1
|
Tong L, Ozes B, Moss K, Myers M, Attia Z, Vetter TA, Trapp BD, Sahenk Z. AAV1.NT3 gene therapy mitigates the severity of autoimmune encephalomyelitis in the mouse model for multiple sclerosis. Gene Ther 2025:10.1038/s41434-025-00518-9. [PMID: 39972161 DOI: 10.1038/s41434-025-00518-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 02/21/2025]
Abstract
Multiple sclerosis (MS) is an immune-mediated chronic inflammatory and neurodegenerative disease of the central nervous system (CNS) affecting more than 2.5 million patients worldwide. Chronic demyelination in the CNS has an important role in perpetuating axonal loss and increases difficulty in promoting remyelination. Therefore, regenerative, and neuroprotective strategies are essential to overcome this impediment to rescue axonal integrity and function. Neurotrophin 3 (NT-3) has immunomodulatory and anti-inflammatory properties, in addition to its well-recognized function in nervous system development, myelination, neuroprotection, and regeneration. For this study, scAAV1.tMCK.NT-3 was delivered to the gastrocnemius muscle of experimental autoimmune encephalomyelitis (EAE) mice, the chronic relapsing mouse model of MS, at 3 weeks post EAE induction. Measurable NT-3 levels were found in serum at 7-weeks post gene delivery. The treated cohort showed improved clinical scores and performed significantly better in rotarod, and grip strength tests compared to their untreated counterparts. Histopathologic studies showed improved remyelination and axon protection. These data correlated with reduced expression of the pro-inflammatory cytokines in brain and spinal cord, and increased percentage of regulatory T cells in the spleens and lymph nodes. Collectively, these findings demonstrate the translational potential of AAV-delivered NT-3 for chronic progressive MS.
Collapse
Affiliation(s)
- Lingying Tong
- Department of Pediatrics, Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Burcak Ozes
- Department of Pediatrics, Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kyle Moss
- Department of Pediatrics, Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Morgan Myers
- Department of Pediatrics, Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Zayed Attia
- Department of Pediatrics, Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Tatyana A Vetter
- Department of Pediatrics, Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics and Neurology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zarife Sahenk
- Department of Pediatrics, Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatrics and Neurology, Nationwide Children's Hospital and The Ohio State University, Columbus, OH, USA.
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
2
|
Lehikoinen J, Strandin T, Parantainen J, Nurmi K, Eklund KK, Rivera FJ, Vaheri A, Tienari PJ. Fibrinolysis associated proteins and lipopolysaccharide bioactivity in plasma and cerebrospinal fluid in multiple sclerosis. J Neuroimmunol 2024; 395:578432. [PMID: 39151321 DOI: 10.1016/j.jneuroim.2024.578432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/24/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
The coagulation cascade and fibrinolysis have links with neuroinflammation and increased activation of the coagulation system has been reported in MS patients. We quantified levels of D-dimer, tissue plasminogen activator (tPA), plasminogen activator inhibitor-1 (PAI-1) and the bioactivity of bacterial lipopolysaccharide (LPS) in cerebrospinal fluid (CSF) and plasma from newly diagnosed untreated MS patients and controls. These molecules showed multiple correlations with each other as well as with age, HLA-DRB1*15:01, body-mass-index and CSF IgG. Our results confirm previous findings of increased plasma PAI-1 and LPS in MS patients compared to controls indicating changes in platelet function and gut permeability in MS.
Collapse
Affiliation(s)
- Joonas Lehikoinen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Neurology, Neurocenter, Helsinki University Hospital, Helsinki, Finland.
| | - Tomas Strandin
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Jukka Parantainen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Katariina Nurmi
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Kari K Eklund
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Rheumatology, Helsinki University Hospital, Helsinki, Finland
| | - Francisco J Rivera
- Translational Regenerative Neurobiology Group (TReN), Molecular and Integrative Biosciences Research Programme (MIBS), Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Pentti J Tienari
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; Department of Neurology, Neurocenter, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
3
|
Comi G, Dalla Costa G, Stankoff B, Hartung HP, Soelberg Sørensen P, Vermersch P, Leocani L. Assessing disease progression and treatment response in progressive multiple sclerosis. Nat Rev Neurol 2024; 20:573-586. [PMID: 39251843 DOI: 10.1038/s41582-024-01006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Progressive multiple sclerosis poses a considerable challenge in the evaluation of disease progression and treatment response owing to its multifaceted pathophysiology. Traditional clinical measures such as the Expanded Disability Status Scale are limited in capturing the full scope of disease and treatment effects. Advanced imaging techniques, including MRI and PET scans, have emerged as valuable tools for the assessment of neurodegenerative processes, including the respective role of adaptive and innate immunity, detailed insights into brain and spinal cord atrophy, lesion dynamics and grey matter damage. The potential of cerebrospinal fluid and blood biomarkers is increasingly recognized, with neurofilament light chain levels being a notable indicator of neuro-axonal damage. Moreover, patient-reported outcomes are crucial for reflecting the subjective experience of disease progression and treatment efficacy, covering aspects such as fatigue, cognitive function and overall quality of life. The future incorporation of digital technologies and wearable devices in research and clinical practice promises to enhance our understanding of functional impairments and disease progression. This Review offers a comprehensive examination of these diverse evaluation tools, highlighting their combined use in accurately assessing disease progression and treatment efficacy in progressive multiple sclerosis, thereby guiding more effective therapeutic strategies.
Collapse
Affiliation(s)
- Giancarlo Comi
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, Milan, Italy.
| | | | - Bruno Stankoff
- Sorbonne Université, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Centre National de la Recherche Scientifique, Inserm, Paris, France
| | - Hans-Peter Hartung
- Brain and Mind Center, University of Sydney, Sydney, Australia
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Per Soelberg Sørensen
- Department of Neurology, Danish Multiple Sclerosis Center, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Patrick Vermersch
- University of Lille, Inserm U1172, Lille Neuroscience & Cognition, Centre Hospitalier Universitaire de Lille, Fédération Hospitalo-Universitaire Precision Medicine in Psychiatry, Lille, France
| | - Letizia Leocani
- Vita-Salute San Raffaele University, Milan, Italy
- Multiple Sclerosis Center, Casa di Cura Igea, Milan, Italy
| |
Collapse
|
4
|
Petersen-Cherubini CL, Liu Y, Deffenbaugh JL, Murphy SP, Xin M, Rau CN, Yang Y, Lovett-Racke AE. Dysregulated autotaxin expression by T cells in multiple sclerosis. J Neuroimmunol 2024; 387:578282. [PMID: 38183947 PMCID: PMC10923181 DOI: 10.1016/j.jneuroim.2023.578282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Multiple sclerosis (MS) is a demyelinating disease characterized by infiltration of autoreactive T cells into the central nervous system (CNS). In order to understand how activated, autoreactive T cells are able to cross the blood brain barrier, the unique molecular characteristics of pathogenic T cells need to be more thoroughly examined. In previous work, our laboratory found autotaxin (ATX) to be upregulated by activated autoreactive T cells in the mouse model of MS. ATX is a secreted glycoprotein that promotes T cell chemokinesis and transmigration through catalysis of lysophoshphatidic acid (LPA). ATX is elevated in the serum of MS patients during active disease phases, and we previously found that inhibiting ATX decreases severity of neurological deficits in the mouse model. In this study, ATX expression was found to be lower in MS patient immune cells during rest, but significantly increased during early activation in a manner not seen in healthy controls. The ribosomal binding protein HuR, which stabilizes ATX mRNA, was also increased in MS patients in a similar pattern to that of ATX, suggesting it may be helping regulate ATX levels after activation. The proinflammatory cytokine interleukin-23 (IL-23) was shown to induce prolonged ATX expression in MS patient Th1 and Th17 cells. Finally, through ChIP, re-ChIP analysis, we show that IL-23 may be signaling through pSTAT3/pSTAT4 heterodimers to induce expression of ATX. Taken together, these findings elucidate cell types that may be contributing to elevated serum ATX levels in MS patients and identify potential drivers of sustained expression in encephalitogenic T cells.
