disease in mice. This study uses a novel mouse model to provide mechanistic insights into the neurodegenerative origin of multiple sclerosis.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shaimaa Mahmoud
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Camille Simard
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Kenzo Yamamoto
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Diwakar Bobbala
- Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Subburaj Ilangumaran
- Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Albert Lamontagne
- Department of Neurology, Faculty of Medicine, MS Clinic, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Samir Jarjoura
- Department of Neurology, Faculty of Medicine, MS Clinic, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Bernard Denault
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Véronique Blais
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Louis Gendron
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - A. Dessa Sadovnick
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Jenny P. Ting
- Department of Microbiology and Immunology, Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Abdelaziz Amrani
- Department of Pediatrics, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Denis Gris
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- * E-mail:
| |
Collapse
|
37
|
Wesselingh R, Butzkueven H, Buzzard K, Tarlinton D, O'Brien TJ, Monif M. Innate Immunity in the Central Nervous System: A Missing Piece of the Autoimmune Encephalitis Puzzle? Front Immunol 2019; 10:2066. [PMID: 31552027 PMCID: PMC6746826 DOI: 10.3389/fimmu.2019.02066] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/15/2019] [Indexed: 12/14/2022] Open
Abstract
The autoimmune encephalitides are a group of autoimmune conditions targeting the central nervous system and causing severe clinical symptoms including drug-resistant seizures, cognitive dysfunction and psychiatric disturbance. Although these disorders appear to be antibody mediated, the role of innate immune responses needs further clarification. Infiltrating monocytes and microglial proliferation at the site of pathology could contribute to the pathogenesis of the disease with resultant blood brain barrier dysfunction, and subsequent activation of adaptive immune response. Both innate and adaptive immune cells can produce pro-inflammatory molecules which can perpetuate ongoing neuroinflammation and drive ongoing seizure activity. Ultimately neurodegenerative changes can ensue with resultant long-term neurological sequelae that can impact on ongoing patient morbidity and quality of life, providing a potential target for future translational research.
Collapse
Affiliation(s)
- Robb Wesselingh
- Department of Neurosciences, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neurosciences, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Katherine Buzzard
- Department of Neurology, Melbourne Health, Melbourne, VIC, Australia.,Department of Neurology, Eastern Health, Melbourne, VIC, Australia
| | - David Tarlinton
- Department of Immunology, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neurosciences, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, Australia.,Department of Neurology, Melbourne Health, Melbourne, VIC, Australia
| | - Mastura Monif
- Department of Neurosciences, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Neurology, Alfred Health, Melbourne, VIC, Australia.,Department of Neurology, Melbourne Health, Melbourne, VIC, Australia
| |
Collapse
|
38
|
Tanabe S, Saitoh S, Miyajima H, Itokazu T, Yamashita T. Microglia suppress the secondary progression of autoimmune encephalomyelitis. Glia 2019; 67:1694-1704. [PMID: 31106910 DOI: 10.1002/glia.23640] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022]
Abstract
Secondary progressive multiple sclerosis (SPMS) is an autoimmune disease of the central nervous system (CNS) characterized by progressive motor dysfunction, sensory deficits, and visual problems. The pathological mechanism of SPMS remains poorly understood. In this study, we investigated the role of microglia, immune cells in the CNS, in a secondary progressive form of experimental autoimmune encephalomyelitis (EAE), the mouse model of SPMS. We induced EAE in nonobese diabetic mice and treated the EAE mice with PLX3397, an antagonist of colony stimulating factor-1 receptor, during secondary progression in order to deplete microglia. The results showed that PLX3397 treatment significantly exacerbated secondary progression of EAE and increased mortality rates. Additionally, histological analysis showed that PLX3397 treatment significantly promoted inflammation, demyelination, and axonal degeneration. Moreover, the number of CD4+ T cells in the spinal cord of EAE mice was expanded due to PLX3397-mediated proliferation. These results suggest that microglia suppressed secondary progression of EAE by inhibiting the proliferation of CD4+ T cells in the CNS.