Collapse
Affiliation(s)
- Cora L Petersen-Cherubini
- The Ohio State University, Neuroscience Graduate Program, 460 West 12th Avenue, Biomedical Research Tower 6894, Columbus, OH 43210, USA; The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA
| | - Yue Liu
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Joshua L Deffenbaugh
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Shawn P Murphy
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Matthew Xin
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA
| | - Christina N Rau
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| | - Yuhong Yang
- The Ohio State University, Wexner Medical Center, Department of Neurology, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA
| | - Amy E Lovett-Racke
- The Ohio State University, Wexner Medical Center, Department of Microbial Infection and Immunity, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA; The Ohio State University, Wexner Medical Center, Department of Neuroscience, 460 West 12th Avenue, Biomedical Research Tower 684, Columbus, OH 43210, USA.
| |
Collapse
|
5
|
Zhang Y, Atkinson J, Burd CE, Graves J, Segal BM. Biological aging in multiple sclerosis. Mult Scler 2023; 29:1701-1708. [PMID: 37877740 PMCID: PMC10843499 DOI: 10.1177/13524585231204122] [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] [Indexed: 10/26/2023]
Abstract
Multiple sclerosis (MS) is most likely to adopt a progressive clinical course during middle age or beyond, and the number of older adults with MS is steadily increasing. Developing new strategies to manage progressive forms of MS, which do not respond to currently available disease-modifying therapies (DMTs), will require a deeper understanding of the mechanisms by which biological aging interacts with pathogenic pathways to propel disability accumulation. In experimental autoimmune encephalomyelitis (EAE), a widely used preclinical mouse model of MS, middle-aged animals experience a more severe and protracted clinical course than their younger counterparts. This exacerbated disease course is accompanied by persistent neuroinflammation. Clinical studies of age-related biomarkers, such as telomere length, senescence markers, and DNA methylation, suggest that biological aging is accelerated in people with MS compared with age- and sex-matched healthy controls. Furthermore, distinguishing biological age from chronological may afford more precision in determining aging effects in MS. Here we review the current literature on aging biology and its impact on MS pathogenesis. Future research on this topic may lead to the development of novel biomarkers and senotherapy agents that slow neurological decline in people with progressive MS by targeting relevant aging-related pathways.
Collapse
Affiliation(s)
- Yinan Zhang
- Department of Neurology, The Ohio State University Wexner
Medical Center, Columbus, OH
| | - Jeffrey Atkinson
- Department of Neurology, The Ohio State University Wexner
Medical Center, Columbus, OH
| | - Christin E. Burd
- Departments of Molecular Genetics, Cancer Biology and
Genetics, The Ohio State University, Columbus, OH
| | - Jennifer Graves
- Department of Neurosciences, University of California San
Diego, San Diego, CA
| | - Benjamin M. Segal
- Department of Neurology, The Ohio State University Wexner
Medical Center, Columbus, OH
| |
Collapse
|
6
|
Adabanya U, Awosika A, Khan A, Oluka E, Adeniyi M. Pediatric multiple sclerosis: an integrated outlook at the interplay between genetics, environment and brain-gut dysbiosis. AIMS Neurosci 2023; 10:232-251. [PMID: 37841344 PMCID: PMC10567585 DOI: 10.3934/neuroscience.2023018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 10/17/2023] Open
Abstract
Multiple sclerosis (MS) is a debilitating autoimmune condition caused by demyelination, neurodegeneration and persistent inflammation of the central nervous system. Pediatric multiple sclerosis (PMS) is a relatively rare form of the disease that affects a significant number of individuals with MS. Environmental exposures, such as viral infections and smoking, can interact with MS-associated human leukocyte antigens (HLA) risk alleles and influence the immune response. Upregulation of immune response results in the disruption of immune balance leading to cascade of inflammatory events. It has also been established that gut microbiome dysbiosis poses a higher risk for pro-inflammation, and it is essentially argued to be the greatest environmental risk factor for MS. Dysbiosis can cause an unusual response from the adaptive immune system and significantly contribute to the development of disease in the host by activating pro-inflammatory pathways that cause immune-mediated disorders such as PMS, rendering the body more vulnerable to foreign attacks due to a weakened immune response. All these dynamic interactions between biological, environmental and genetic factors based on epigenetic study has further revealed that upregulation or downregulation of some genes/enzyme in the central nervous system white matter of MS patients produces a less stable form of myelin basic protein and ultimately leads to the loss of immune tolerance. The diagnostic criteria and treatment options for PMS are constantly evolving, making it crucial to have a better understanding of the disease burden on a global and regional scale. The findings from this review will aid in deepening the understanding of the interplay between genetic and environmental risk factors, as well as the role of the gut microbiome in the development of pediatric multiple sclerosis. As a result, healthcare professionals will be kept abreast of the early diagnostic criteria, accurately delineating other conditions that can mimic pediatric MS and to provide comprehensive care to individuals with PMS based on the knowledge gained from this research.
Collapse
Affiliation(s)
- Uzochukwu Adabanya
- Anatomical Sciences, Edward Via College of Osteopathic Medicine, Monroe, USA
| | - Ayoola Awosika
- College of Medicine, University of Illinois, Chicago, USA
| | - Anosh Khan
- Emergency Medicine, Trinity health Livonia Hospital, Livonia USA
| | - Ejike Oluka
- Department of pathophysiology, St. George's University School of Medicine, Grenada
| | - Mayowa Adeniyi
- Department of Physiology, Federal University of Health Sciences Otukpo, Benue State, Nigeria
| |
Collapse
|
7
|
Palacio PL, Pleet ML, Reátegui E, Magaña SM. Emerging role of extracellular vesicles in multiple sclerosis: From cellular surrogates to pathogenic mediators and beyond. J Neuroimmunol 2023; 377:578064. [PMID: 36934525 PMCID: PMC10124134 DOI: 10.1016/j.jneuroim.2023.578064] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/06/2023] [Accepted: 03/05/2023] [Indexed: 03/16/2023]
Abstract
Multiple Sclerosis (MS) is a chronic, inflammatory demyelinating disease of the central nervous system (CNS) driven by a complex interplay of genetic and environmental factors. While the therapeutic arsenal has expanded significantly for management of relapsing forms of MS, treatment of individuals with progressive MS is suboptimal. This treatment inequality is in part due to an incomplete understanding of pathomechanisms at different stages of the disease-underscoring the critical need for new biomarkers. Extracellular vesicles (EVs) and their bioactive cargo have emerged as endogenous nanoparticles with great theranostic potential-as diagnostic and prognostic biomarkers and ultimately as therapeutic candidates for precision nanotherapeutics. The goals of this review are to: 1) summarize the current data investigating the role of EVs and their bioactive cargo in MS pathogenesis, 2) provide a high level overview of advances and challenges in EV isolation and characterization for translational studies, and 3) conclude with future perspectives on this evolving field.
Collapse
Affiliation(s)
- Paola Loreto Palacio
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Michelle L Pleet
- Viral Immunology Section, Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eduardo Reátegui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Setty M Magaña
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
8
|
Age-related changes in multiple sclerosis and experimental autoimmune encephalomyelitis. Semin Immunol 2022; 59:101631. [PMID: 35752572 DOI: 10.1016/j.smim.2022.101631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
A better understanding of the pathological mechanisms that drive neurodegeneration in people living with multiple sclerosis (MS) is needed to design effective therapies to treat and/or prevent disease progression. We propose that CNS-intrinsic inflammation and re-modelling of the sub-arachnoid space of the leptomeninges sets the stage for neurodegeneration from the earliest stages of MS. While neurodegenerative processes are clinically silent early in disease, ageing results in neurodegenerative changes that become clinically manifest as progressive disability. Here we review pathological correlates of MS disease progression, highlight emerging mouse models that mimic key progressive changes in MS, and provide new perspectives on therapeutic approaches to protect against MS-associated neurodegeneration.
Collapse
|
9
|
Vollmer TL, Nair KV, Williams IM, Alvarez E. Multiple Sclerosis Phenotypes as a Continuum: The Role of Neurologic Reserve. Neurol Clin Pract 2021; 11:342-351. [PMID: 34476126 PMCID: PMC8382415 DOI: 10.1212/cpj.0000000000001045] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022]
Abstract
Purpose of Review This review presents the hypothesis that loss of neurologic reserve explains onset of progressive multiple sclerosis (PrMS). Recent Findings Evidence supporting the separate classification of PrMS and relapsing multiple sclerosis (RMS) is limited and does not explain PrMS or the response of these patients to therapy. Summary We argue that multiple sclerosis (MS) progresses along a continuum from RMS to PrMS, with differing levels of neurologic reserve accounting for phenotypic differences. In early MS, inflammation causes brain atrophy with symptoms buffered by neurologic reserve. As brain loss from normal aging and MS continues, reserve is depleted and effects of subclinical MS disease activity and aging are unmasked, manifesting as PrMS. Most therapies show limited benefit in PrMS; patients are older, have fewer inflammatory events, and the effects of aging cause continued loss of neurologic function, even if inflammation is terminated. Loss of neurologic reserve means patients with PrMS cannot recover function, unlike patients with RMS.
Collapse
Affiliation(s)
- Timothy L Vollmer
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| | - Kavita V Nair
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| | - Ian M Williams
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| | - Enrique Alvarez
- Department of Neurology (TLV, KVN, EA), University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora; Department of Clinical Pharmacy (KVN), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora; and Oxford PharmaGenesis (IMW), United Kingdom
| |
Collapse
|
10
|
Hjæresen S, Sejbaek T, Axelsson M, Vinsløv-Jensen H, Mortensen SK, Pihl-Jensen G, Novakova L, Christensen JDR, Pedersen CB, Halle B, Poulsen FR, Lautrup Frederiksen J, Zhang M, Benedikz E, Lycke J, Illes Z, Fex Svenningsen Å. The levels of the serine protease HTRA1 in cerebrospinal fluid correlate with progression and disability in multiple sclerosis. J Neurol 2021; 268:3316-3324. [PMID: 33661357 DOI: 10.1007/s00415-021-10489-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND High Temperature Requirement Serine Protease A1 (HTRA1) degrades extracellular matrix molecules (ECMs) and growth factors. It interacts with several proteins implicated in multiple sclerosis (MS), but has not previously been linked to the disease. OBJECTIVE Investigate the levels of HTRA1 in cerebrospinal fluid (CSF) in different subtypes of MS and brain tissue. METHODS Using ELISA, HTRA1 levels were compared in CSF from untreated patients with relapsing-remitting MS (RRMS, n = 23), secondary progressive MS (SPMS, n = 26) and healthy controls (HCs, n = 26). The effect of disease modifying therapies (DMTs) were examined in both patient groups. Cellular distribution in human brain was studied using immunochemistry and the oligointernode database, based on a single-nuclei RNA expression map. RESULTS HTRA1 increased in RRMS and SPMS compared to HCs. DMT decreased HTRA1 levels in both types of MS. Using ROC analysis, HTRA1 cut-offs could discriminate HCs from RRMS patients with 100% specificity and 82.6% sensitivity. In the brain, HTRA1 was expressed in glia and neurons. CONCLUSION HTRA1 is a promising CSF biomarker for MS correlating with disease- and disability progression. Most cell species of the normal and diseased CNS express HTRA1 and the expression pattern could reflect pathological processes involved in MS pathogenesis.
Collapse
Affiliation(s)
- Simone Hjæresen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Tobias Sejbaek
- Department of Regional Health Research, University of Southern Denmark, 5000, Odense, Denmark
- Department of Neurology, Southwest Jutland University Hospital, 6700, Esbjerg, Denmark
| | - Marcus Axelsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Helle Vinsløv-Jensen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
| | - Sif Kløvedal Mortensen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
| | - Gorm Pihl-Jensen
- Department of Neurology, University of Copenhagen, Rigshospitalet Glostrup, Copenhagen, Denmark
| | - Lenka Novakova
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Christian Bonde Pedersen
- Department of Neurosurgery, Clinical Institute, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Bo Halle
- Department of Neurosurgery, Clinical Institute, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Frantz Rom Poulsen
- Department of Neurosurgery, Clinical Institute, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | | | - Mengliang Zhang
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Eirikur Benedikz
- Faculty of Health Sciences, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
| | - Jan Lycke
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, and Department of Neurology, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Zsolt Illes
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark
- Department of Neurology, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark
| | - Åsa Fex Svenningsen
- Department of Molecular Medicine- Neurobiology Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000, Odense, Denmark.
- University of Southern Denmark, BRIDGE, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
11
|
Mockus TE, Munie A, Atkinson JR, Segal BM. Encephalitogenic and Regulatory CD8 T Cells in Multiple Sclerosis and Its Animal Models. THE JOURNAL OF IMMUNOLOGY 2021; 206:3-10. [PMID: 33443060 DOI: 10.4049/jimmunol.2000797] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS), a neuroinflammatory disease that affects millions worldwide, is widely thought to be autoimmune in etiology. Historically, research into MS pathogenesis has focused on autoreactive CD4 T cells because of their critical role in the animal model, experimental autoimmune encephalomyelitis, and the association between MS susceptibility and single-nucleotide polymorphisms in the MHC class II region. However, recent studies have revealed prominent clonal expansions of CD8 T cells within the CNS during MS. In this paper, we review the literature on CD8 T cells in MS, with an emphasis on their potential effector and regulatory properties. We discuss the impact of disease modifying therapies, currently prescribed to reduce MS relapse rates, on CD8 T cell frequency and function. A deeper understanding of the role of CD8 T cells in MS may lead to the development of more effective and selective immunomodulatory drugs for particular subsets of patients.
Collapse
Affiliation(s)
- Taryn E Mockus
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Ashley Munie
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Jeffrey R Atkinson
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210; .,Neuroscience Research Institute, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
12
|
Multiple sclerosis patients have reduced resting and increased activated CD4 +CD25 +FOXP3 +T regulatory cells. Sci Rep 2021; 11:10476. [PMID: 34006899 PMCID: PMC8131694 DOI: 10.1038/s41598-021-88448-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/01/2021] [Indexed: 12/26/2022] Open
Abstract
Resting and activated subpopulations of CD4+CD25+CD127loT regulatory cells (Treg) and CD4+CD25+CD127+ effector T cells in MS patients and in healthy individuals were compared. Peripheral blood mononuclear cells isolated using Ficoll Hypaque were stained with monoclonal antibodies and analysed by flow cytometer. CD45RA and Foxp3 expression within CD4+ cells and in CD4+CD25+CD127loT cells identified Population I; CD45RA+Foxp3+, Population II; CD45RA−Foxp3hi and Population III; CD45RA−Foxp3+ cells. Effector CD4+CD127+ T cells were subdivided into Population IV; memory /effector CD45RA− CD25−Foxp3− and Population V; effector naïve CD45RA+CD25−Foxp3−CCR7+ and terminally differentiated RA+ (TEMRA) effector memory cells. Chemokine receptor staining identified CXCR3+Th1-like Treg, CCR6+Th17-like Treg and CCR7+ resting Treg. Resting Treg (Population I) were reduced in MS patients, both in untreated and treated MS compared to healthy donors. Activated/memory Treg (Population II) were significantly increased in MS patients compared to healthy donors. Activated effector CD4+ (Population IV) were increased and the naïve/ TEMRA CD4+ (Population V) were decreased in MS compared to HD. Expression of CCR7 was mainly in Population I, whereas expression of CCR6 and CXCR3 was greatest in Populations II and intermediate in Population III. In MS, CCR6+Treg were lower in Population III. This study found MS is associated with significant shifts in CD4+T cells subpopulations. MS patients had lower resting CD4+CD25+CD45RA+CCR7+ Treg than healthy donors while activated CD4+CD25hiCD45RA−Foxp3hiTreg were increased in MS patients even before treatment. Some MS patients had reduced CCR6+Th17-like Treg, which may contribute to the activity of MS.
Collapse
|
13
|
Aqel SI, Yang X, Kraus EE, Song J, Farinas MF, Zhao EY, Pei W, Lovett-Racke AE, Racke MK, Li C, Yang Y. A STAT3 inhibitor ameliorates CNS autoimmunity by restoring Teff:Treg balance. JCI Insight 2021; 6:142376. [PMID: 33411696 PMCID: PMC7934926 DOI: 10.1172/jci.insight.142376] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Reestablishing an appropriate balance between T effector cells (Teff) and Tregs is essential for correcting autoimmunity. Multiple sclerosis (MS) is an immune-mediated chronic CNS disease characterized by neuroinflammation, demyelination, and neuronal degeneration, in which the Teff:Treg balance is skewed toward pathogenic Teffs Th1 and Th17 cells. STAT3 is a key regulator of Teff:Treg balance. Using the structure-based design, we have developed a potentially novel small-molecule prodrug LLL12b that specifically inhibits STAT3 and suppresses Th17 differentiation and expansion. Moreover, LLL12b regulates the fate decision between Th17 and Tregs in an inflammatory environment, shifting Th17:Treg balance toward Tregs and favoring the resolution of inflammation. Therapeutic administration of LLL12b after disease onset significantly suppresses disease progression in adoptively transferred, chronic, and relapsing-remitting experimental autoimmune encephalomyelitis. Disease relapses were also significantly suppressed by LLL12b given during the remission phase. Additionally, LLL12b shifts Th17:Treg balance of CD4+ T cells from MS patients toward Tregs and increases Teff sensitivity to Treg-mediated suppression. These data suggest that selective inhibition of STAT3 by the small molecule LLL12b recalibrates the effector and regulatory arms of CD4+ T responses, representing a potentially clinically translatable therapeutic strategy for MS.