Collapse
Affiliation(s)
- Shogo Tanabe
- Department of Molecular Neuroscience, World Premier International, Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka, Japan.,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan
| | - Shohei Saitoh
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan
| | - Hisao Miyajima
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita-shi, Osaka, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, World Premier International, Immunology Frontier Research Center, Osaka University, Suita-shi, Osaka, Japan.,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita-shi, Osaka, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita-shi, Osaka, Japan
| |
Collapse
|
39
|
Kumar V. Toll-like receptors in the pathogenesis of neuroinflammation. J Neuroimmunol 2019; 332:16-30. [PMID: 30928868 DOI: 10.1016/j.jneuroim.2019.03.012] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Toll-like receptors (TLRs) are discovered as crucial pattern recognition receptors (PRRs) involved in the recognition of pathogen-associated molecular patterns (PAMPs). Later studies showed their involvement in the recognition of various damage/danger-associated molecular patterns (DAMPs) generated by host itself. Thus, TLRs are capable of recognizing wide-array of patterns/molecules derived from pathogens and host as well and initiating a proinflammatory immune response through the activation of NF-κB and other transcription factors causing synthesis of proinflammatory molecules. The process of neuroinflammation is seen under both sterile and infectious inflammatory diseases of the central nervous system (CNS) and may lead to the development of neurodegeneration. The present article is designed to highlight the importance of TLRs in the pathogenesis of neuroinflammation under diverse conditions. TLRs are expressed by various immune cells present in CNS along with neurons. However out of thirteen TLRs described in mammals, some are present and active in these cells, while some are absent and are described in detail in main text. The role of various immune cells present in the brain and their role in the pathogenesis of neuroinflammation depending on the type of TLR expressed is described. Thereafter the role of TLRs in bacterial meningitis, viral encephalitis, stroke, Alzheimer's disease (AD), Parkinson's disease (PD), and autoimmune disease including multiple sclerosis (MS) is described. The article is designed for both neuroscientists needing information regarding TLRs in neuroinflammation and TLR biologists or immunologists interested in neuroinflammation.
Collapse
Affiliation(s)
- V Kumar
- Children Health Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
40
|
Adkins Y, Soulika AM, Mackey B, Kelley DS. Docosahexaenoic acid (22:6n-3) Ameliorated the Onset and Severity of Experimental Autoimmune Encephalomyelitis in Mice. Lipids 2019; 54:13-23. [PMID: 30762234 DOI: 10.1002/lipd.12130] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/11/2018] [Accepted: 01/03/2019] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis (MS) is a neurologic autoimmune disease, which is the leading cause of nontraumatic neurologic disability in young adults in United States and Europe. n-3 polyunsaturated fatty acids (PUFA) are reported to mitigate severity of this disease. Recent studies suggest that phospholipid (PL) form of dietary n-3 PUFA may lead to their higher tissue accretion than triacylglycerol (TAG) form. We compared efficacy of PL-docosahexaenoic acid (22:6n-3) (DHA) and TAG-DHA on onset and severity of experimental autoimmune encephalomyelitis (EAE) in a mouse model of MS. Female mice were fed low alpha-linolenic acid (18:3n-3) (ALA) diet (control) for 2 weeks and then fed either control, 0.3%, or 1.0% DHA (PL or TAG) for 4 weeks pre-EAE induction and 4 weeks post-EAE induction. The brain and spinal cord n-6:n-3 ratio was significantly lower in all mice fed DHA compared to control. EAE onset was delayed in mice fed both DHA forms and concentrations, except for 1% TAG-DHA. The inverse association between the EAE score and the brain DHA concentration was nonsignificant at the end of the study (p = 0.08). Daily EAE scores of mice fed different DHA diets did not differ from control, however, the score of all DHA groups combined during days 9-16 was lower (p = 0.028) compared to the control. During days 17-22, the EAE score trended lower in 0.3% TAG-DHA and during days 23-28, the EAE score trended lower in both PL-DHA groups than those in all other groups. These findings suggest that TAG-DHA may be more effective than PL-DHA in the early phases of EAE, and in the final outcome, PL-DHA may be more effective than TAG-DHA.