Collapse
Affiliation(s)
- Saba I Aqel
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Xiaozhi Yang
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Emma E Kraus
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Jinhua Song
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Marissa F Farinas
- Neuroscience program, College of Arts and Sciences, OSU, Columbus, Ohio, USA
| | - Erin Y Zhao
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Wei Pei
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, OSU Wexner Medical Center, Columbus, Ohio, USA
| | - Michael K Racke
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA.,Quest Diagnostics, Secaucus, New Jersey, USA
| | - Chenglong Li
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Yuhong Yang
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA.,Department of Microbial Infection and Immunity, OSU Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
14
|
Maltby VE, Lea RA, Burnard S, Xavier A, Van Cao T, White N, Kennedy D, Groen K, Sanders KA, Seeto R, Bray S, Gresle M, Laverick L, Butzkueven H, Scott RJ, Lechner-Scott J. Epigenetic differences at the HTR2A locus in progressive multiple sclerosis patients. Sci Rep 2020; 10:22217. [PMID: 33335118 PMCID: PMC7747721 DOI: 10.1038/s41598-020-78809-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 11/22/2020] [Indexed: 11/30/2022] Open
Abstract
The pathology of progressive multiple sclerosis (MS) is poorly understood. We have previously assessed DNA methylation in the CD4+ T cells of relapsing–remitting (RR) MS patients compared to healthy controls and identified differentially methylated regions (DMRs) in HLA-DRB1 and RNF39. This study aimed to investigate the DNA methylation profiles of the CD4+ T cells of progressive MS patients. DNA methylation was measured in two separate case/control cohorts using the Illumina 450K/EPIC arrays and data was analysed with the Chip Analysis Methylation Pipeline (ChAMP). Single nucleotide polymorphisms (SNPs) were assessed using the Illumina Human OmniExpress24 arrays and analysed using PLINK. Expression was assessed using the Illumina HT12 array and analysed in R using a combination of Limma and Illuminaio. We identified three DMRs at HTR2A, SLC17A9 and HDAC4 that were consistent across both cohorts. The DMR at HTR2A is located within the bounds of a haplotype block; however, the DMR remained significant after accounting for SNPs in the region. No expression changes were detected in any DMRs. HTR2A is differentially methylated in progressive MS independent of genotype. This differential methylation is not evident in RRMS, making it a potential biomarker of progressive disease.
Collapse
Affiliation(s)
- Vicki E Maltby
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Rodney A Lea
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Sean Burnard
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Alexandre Xavier
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Thao Van Cao
- Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Nicole White
- Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Daniel Kennedy
- Institute of Health and Biomedical Innovations, Genomics Research Centre, Queensland University of Technology, Kelvin Grove, QLD, 4059, Australia
| | - Kira Groen
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Katherine A Sanders
- Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.,Centre for Anatomical and Human Sciences, Hull York Medical School, Hull, UK
| | - Rebecca Seeto
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Samara Bray
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia.,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Melissa Gresle
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia.,Royal Melbourne Hospital, Melbourne, VIC, Australia.,MS and Neuroimmunology Unit, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Louise Laverick
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia.,Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Alfred Hospital, Melbourne, VIC, Australia.,MS and Neuroimmunology Unit, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia.,Division of Molecular Genetics, Pathology North, John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia.,Centre for Cancer Research, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Jeannette Lechner-Scott
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2308, Australia. .,Centre for Brain and Mental Health, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia. .,Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
15
|
Tavazzi E, Zivadinov R, Dwyer MG, Jakimovski D, Singhal T, Weinstock-Guttman B, Bergsland N. MRI biomarkers of disease progression and conversion to secondary-progressive multiple sclerosis. Expert Rev Neurother 2020; 20:821-834. [PMID: 32306772 DOI: 10.1080/14737175.2020.1757435] [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] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Conventional imaging measures remain a key clinical tool for the diagnosis multiple sclerosis (MS) and monitoring of patients. However, most measures used in the clinic show unsatisfactory performance in predicting disease progression and conversion to secondary progressive MS. AREAS COVERED Sophisticated imaging techniques have facilitated the identification of imaging biomarkers associated with disease progression, such as global and regional brain volume measures, and with conversion to secondary progressive MS, such as leptomeningeal contrast enhancement and chronic inflammation. The relevance of emerging imaging approaches partially overcoming intrinsic limitations of traditional techniques is also discussed. EXPERT OPINION Imaging biomarkers capable of detecting tissue damage early on in the disease, with the potential to be applied in multicenter trials and at an individual level in clinical settings, are strongly needed. Several measures have been proposed, which exploit advanced imaging acquisitions and/or incorporate sophisticated post-processing, can quantify irreversible tissue damage. The progressively wider use of high-strength field MRI and the development of more advanced imaging techniques will help capture the missing pieces of the MS puzzle. The ability to more reliably identify those at risk for disability progression will allow for earlier intervention with the aim to favorably alter the disease course.
Collapse
Affiliation(s)
- Eleonora Tavazzi
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York , Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York , Buffalo, NY, USA.,Translational Imaging Center, Clinical and Translational Science Institute, University at Buffalo, The State University of New York , Buffalo, NY, USA
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York , Buffalo, NY, USA
| | - Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York , Buffalo, NY, USA
| | - Tarun Singhal
- PET Imaging Program in Neurologic Diseases and Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Disease, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School , Boston, MA, USA
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York , Buffalo, NY, USA
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York , Buffalo, NY, USA.,IRCCS, Fondazione Don Carlo Gnocchi , Milan, Italy
| |
Collapse
|
16
|
Kalra S, Lowndes C, Durant L, Strange RC, Al-Araji A, Hawkins CP, Curnow SJ. Th17 cells increase in RRMS as well as in SPMS, whereas various other phenotypes of Th17 increase in RRMS only. Mult Scler J Exp Transl Clin 2020; 6:2055217319899695. [PMID: 32064115 PMCID: PMC6990617 DOI: 10.1177/2055217319899695] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 11/17/2022] Open
Abstract
Background The nature and extent of inflammation seen in multiple sclerosis (MS) varies throughout the course of the disease. Changes seen in CD4+ T-helper cells in relapsing–remitting (RR) MS and secondary progressive (SP) MS might differ qualitatively and/or quantitatively. Objective The objective of this paper is to study the frequencies of all major CD4+ T-helper subtypes – Th17, Th22 and Th1 lineage cells – in relapse, remission and secondary progression alongside CCR6 status, a chemokine receptor involved in migration of these cells into the central nervous system. Methods We compared 100 patients (50 RRMS and 50 SPMS) and 50 healthy volunteers and performed flow cytometric analysis of lymphocytes in blood samples. Results We demonstrated raised frequencies of various cell types along the Th17 axis; Th17, Th17.1 (IL-17+ interferon gamma+) and dual IL-17+ IL-22+ cells in RRMS. Th22 and CCR6+ Th1 cells (nonclassical Th1) were also increased in RRMS. All these cells were CCR6+. Only Th17 frequencies were elevated in SPMS. Conclusions Increased frequencies of Th17 cells are implicated both in RRMS and SPMS. The CCR6 pathway includes Th17, Th22 and Th1 nonclassical cells, of which Th22 and Th1 cells represent the greatest subsets in MS.