Collapse
Affiliation(s)
- Yuriko Adkins
- USDA, ARS, Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA
| | - Athena M Soulika
- Department of Dermatology, University of California Davis Medical Center and Shriners Hospital for Children, Northern California, 2425 Stockton Boulevard, Sacramento, CA 95816, USA
| | - Bruce Mackey
- USDA, ARS, Western Regional Research Center, 800 Buchanan St Albany, CA 94710, USA
| | - Darshan S Kelley
- USDA, ARS, Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA.,Department of Nutrition, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
41
|
Knier B, Hiltensperger M, Sie C, Aly L, Lepennetier G, Engleitner T, Garg G, Muschaweckh A, Mitsdörffer M, Koedel U, Höchst B, Knolle P, Gunzer M, Hemmer B, Rad R, Merkler D, Korn T. Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity. Nat Immunol 2018; 19:1341-1351. [PMID: 30374128 PMCID: PMC6241855 DOI: 10.1038/s41590-018-0237-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 09/13/2018] [Indexed: 01/25/2023]
Abstract
Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) have been characterized in the context of malignancies. Here we show that PMN-MDSCs can restrain B cell accumulation during central nervous system (CNS) autoimmunity. Ly6G+ cells were recruited to the CNS during experimental autoimmune encephalomyelitis (EAE), interacted with B cells that produced the cytokines GM-CSF and interleukin-6 (IL-6), and acquired properties of PMN-MDSCs in the CNS in a manner dependent on the signal transducer STAT3. Depletion of Ly6G+ cells or dysfunction of Ly6G+ cells through conditional ablation of STAT3 led to the selective accumulation of GM-CSF-producing B cells in the CNS compartment, which in turn promoted an activated microglial phenotype and lack of recovery from EAE. The frequency of CD138+ B cells in the cerebrospinal fluid (CSF) of human subjects with multiple sclerosis was negatively correlated with the frequency of PMN-MDSCs in the CSF. Thus PMN-MDSCs might selectively control the accumulation and cytokine secretion of B cells in the inflamed CNS.
Collapse
Affiliation(s)
- Benjamin Knier
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael Hiltensperger
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christopher Sie
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lilian Aly
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gildas Lepennetier
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Engleitner
- Institute of Molecular Oncology and Functional Genomics, TranslaTUM Cancer Center, Technical University of Munich, Munich, Germany.,Department of Medicine II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Garima Garg
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Andreas Muschaweckh
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Meike Mitsdörffer
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Uwe Koedel
- Department of Neurology, Klinikum Grosshadern, Ludwig Maximilians University Munich, Munich, Germany
| | - Bastian Höchst
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Percy Knolle
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Munich, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, TranslaTUM Cancer Center, Technical University of Munich, Munich, Germany.,Department of Medicine II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, University of Geneva, Geneva, Switzerland
| | - Thomas Korn
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany. .,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
42
|
Role of Inflammasomes in Neuroimmune and Neurodegenerative Diseases: A Systematic Review. Mediators Inflamm 2018; 2018:1549549. [PMID: 29849483 PMCID: PMC5932495 DOI: 10.1155/2018/1549549] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/26/2017] [Accepted: 01/01/2018] [Indexed: 12/31/2022] Open
Abstract
Inflammasomes are multiprotein complexes that can sense pathogen-associated molecular patterns and damage-associated molecular signals. They are involved in the initiation and development of inflammation via activation of IL-1β and IL-18. Many recent studies suggest a strong correlation between inflammasomes and neurological diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), and Parkinson's disease (PD). Several components of inflammasomes, such as nucleotide-binding oligomerization domain- (NOD-) like receptor, absent in melanoma 2- (AIM2-) like receptors (ALRs), apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and caspase-1, as well as the upstream factors and downstream effectors, are associated with the initiation and development of MS and its animal model, experimental autoimmune encephalomyelitis. Additionally, inflammasomes affect the efficacy of interferon-β therapy in patients with MS. Finally, the strong association of inflammasomes with AD and PD needs to be further studied. In this review of latest literatures, we comprehensively tease out diverse roles of different kinds of inflammasomes in neuroimmune and neurodegenerative diseases, especially in the perspective of double roles involved in pathogenesis, and identify future research priorities.
Collapse
|
43
|
Pierson ER, Wagner CA, Goverman JM. The contribution of neutrophils to CNS autoimmunity. Clin Immunol 2018; 189:23-28. [PMID: 27377536 PMCID: PMC5203971 DOI: 10.1016/j.clim.2016.06.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is believed to be initiated when myelin-specific T cells infiltrate the central nervous system (CNS), triggering subsequent recruitment of inflammatory leukocytes to the CNS. The contribution of neutrophils to CNS autoimmune disease has been underappreciated, but several studies in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, indicate that neutrophils have an important role in inflammation. Neutrophils are hypothesized to contribute to the pathogenesis of EAE by producing cytokines and promoting breakdown of the blood brain barrier. Neutrophils may also influence the manifestation of EAE by facilitating parenchymal brain inflammation. This review summarizes evidence supporting a functional role for neutrophils in EAE and MS, highlighting the differential regulation of neutrophil recruitment in the brain and spinal cord.