Collapse
Affiliation(s)
- S Kalra
- Royal Stoke MS Centre of Excellence, Neurology Department, University Hospital North Midlands NHS Trust, UK
| | - C Lowndes
- Royal Stoke MS Centre of Excellence, Neurology Department, University Hospital North Midlands NHS Trust, UK
| | - L Durant
- Centre for Translational Inflammation Research, Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK
| | - R C Strange
- Institute for Science and Technology in Medicine, Keele University Medical School, UK
| | - A Al-Araji
- Royal Stoke MS Centre of Excellence, Neurology Department, University Hospital North Midlands NHS Trust, UK
| | - Clive P Hawkins
- Royal Stoke MS Centre of Excellence, Neurology Department, University Hospital North Midlands NHS Trust, UK
| | - S John Curnow
- Centre for Translational Inflammation Research, Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, UK
| |
Collapse
|
17
|
Shaker OG, Golam RM, Ayoub S, Daker LI, Elguaad MKA, Said ES, Khalil MAF. Correlation between LincR-Gng2-5'and LincR-Epas1-3'as with the severity of multiple sclerosis in Egyptian patients. Int J Neurosci 2019; 130:515-521. [PMID: 31790618 DOI: 10.1080/00207454.2019.1695610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Multiple sclerosis (MS) is an immune-mediated disorder. Long noncoding RNAs (lncRNAs, LncR, Linc RNA) have role in many autoimmune and inflammatory disorders, including MS. LincR-Gng2-5 AS locus in T helper 1 cell (TH1) and LincR-Epas1-3AS in T helper 2 cell (TH2) cell were located in a genomic region rich in genes code for proteins with immune regulatory function. Our aim was to evaluate the LincR-Gng2-5' and LincR-Epas1-3'AS fold change in blood of MS patients versus healthy controls and correlate it with disease severity, assessed based on Expanded Disability Status Scale (EDSS).Material and Methods: Sixty MS patients 42 relapsing remitting (RR, RRMS), 18 Secondary progressive (SP, SPMS) and sixty controls (age-matched and sex-matched) were studied. Blood of patients and control group undergone the investigation of LincR-Gng2-5' and LincR-Epas1-3'AS fold change by real-time PCR. Fold change >2 and p < .05 represent significant result.Results: LincR-Gng2-5' was significantly upregulated in MS patients with mean fold change (2.559) and (p = .03). Meanwhile, LincR-Epas1-3'AS levels were significantly downregulated with mean fold change (0.5964) and (p < .004). Patients with SP showed a significantly higher level of LincR-Gng2-5-fold change (3.71 ± 0.7) than that of RR (1.33 ± 0.3). LincR-Epas1-3'AS was markedly reduced among SP (0.43 ± 0.2) than that of RR (0.66 ± 0.1) but with no significant difference. As regards disease severity (EDSS); there was a significant positive correlation with LincR-Gng2-5 and negative correlation with LincR-Epas1-3'AS. LincR- Gng2-5and LincR-Epas1-3'AS, both are dysregulated in MS patient suggesting a role in disease pathogenesis.Conclusion: LincR-Gng2-5 AS and LincR-Epas1-3'AS fold change are correlated to MS severity (EDSS).
Collapse
Affiliation(s)
- Olfat G Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Rehab M Golam
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Shymaa Ayoub
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Lamiaa I Daker
- Department of Neurology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Mohamed K Abd Elguaad
- Department of Medical Physiology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Eman S Said
- Department of Clinical Pharmacology, Faculty of Medicine, Fayoum University, Fayoum, Egypt.,Department of Pharmacology and Toxicology Collage of Pharmacy, Qassim University, Saudi Arabia
| | - Mahmoud A F Khalil
- Department of Microbiology and Immunology, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
18
|
Haghmorad D, Yazdanpanah E, Jadid Tavaf M, Zargarani S, Soltanmohammadi A, Mahmoudi MB, Mahmoudi M. Prevention and treatment of experimental autoimmune encephalomyelitis induced mice with 1, 25-dihydroxyvitamin D 3. Neurol Res 2019; 41:943-957. [PMID: 31402771 DOI: 10.1080/01616412.2019.1650218] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a complex inflammatory and demyelinating disease of the central nervous system (CNS) frequently starts in young adulthood. Demyelination, inflammatory and axonal damage in the CNS is the pathological hallmark of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. 1, 25-dihydroxyvitamin D3 (Vitamin D3) is involved in calcium regulation, phosphorus homeostasis, and bone mineralization. In addition, vitamin D3 has potential inhibitory effects on immune cells in various inflammatory and autoimmunity disease. C57BL/6 female mice were divided into prevention groups (low, middle and high doses) and treatment groups (middle and high doses). Prevention groups received vitamin D3 2 weeks before EAE induction, and treatment groups were treated with vitamin D3 simultaneous with EAE induction. Vitamin D3 inhibits the development of EAE in a dose-dependent manner. Histological studies revealed reduced demyelination and limited infiltration into CNS, moreover vitamin D3 increased the production of IL-4, IL-10, and TGF-β, while a significant reduction in the production of IFN-γ, IL-6, TNF-α, and IL-17 was observed. Flow cytometry results for CD4+ T cell subsets in compliance with ELISA cytokine assay results showed a significant decrease in the percentage of Th1 and Th17, but also a significant increase in the percentage of Th2 and Treg for middle and high dose vitamin D3 treated mice. Real-time PCR results indicated that middle and high dose vitamin D3 treatment reduced T-bet and ROR-γt expression, but enhanced GATA3 and Foxp3 expression. Real-Time PCR results in CNS for T cell subsets related cytokines and transcription factors supported the results of flow cytometry and ELISA. This study indicated that middle and high doses of vitamin D3 deviate the balance between Th1/Th2 and Th17/Treg to Th2 and Treg. Moreover, vitamin D3 could reduce the incidence and severity of EAE clinical disease.
Collapse
Affiliation(s)
- Dariush Haghmorad
- Department of Pathology and Laboratory Medicine, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran.,Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Department of Immunology and Allergy, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Maryam Jadid Tavaf
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | - Simin Zargarani
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | - Azita Soltanmohammadi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences , Semnan , Iran
| | | | - Mahmoud Mahmoudi
- Immunology Research Center, Department of Immunology and Allergy, Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
19
|
The Diversity of Encephalitogenic CD4+ T Cells in Multiple Sclerosis and Its Animal Models. J Clin Med 2019; 8:jcm8010120. [PMID: 30669462 PMCID: PMC6352150 DOI: 10.3390/jcm8010120] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 02/06/2023] Open
Abstract
Autoreactive CD4+ T cells, which target antigens in central nervous system (CNS) myelin, are widely believed to play a critical role in the pathogenesis of multiple sclerosis (MS) in concert with other immune effectors. This theory is supported by data from animal model experiments, genome-wide association studies, and immune profiles of individuals with MS. Furthermore, disease modifying agents that target lymphocytes significantly reduce the rate of MS clinical exacerbations. However, the properties of myelin-reactive CD4+ T cells that are critical for their pathogenic activities are not understood completely. This article reviews the literature on encephalitogenic CD4+ T cells, with an emphasis on T-helper (Th) lineage and cytokine production. An increased understanding of the spectrum of encephalitogenic T cells and how they differ from protective subsets is necessary for the development of the next generation of more effective and safer immunomodulatory therapies customized for individuals with MS and related disorders.
Collapse
|
20
|
Ramirez SH, Andrews AM, Paul D, Pachter JS. Extracellular vesicles: mediators and biomarkers of pathology along CNS barriers. Fluids Barriers CNS 2018; 15:19. [PMID: 29960602 PMCID: PMC6026502 DOI: 10.1186/s12987-018-0104-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous, nano-sized vesicles that are shed into the blood and other body fluids, which disperse a variety of bioactive molecules (e.g., protein, mRNA, miRNA, DNA and lipids) to cellular targets over long and short distances. EVs are thought to be produced by nearly every cell type, however this review will focus specifically on EVs that originate from cells at the interface of CNS barriers. Highlighted topics include, EV biogenesis, the production of EVs in response to neuroinflammation, role in intercellular communication and their utility as a therapeutic platform. In this review, novel concepts regarding the use of EVs as biomarkers for BBB status and as facilitators for immune neuroinvasion are also discussed. Future directions and prospective are covered along with important unanswered questions in the field of CNS endothelial EV biology.
Collapse
Affiliation(s)
- Servio H Ramirez
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, 3500 N Broad St, Philadelphia, PA, 19140, USA. .,Shriners Hospital Pediatric Research Center, Philadelphia, PA, 19140, USA. .,Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| | - Allison M Andrews
- Department of Pathology and Laboratory Medicine, The Lewis Katz School of Medicine at Temple University, 3500 N Broad St, Philadelphia, PA, 19140, USA.,Center for Substance Abuse Research, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Debayon Paul
- Department of Immunology, Blood-Brain Barrier Laboratory & Laser Capture Microdissection Core, UConn Health, 263 Farmington Ave., Farmington, CT, 06070, USA
| | - Joel S Pachter
- Department of Immunology, Blood-Brain Barrier Laboratory & Laser Capture Microdissection Core, UConn Health, 263 Farmington Ave., Farmington, CT, 06070, USA.