Collapse
Affiliation(s)
- Emily R Pierson
- Department of Immunology, University of Washington, Box 358059, 750 Republican St., Seattle, WA 98109-8509, USA
| | - Catriona A Wagner
- Department of Immunology, University of Washington, Box 358059, 750 Republican St., Seattle, WA 98109-8509, USA
| | - Joan M Goverman
- Department of Immunology, University of Washington, Box 358059, 750 Republican St., Seattle, WA 98109-8509, USA.
| |
Collapse
|
44
|
Silva BA, Leal MC, Farías MI, Avalos JC, Besada CH, Pitossi FJ, Ferrari CC. A new focal model resembling features of cortical pathology of the progressive forms of multiple sclerosis: Influence of innate immunity. Brain Behav Immun 2018; 69:515-531. [PMID: 29378262 DOI: 10.1016/j.bbi.2018.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating disease of unknown aetiology that causes neurological disabilities in young adults. MS displays different clinical patterns, including recurrent episodes with remission periods ("relapsing-remitting MS" (RRMS)), which can progress over several years to a secondary progressive form (SPMS). However, 10% of patients display persistent progression at the onset of disease ("primary progressive MS" (PPMS)). Currently, no specific therapeutic agents are available for the progressive forms, mainly because the underlying pathogenic mechanisms are not clear and because no animal models have been specifically developed for these forms. The development of MS animal models is required to clarify the pathological mechanisms and to test novel therapeutic agents. In the present work, we overexpressed interleukin 1 beta (IL-1β) in the cortex to develop an animal model reflecting the main pathological hallmarks of MS. The treated animals presented with neuroinflammation, demyelination, glial activation, and neurodegeneration along with cognitive symptoms and MRI images consistent with MS pathology. We also demonstrated the presence of meningeal inflammation close to cortical lesions, with characteristics similar to those described in MS patients. Systemic pro-inflammatory stimulation caused a flare-up of the cortical lesions and behavioural symptoms, including impairment of working memory and the appearance of anxiety-like symptoms. Our work demonstrated induced cortical lesions, reflecting the main histopathological hallmarks and cognitive impairments characterizing the cortical pathology described in MS patients with progressive forms of the disease.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - María Celeste Leal
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - María Isabel Farías
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | | | | | - Fernando Juan Pitossi
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina.
| |
Collapse
|
45
|
Cell-specific and region-specific transcriptomics in the multiple sclerosis model: Focus on astrocytes. Proc Natl Acad Sci U S A 2018; 115:E302-E309. [PMID: 29279367 PMCID: PMC5777065 DOI: 10.1073/pnas.1716032115] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Changes in gene expression that occur across the central nervous system (CNS) during neurological diseases do not address the heterogeneity of cell types from one CNS region to another and are complicated by alterations in cellular composition during disease. Multiple sclerosis (MS) is multifocal by definition. Here, a cell-specific and region-specific transcriptomics approach was used to determine gene expression changes in astrocytes in the most widely used MS model, experimental autoimmune encephalomyelitis (EAE). Astrocyte-specific RNAs from various neuroanatomic regions were attained using RiboTag technology. Sequencing and bioinformatics analyses showed that EAE-induced gene expression changes differed between neuroanatomic regions when comparing astrocytes from spinal cord, cerebellum, cerebral cortex, and hippocampus. The top gene pathways that were changed in astrocytes from spinal cord during chronic EAE involved decreases in expression of cholesterol synthesis genes while immune pathway gene expression in astrocytes was increased. Optic nerve from EAE and optic chiasm from MS also showed decreased cholesterol synthesis gene expression. The potential role of cholesterol synthesized by astrocytes during EAE and MS is discussed. Together, this provides proof-of-concept that a cell-specific and region-specific gene expression approach can provide potential treatment targets in distinct neuroanatomic regions during multifocal neurological diseases.