| |
Collapse
|
21
|
Iacobaeus E, Douagi I, Jitschin R, Marcusson-Ståhl M, Andrén AT, Gavin C, Lefsihane K, Davies LC, Mougiakakos D, Kadri N, Le Blanc K. Phenotypic and functional alterations of myeloid-derived suppressor cells during the disease course of multiple sclerosis. Immunol Cell Biol 2018; 96:820-830. [PMID: 29569304 DOI: 10.1111/imcb.12042] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system involving dysregulated encephalitogenic T cells. Myeloid-derived suppressor cells (MDSCs) have been recognized for their important function in regulating T-cell responses. Recent studies have indicated a role for MDSCs in autoimmune diseases, but their significance in MS is not clear. Here, we assessed the frequencies of CD14+ HLA-DRlow monocytic MDSCs (Mo-MDSCs) and CD33+ CD15+ CD11b+ HLA-DRlow granulocytic MDSCs (Gr-MDSCs) and investigated phenotypic and functional differences of Mo-MDSCs at different clinical stages of MS and in healthy subjects (HC). Increased frequencies of Mo-MDSCs (P < 0.05) and Gr-MDSCs (P < 0.05) were observed in relapsing-remitting MS patients during relapse (RRMS-relapse) compared to stable RRMS (RRMS-rem). Secondary progressive MS (SPMS) patients displayed a decreased frequency of Mo-MDSCs and Gr-MDSCs compared to HC (P < 0.05). Mo-MDSCs within RRMS patients expressed significantly higher cell surface protein levels of CD86 and CD163 compared to SPMS patients. Mo-MDSCs within SPMS exhibited decreased mRNA expression of interleukin-10 and heme oxygenase 1 compared to RRMS and HC. Analysis of T-cell regulatory function of Mo-MDSCs demonstrated T-cell suppressive capacity in RRMS and HCs, while Mo-MDSCs of SPMS promoted autologous T-cell proliferation, which aligned with a differential cytokine profile compared to RRMS and HCs. This study is the first to show phenotypic and functional shifts of MDSCs between clinical stages of MS, suggesting a role for MDSCs as a therapeutic target to prevent MS disease progression.
Collapse
Affiliation(s)
- Ellen Iacobaeus
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Division of Neurology, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Iyadh Douagi
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Regina Jitschin
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Maritha Marcusson-Ståhl
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anton Törnqvist Andrén
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Caroline Gavin
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Katia Lefsihane
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Lindsay C Davies
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Dimitrios Mougiakakos
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Nadir Kadri
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Katarina Le Blanc
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
22
|
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: 84] [Impact Index Per Article: 12.0] [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
|
23
|
Elieh-Ali-Komi D, Cao Y. Role of Mast Cells in the Pathogenesis of Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. Clin Rev Allergy Immunol 2018; 52:436-445. [PMID: 28025778 DOI: 10.1007/s12016-016-8595-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune disorder of the central nervous system (CNS), characterized by recurrent episodes of inflammatory demyelination and consequent axonal deterioration. The hallmark of the disease is the demyelinated plaque, a hypocellular area characterized by formation of astrocytic scars and infiltration of mononuclear cells. Recent studies have revealed that both innate and adaptive immune cells contribute to the pathogenesis of MS and its experimental autoimmune encephalomyelitis (EAE) model. Here, we review the current understanding of the role of mast cells in the pathogenesis of MS and EAE. Mast cells may act at the early stage that promote demyelination through interactions among mast cells, neurons, and other immune cells to mediate neuroinflammation. Studies from EAE model suggest that mast cells regulate adaptive autoimmune responses, present myelin antigens to T cells, disrupt the blood-brain barrier, and permit the entry of inflammatory cells and mediators into the CNS. Depletion or limiting mast cells could be a new promising therapeutic target for MS and EAE.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Immunology Research Center, Department of Immunology, and Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yonghao Cao
- Center for Synthetic Biology Engineering Research, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China. .,Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
24
|
Liu Y, Gibson SA, Benveniste EN, Qin H. Opportunities for Translation from the Bench: Therapeutic Intervention of the JAK/STAT Pathway in Neuroinflammatory Diseases. Crit Rev Immunol 2018; 35:505-27. [PMID: 27279046 DOI: 10.1615/critrevimmunol.2016015517] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathogenic CD4+ T cells and myeloid cells play critical roles in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. These immune cells secrete aberrantly high levels of pro-inflammatory cytokines that pathogenically bridge the innate and adaptive immune systems and damage neurons and oligodendrocytes. These cytokines include interleukin-2 (IL-2), IL-6, IL-12, IL-21, IL-23, granulocyte macrophage-colony stimulating factor (GM-CSF), and interferon-γ (IFN-γ). It is, therefore, not surprising that both the dysregulated expression of these cytokines and the subsequent activation of their downstream signaling cascades is a common feature in MS/EAE. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is utilized by numerous cytokines for signal transduction and is essential for the development and regulation of immune responses. Unbridled activation of the JAK/STAT pathway by pro-inflammatory cytokines has been demonstrated to be critically involved in the pathogenesis of MS/EAE. In this review, we discuss recent advancements in our understanding of the involvement of the JAK/STAT signaling pathway in the pathogenesis of MS/EAE, with a particular focus on therapeutic approaches to target the JAK/STAT pathway.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294; Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sara A Gibson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| |
Collapse
|
25
|
Giles DA, Washnock-Schmid JM, Duncker PC, Dahlawi S, Ponath G, Pitt D, Segal BM. Myeloid cell plasticity in the evolution of central nervous system autoimmunity. Ann Neurol 2018; 83:131-141. [PMID: 29283442 PMCID: PMC5876132 DOI: 10.1002/ana.25128] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/22/2017] [Accepted: 12/26/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Myeloid cells, including macrophages and dendritic cells, are a prominent component of central nervous system (CNS) infiltrates during multiple sclerosis (MS) and the animal model experimental autoimmune encephalomyelitis (EAE). Although myeloid cells are generally thought to be proinflammatory, alternatively polarized subsets can serve noninflammatory and/or reparative functions. Here we investigate the heterogeneity and biological properties of myeloid cells during central nervous system autoimmunity. METHODS Myeloid cell phenotypes in chronic active MS lesions were analyzed by immunohistochemistry. In addition, immune cells were isolated from the CNS during exacerbations and remissions of EAE and characterized by flow cytometric, genetic, and functional assays. RESULTS Myeloid cells expressing inducible nitric oxide synthase (iNOS), indicative of a proinflammatory phenotype, were detected in the actively demyelinating rim of chronic active MS lesions, whereas macrophages expressing mannose receptor (CD206), a marker of alternatively polarized human myeloid cells, were enriched in the quiescent lesion core. During EAE, CNS-infiltrating myeloid cells, as well as microglia, shifted from expression of proinflammatory markers to expression of noninflammatory markers immediately prior to clinical remissions. Murine CNS myeloid cells expressing the alternative lineage marker arginase-1 (Arg1) were partially derived from iNOS+ precursors and were deficient in activating encephalitogenic T cells compared with their Arg1- counterparts. INTERPRETATION These observations demonstrate the heterogeneity of CNS myeloid cells, their evolution during the course of autoimmune demyelinating disease, and their plasticity on the single cell level. Future therapeutic strategies for disease modification in individuals with MS may be focused on accelerating the transition of CNS myeloid cells from a proinflammatory to a noninflammatory phenotype. Ann Neurol 2018;83:131-141.
Collapse
Affiliation(s)
- David A. Giles
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA
| | - Jesse M. Washnock-Schmid
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Patrick C. Duncker
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Somiah Dahlawi
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Gerald Ponath
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - David Pitt
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA
| | - Benjamin M. Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Duncker PC, Stoolman JS, Huber AK, Segal BM. GM-CSF Promotes Chronic Disability in Experimental Autoimmune Encephalomyelitis by Altering the Composition of Central Nervous System-Infiltrating Cells, but Is Dispensable for Disease Induction. THE JOURNAL OF IMMUNOLOGY 2017; 200:966-973. [PMID: 29288202 DOI: 10.4049/jimmunol.1701484] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/01/2017] [Indexed: 11/19/2022]
Abstract
GM-CSF has been portrayed as a critical cytokine in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and, ostensibly, in multiple sclerosis. C57BL/6 mice deficient in GM-CSF are resistant to EAE induced by immunization with myelin oligodendrocyte glycoprotein (MOG)35-55 The mechanism of action of GM-CSF in EAE is poorly understood. In this study, we show that GM-CSF augments the accumulation of MOG35-55-specific T cells in the skin draining lymph nodes of primed mice, but it is not required for the development of encephalitogenic T cells. Abrogation of GM-CSF receptor signaling in adoptive transfer recipients of MOG35-55-specific T cells did not alter the incidence of EAE or the trajectory of its initial clinical course, but it limited the extent of chronic CNS tissue damage and neurologic disability. The attenuated clinical course was associated with a relative dearth of MOG35-55-specific T cells, myeloid dendritic cells, and neutrophils, as well as an abundance of B cells, within CNS infiltrates. Our data indicate that GM-CSF drives chronic tissue damage and disability in EAE via pleiotropic pathways, but it is dispensable during early lesion formation and the onset of neurologic deficits.
Collapse
Affiliation(s)
- Patrick C Duncker
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109.,Multiple Sclerosis Center, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109.,Graduate Program in Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109; and
| | - Joshua S Stoolman
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109.,Multiple Sclerosis Center, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109.,Graduate Program in Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109; and
| | - Amanda K Huber
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109.,Multiple Sclerosis Center, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Benjamin M Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109; .,Multiple Sclerosis Center, Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109.,Graduate Program in Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109; and.,Neuroscience Program, University of Michigan School of Medicine, Ann Arbor, MI 48109
| |
Collapse
|
27
|
Shrestha B, Jiang X, Ge S, Paul D, Chianchiano P, Pachter JS. Spatiotemporal resolution of spinal meningeal and parenchymal inflammation during experimental autoimmune encephalomyelitis. Neurobiol Dis 2017; 108:159-172. [PMID: 28844788 DOI: 10.1016/j.nbd.2017.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/10/2017] [Accepted: 08/18/2017] [Indexed: 01/14/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) induced by active immunization of C57BL/6 mice with peptide from myelin oligodendrocyte protein (MOG35-55), is a neuroinflammatory, demyelinating disease widely recognized as an animal model of multiple sclerosis (MS). Typically, EAE presents with an ascending course of paralysis, and inflammation that is predominantly localized to the spinal cord. Recent studies have further indicated that inflammation - in both MS and EAE - might initiate within the meninges and propagate from there to the underlying parenchyma. However, the patterns of inflammation within the respective meningeal and parenchymal compartments along the length of the spinal cord, and the progression with which these patterns develop during EAE, have yet to be detailed. Such analysis could hold key to identifying factors critical for spreading, as well as constraining, inflammation along the neuraxis. To address this issue, high-resolution 3-dimensional (3D) confocal microscopy was performed to visualize, in detail, the sequence of leukocyte infiltration at distinct regions of the spinal cord. High quality virtual slide scanning for imaging the entire spinal cord using epifluorescence was further conducted to highlight the directionality and relative degree of inflammation. Meningeal inflammation was found to precede parenchymal inflammation at all levels of the spinal cord, but did not develop equally or simultaneously throughout the subarachnoid space (SAS) of the meninges. Instead, meningeal inflammation was initially most obvious in the caudal SAS, from which it progressed to the immediate underlying parenchyma, paralleling the first signs of clinical disease in the tail and hind limbs. Meningeal inflammation could then be seen to extend in the caudal-to-rostral direction, followed by a similar, but delayed, trajectory of parenchymal inflammation. To additionally determine whether the course of ascending paralysis and leukocyte infiltration during EAE is reflected in differences in inflammatory gene expression by meningeal and parenchymal microvessels along the spinal cord, laser capture microdissection (LCM) coupled with gene expression profiling was performed. Expression profiles varied between these respective vessel populations at both the cervical and caudal levels of the spinal cord during disease progression, and within each vessel population at different levels of the cord at a given time during disease. These results reinforce a significant role for the meninges in the development and propagation of central nervous system inflammation associated with MS and EAE.
Collapse
Affiliation(s)
- Bandana Shrestha
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Xi Jiang
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Shujun Ge
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Debayon Paul
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Peter Chianchiano
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| | - Joel S Pachter
- Blood-Brain Barrier Laboratory, Dept. of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, United States.
| |
Collapse
|
28
|
Arellano G, Acuña E, Reyes LI, Ottum PA, De Sarno P, Villarroel L, Ciampi E, Uribe-San Martín R, Cárcamo C, Naves R. Th1 and Th17 Cells and Associated Cytokines Discriminate among Clinically Isolated Syndrome and Multiple Sclerosis Phenotypes. Front Immunol 2017; 8:753. [PMID: 28713377 PMCID: PMC5491887 DOI: 10.3389/fimmu.2017.00753] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/13/2017] [Indexed: 12/02/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory, and demyelinating disease of the central nervous system. It is a heterogeneous pathology that can follow different clinical courses, and the mechanisms that underlie the progression of the immune response across MS subtypes remain incompletely understood. Here, we aimed to determine differences in the immunological status among different MS clinical subtypes. Blood samples from untreated patients diagnosed with clinically isolated syndrome (CIS) (n = 21), different clinical forms of MS (n = 62) [relapsing–remitting (RRMS), secondary progressive, and primary progressive], and healthy controls (HCs) (n = 17) were tested for plasma levels of interferon (IFN)-γ, IL-10, TGF-β, IL-17A, and IL-17F by immunoanalysis. Th1 and Th17 lymphocyte frequencies were determined by flow cytometry. Our results showed that IFN-γ levels and the IFN-γ/IL-10 ratio were higher in CIS patients than in RRMS patients and HC. Th1 cell frequencies were higher in CIS and RRMS than in progressive MS, and RRMS had a higher Th17 frequency than CIS. The Th1/Th17 cell ratio was skewed toward Th1 in CIS compared to MS phenotypes and HC. Receiver operating characteristic statistical analysis determined that IFN-γ, the IFN-γ/IL-10 ratio, Th1 cell frequency, and the Th1/Th17 cell ratio discriminated among CIS and MS subtypes. A subanalysis among patients expressing high IL-17F levels showed that IL-17F and the IFN-γ/IL-17F ratio discriminated between disease subtypes. Overall, our data showed that CIS and MS phenotypes displayed distinct Th1- and Th17-related cytokines and cell profiles and that these immune parameters discriminated between clinical forms. Upon validation, these parameters might be useful as biomarkers to predict disease progression.
Collapse
Affiliation(s)
- Gabriel Arellano
- School of Medicine, Institute of Biomedical Sciences (ICBM), Universidad de Chile, Santiago, Chile
| | - Eric Acuña
- School of Medicine, Institute of Biomedical Sciences (ICBM), Universidad de Chile, Santiago, Chile
| | - Lilian I Reyes
- Faculty of Science, Universidad San Sebastián, Santiago, Chile
| | - Payton A Ottum
- School of Medicine, Institute of Biomedical Sciences (ICBM), Universidad de Chile, Santiago, Chile
| | - Patrizia De Sarno
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Luis Villarroel
- Department of Public Health, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ethel Ciampi
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Neurology Service, Hospital Sotero del Río, Santiago, Chile
| | - Reinaldo Uribe-San Martín
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Neurology Service, Hospital Sotero del Río, Santiago, Chile
| | - Claudia Cárcamo
- Department of Neurology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Naves
- School of Medicine, Institute of Biomedical Sciences (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
29
|
Yang H, Liu C, Jiang J, Wang Y, Zhang X. Celastrol Attenuates Multiple Sclerosis and Optic Neuritis in an Experimental Autoimmune Encephalomyelitis Model. Front Pharmacol 2017; 8:44. [PMID: 28239352 PMCID: PMC5301323 DOI: 10.3389/fphar.2017.00044] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 01/20/2017] [Indexed: 01/21/2023] Open
Abstract
This study was aimed to evaluate the effects of celastrol, a natural compound with multiple bioactivities, on multiple sclerosis and optic neuritis (ON) in rat experimental autoimmune encephalomyelitis (EAE). EAE was induced in Sprague Dawley rats using myelin basic protein, and the animals received daily intraperitoneal injections of celastrol or vehicle for 13 days. The EAE rats showed abnormal neurobehavior and inflammatory infiltration and demyelination in the spinal cord. Significantly upregulated mRNA expression of pro-inflammatory cytokines interferon-γ and interleukin-17 and downregulated anti-inflammatory cytokines interleukin-4 were found in the spinal cord of EAE rats. In the study of ON, severely inflammatory responses like in the spinal cord were also seen in the optic nerve, as well as obvious microgliosis. Furthermore, activation of nuclear factor kappa-B and upregulated inducible nitric oxide synthase was observed in the optic nerve. In addition, apoptosis of retinal ganglion cells and dysregulation of apoptotic-associated proteins in the optic nerve were found in EAE rats. Treatment of celastrol potently restored these changes. In most of the indexes, the effects of high dose of celastrol were better than the low dose. Our data conclude that administration of celastrol attenuates multiple sclerosis and ON in EAE via anti-inflammatory and anti-apoptotic effects. These findings provide new pre-clinical evidence for the use of celastrol in treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Hongbin Yang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University Harbin, China
| | - Chang Liu
- Department of Neurology, Harbin Fourth Hospital Harbin, China
| | - Jie Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University Harbin, China
| | - Yuena Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University Harbin, China
| | - Xiaoyu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University Harbin, China