Collapse
|
46
|
Martín-Álvarez R, Paúl-Fernández N, Palomo V, Gil C, Martínez A, Mengod G. A preliminary investigation of phoshodiesterase 7 inhibitor VP3.15 as therapeutic agent for the treatment of experimental autoimmune encephalomyelitis mice. J Chem Neuroanat 2017; 80:27-36. [DOI: 10.1016/j.jchemneu.2016.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/10/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
|
47
|
Casserly CS, Nantes JC, Whittaker Hawkins RF, Vallières L. Neutrophil perversion in demyelinating autoimmune diseases: Mechanisms to medicine. Autoimmun Rev 2017; 16:294-307. [DOI: 10.1016/j.autrev.2017.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
|
48
|
Lassmann H, Bradl M. Multiple sclerosis: experimental models and reality. Acta Neuropathol 2017; 133:223-244. [PMID: 27766432 PMCID: PMC5250666 DOI: 10.1007/s00401-016-1631-4] [Citation(s) in RCA: 351] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023]
Abstract
One of the most frequent statements, provided in different variations in the introduction of experimental studies on multiple sclerosis (MS), is that "Multiple sclerosis is a demyelinating autoimmune disease and experimental autoimmune encephalomyelitis (EAE) is a suitable model to study its pathogenesis". However, so far, no single experimental model covers the entire spectrum of the clinical, pathological, or immunological features of the disease. Many different models are available, which proved to be highly useful for studying different aspects of inflammation, demyelination, remyelination, and neurodegeneration in the central nervous system. However, the relevance of results from such models for MS pathogenesis has to be critically validated. Current EAE models are mainly based on inflammation, induced by auto-reactive CD4+ T-cells, and these models reflect important aspects of MS. However, pathological data and results from clinical trials in MS indicate that CD8+ T-cells and B-lymphocytes may play an important role in propagating inflammation and tissue damage in established MS. Viral models may reflect key features of MS-like inflammatory demyelination, but are difficult to use due to their very complex pathogenesis, involving direct virus-induced and immune-mediated mechanisms. Furthermore, evidence for a role of viruses in MS pathogenesis is indirect and limited, and an MS-specific virus infection has not been identified so far. Toxic models are highly useful to unravel mechanisms of de- and remyelination, but do not reflect other important aspects of MS pathology and pathogenesis. For all these reasons, it is important to select the right experimental model to answer specific questions in MS research.
Collapse
Affiliation(s)
- Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| | - Monika Bradl
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| |
Collapse
|
49
|
Pleasure D. Good Things Come in Threes: Genetically Engineered Neural Stem Cells Mitigate Chronic CNS Autoimmunity. Mol Ther 2016; 24:1338-9. [PMID: 27578282 DOI: 10.1038/mt.2016.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- David Pleasure
- Department of Neurology, University of California-Davis, Sacramento, California, USA
| |
Collapse
|
50
|
Thalamus Degeneration and Inflammation in Two Distinct Multiple Sclerosis Animal Models. J Mol Neurosci 2016; 60:102-14. [PMID: 27491786 DOI: 10.1007/s12031-016-0790-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/21/2016] [Indexed: 12/31/2022]
Abstract
There is a broad consensus that multiple sclerosis (MS) represents more than an inflammatory disease: it harbors several characteristic aspects of a classical neurodegenerative disorder, i.e., damage to axons, synapses, and nerve cell bodies. While several accepted paraclinical methods exist to monitor the inflammatory-driven aspects of the disease, techniques to monitor progression of early and late neurodegeneration are still in their infancy and have not been convincingly validated. It was speculated that the thalamus with its multiple reciprocal connections is sensitive to inflammatory processes occurring in different brain regions, thus acting as a "barometer" for diffuse brain parenchymal damage in MS. To what extent the thalamus is affected in commonly applied MS animal models is, however, not known. In this article we describe direct and indirect damage to the thalamus in two distinct MS animal models. In the cuprizone model, we observed primary oligodendrocyte stress which is followed by demyelination, microglia/astrocyte activation, and acute axonal damage. These degenerative cuprizone-induced lesions were found to be more severe in the lateral compared to the medial part of the thalamus. In MOG35-55-induced EAE, in contrast, most parts of the forebrain, including the thalamus were not directly involved in the autoimmune attack. However, important thalamic afferent fiber tracts, such as the spinothalamic tract were inflamed and demyelinated on the spinal cord level. Quantitative immunohistochemistry revealed that this spinal cord inflammatory-demyelination is associated with neuronal loss within the target region of the spinothalamic tract, namely the sensory ventral posterolateral nucleus of the thalamus. This study highlights the possibility of trans-neuronal degeneration as one mechanism of secondary neuronal damage in MS. Further studies are now warranted to investigate involved cell types and cellular mechanisms.
Collapse
|