| |
Collapse
|
30
|
Sanders KA, Benton MC, Lea RA, Maltby VE, Agland S, Griffin N, Scott RJ, Tajouri L, Lechner-Scott J. Next-generation sequencing reveals broad down-regulation of microRNAs in secondary progressive multiple sclerosis CD4+ T cells. Clin Epigenetics 2016; 8:87. [PMID: 27570566 PMCID: PMC5002332 DOI: 10.1186/s13148-016-0253-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/09/2016] [Indexed: 01/06/2023] Open
Abstract
Background Immunoactivation is less evident in secondary progressive MS (SPMS) compared to relapsing-remitting disease. MicroRNA (miRNA) expression is integral to the regulation of gene expression; determining their impact on immune-related cell functions, especially CD4+ T cells, during disease progression will advance our understanding of MS pathophysiology. This study aimed to compare miRNA profiles of CD4+ T cells from SPMS patients to healthy controls (HC) using whole miRNA transcriptome next-generation sequencing (NGS). Total RNA was extracted from CD4+ T cells and miRNA expression patterns analyzed using Illumina-based small-RNA NGS in 12 SPMS and 12 HC samples. Results were validated in a further cohort of 12 SPMS and 10 HC by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Results The ten most dysregulated miRNAs identified by NGS were selected for qPCR confirmation; five (miR-21-5p, miR-26b-5p, miR-29b-3p, miR-142-3p, and miR-155-5p) were confirmed to be down-regulated in SPMS (p < 0.05). SOCS6 is targeted by eight of these ten miRNAs. Consistent with this, SOCS6 expression is up-regulated in SPMS CD4+ T cells (p < 0.05). This is of particular interest as SOCS6 has previously been shown to act as a negative regulator of T cell activation. Conclusions Ninety-seven percent of miRNA candidates identified by NGS were down-regulated in SPMS. The down-regulation of miRNAs and increased expression of SOCS6 in SPMS CD4+ T cells may contribute to reduced immune system activity in progressive MS. Electronic supplementary material The online version of this article (doi:10.1186/s13148-016-0253-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katherine A Sanders
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland 4226 Australia ; Centre for Information-Based Medicine, Hunter Medical Research Institute, Newcastle, New South Wales 2305 Australia ; School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales 2308 Australia
| | - Miles C Benton
- Institute of Health and Biomedical Innovation, Genomics Research Centre, Brisbane, Queensland 4059 Australia
| | - Rod A Lea
- Institute of Health and Biomedical Innovation, Genomics Research Centre, Brisbane, Queensland 4059 Australia ; Centre for Information-Based Medicine, Hunter Medical Research Institute, Newcastle, New South Wales 2305 Australia
| | - Vicki E Maltby
- Centre for Information-Based Medicine, Hunter Medical Research Institute, Newcastle, New South Wales 2305 Australia ; School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales 2308 Australia
| | - Susan Agland
- Department of Neurology, Division of Medicine, John Hunter Hospital, Locked Bag 1, Hunter Region Mail Centre, Newcastle, NSW 2310 Australia
| | - Nathan Griffin
- Centre for Information-Based Medicine, Hunter Medical Research Institute, Newcastle, New South Wales 2305 Australia ; School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales 2308 Australia
| | - Rodney J Scott
- Centre for Information-Based Medicine, Hunter Medical Research Institute, Newcastle, New South Wales 2305 Australia ; School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales 2308 Australia ; Division of Molecular Genetics, Pathology North, Newcastle, New South Wales 2305 Australia
| | - Lotti Tajouri
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland 4226 Australia
| | - Jeannette Lechner-Scott
- Centre for Information-Based Medicine, Hunter Medical Research Institute, Newcastle, New South Wales 2305 Australia ; Department of Neurology, Division of Medicine, John Hunter Hospital, Locked Bag 1, Hunter Region Mail Centre, Newcastle, NSW 2310 Australia ; School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales 2308 Australia
| |
Collapse
|
31
|
Yokoyama K, Hattori N. Immunomodulatory effects of glatiramer acetate as they relate to stage-specific immune dysregulation in multiple sclerosis. Nihon Yakurigaku Zasshi 2016; 148:105-20. [PMID: 27478050 DOI: 10.1254/fpj.148.105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
32
|
Li X, Zhang Y, Yan Y, Ciric B, Ma CG, Chin J, Curtis M, Rostami A, Zhang GX. LINGO-1-Fc-Transduced Neural Stem Cells Are Effective Therapy for Chronic Stage Experimental Autoimmune Encephalomyelitis. Mol Neurobiol 2016; 54:4365-4378. [PMID: 27344330 DOI: 10.1007/s12035-016-9994-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/14/2016] [Indexed: 12/11/2022]
Abstract
The chronic stage multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system (CNS), remains refractory to current treatments. This refractory nature may be due to the fact that current treatments are primarily immunomodulatory, which prevent further demyelination but lack the capacity to promote remyelination. Several approaches, including transplantation of neural stem cells (NSCs) or antagonists to LINGO-1, a key part of the receptor complex for neuroregeneration inhibitors, have been effective in suppressing the acute stage of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. However, their effect on the chronic stage EAE is not known. Here, we show that transplantation of NSCs had only a slight therapeutic effect when treatment started at the chronic stage of EAE (e.g., injected at day 40 postimmunization). However, NSCs engineered to produce LINGO-1-Fc, a soluble LINGO-1 antagonist, significantly promoted neurological recovery as demonstrated by amelioration of clinical signs, improvement in axonal integrity, and enhancement of oligodendrocyte maturation and neuron repopulation. Significantly enhanced NAD production and Sirt2 expression were also found in the CNS of mice treated with LINGO-1-Fc-producing NSC. Moreover, differentiation of LINGO-1-Fc-producing NSCs into oligodendrocytes in vitro was largely diminished by an NAMPT inhibitor, indicating that LINGO-1-Fc enhances the NAMPT/NAD/Sirt2 pathway. Together, our study establishes a CNS-targeted, novel LINGO-1-Fc delivery system using NSCs, which represents a novel and effective NSC-based gene therapy approach for the chronic stage of MS.
Collapse
Affiliation(s)
- Xing Li
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuan Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yaping Yan
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cun-Gen Ma
- Institute of Brain Science, Department of Neurology, Shanxi Datong University Medical School, Datong, China
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Mark Curtis
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Dudvarski Stankovic N, Teodorczyk M, Ploen R, Zipp F, Schmidt MHH. Microglia-blood vessel interactions: a double-edged sword in brain pathologies. Acta Neuropathol 2016; 131:347-63. [PMID: 26711460 DOI: 10.1007/s00401-015-1524-y] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/09/2015] [Accepted: 12/12/2015] [Indexed: 12/12/2022]
Abstract
Microglia are long-living resident immune cells of the brain, which secure a stable chemical and physical microenvironment necessary for the proper functioning of the central nervous system (CNS). These highly dynamic cells continuously scan their environment for pathogens and possess the ability to react to damage-induced signals in order to protect the brain. Microglia, together with endothelial cells (ECs), pericytes and astrocytes, form the functional blood-brain barrier (BBB), a specialized endothelial structure that selectively separates the sensitive brain parenchyma from blood circulation. Microglia are in bidirectional and permanent communication with ECs and their perivascular localization enables them to survey the influx of blood-borne components into the CNS. Furthermore, they may stimulate the opening of the BBB, extravasation of leukocytes and angiogenesis. However, microglia functioning requires tight control as their dysregulation is implicated in the initiation and progression of numerous neurological diseases. Disruption of the BBB, changes in blood flow, introduction of pathogens in the sensitive CNS niche, insufficient nutrient supply, and abnormal secretion of cytokines or expression of endothelial receptors are reported to prime and attract microglia. Such reactive microglia have been reported to even escalate the damage of the brain parenchyma as is the case in ischemic injuries, brain tumors, multiple sclerosis, Alzheimer's and Parkinson's disease. In this review, we present the current state of the art of the causes and mechanisms of pathological interactions between microglia and blood vessels and explore the possibilities of targeting those dysfunctional interactions for the development of future therapeutics.
Collapse
Affiliation(s)
- Nevenka Dudvarski Stankovic
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Marcin Teodorczyk
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Robert Ploen
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Research Center for Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Research Center for Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Mirko H H Schmidt
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
34
|
Mormile R. Multiple sclerosis and susceptibility to celiac disease: an osteopontin gene haplotypes affair? Immunol Lett 2015; 163:132-133. [PMID: 25476232 DOI: 10.1016/j.imlet.2014.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 11/23/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Raffaella Mormile
- Division of Pediatrics and Neonatology, Moscati Hospital, Aversa, Italy.
| |
Collapse
|
35
|
Upregulation of immunomodulatory molecules by matrine treatment in experimental autoimmune encephalomyelitis. Exp Mol Pathol 2014; 97:470-6. [DOI: 10.1016/j.yexmp.2014.10.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/06/2014] [Indexed: 11/18/2022]
|