1
|
Ibrahim Fouad G, Mabrouk M, El-Sayed SAM, Abdelhameed MF, Rizk MZ, Beherei HH. Berberine-loaded iron oxide nanoparticles alleviate cuprizone-induced astrocytic reactivity in a rat model of multiple sclerosis. Biometals 2025; 38:203-229. [PMID: 39543075 PMCID: PMC11754386 DOI: 10.1007/s10534-024-00648-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/20/2024] [Indexed: 11/17/2024]
Abstract
Berberine (BBN) is a naturally occurring alkaloid as a secondary metabolite in many plants and exhibits several benefits including neuroprotective activities. However, data on the neuromodulating potential of nanoformulated BBN are still lacking. In the present study, BBN loaded within iron oxide nanoparticles (BBN-IONP) were prepared and characterized by transmission electron microscopy FTIR, X-ray photoelectron spectroscopy particle-size distribution, zeta potential, and HPLC. The remyelinating neuroprotective potential of BBN-IONP relative to free BBN was evaluated against cuprizone (CPZ)-induced neurotoxicity (rats administered 0.2% CPZ powder (w/w) for five weeks). CPZ rats were treated with either free BBN or IONP-BBN (50 mg/kg/day, orally) for 14 days. Cognitive function was estimated using Y-maze. Biochemically, total antioxidant capacity lipid peroxides and reduced glutathione in the brain tissue, as well as, serum interferon-gamma levels were estimated. Moreover, the genetic expression contents of myelin basic protein Matrix metallopeptidase-9 Tumor necrosis factor-α (TNF-α), and S100β were measured. The histopathological patterns and immunohistochemical assessment of Glial Fibrillary Acidic Protein in both cerebral cortex and hippocampus CA1 regions were investigated. CPZ-rats treated with either free BBN or IONP-BBN demonstrated memory restoring, anti-oxidative, anti-inflammatory, anti-astrocytic, and remyelinating activities. Comparing free BBN with IONP-BBN revealed that the latter altered the neuromodulating activities of BBN, showing superior neuroprotective activities of IONP-BBN relative to BBN. In conclusion, both forms of BBN possess neuroprotective potential. However, the use of IONPs for brain delivery and the safety of these nano-based forms need further investigation.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt.
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Sara A M El-Sayed
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mohamed F Abdelhameed
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt
| | - Maha Z Rizk
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| |
Collapse
|
2
|
Geraldes R, Arrambide G, Banwell B, Rovira À, Cortese R, Lassmann H, Messina S, Rocca MA, Waters P, Chard D, Gasperini C, Hacohen Y, Mariano R, Paul F, DeLuca GC, Enzinger C, Kappos L, Leite MI, Sastre-Garriga J, Yousry T, Ciccarelli O, Filippi M, Barkhof F, Palace J. The influence of MOGAD on diagnosis of multiple sclerosis using MRI. Nat Rev Neurol 2024; 20:620-635. [PMID: 39227463 DOI: 10.1038/s41582-024-01005-2] [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: 07/26/2024] [Indexed: 09/05/2024]
Abstract
Myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD) is an immune-mediated demyelinating disease that is challenging to differentiate from multiple sclerosis (MS), as the clinical phenotypes overlap, and people with MOGAD can fulfil the current MRI-based diagnostic criteria for MS. In addition, the MOG antibody assays that are an essential component of MOGAD diagnosis are not standardized. Accurate diagnosis of MOGAD is crucial because the treatments and long-term prognosis differ from those for MS. This Expert Recommendation summarizes the outcomes from a Magnetic Resonance Imaging in MS workshop held in Oxford, UK in May 2022, in which MS and MOGAD experts reflected on the pathology and clinical features of these disorders, the contributions of MRI to their diagnosis and the clinical use of the MOG antibody assay. We also critically reviewed the literature to assess the validity of distinctive imaging features in the current MS and MOGAD criteria. We conclude that dedicated orbital and spinal cord imaging (with axial slices) can inform MOGAD diagnosis and also illuminate differential diagnoses. We provide practical guidance to neurologists and neuroradiologists on how to navigate the current MOGAD and MS criteria. We suggest a strategy that includes useful imaging discriminators on standard clinical MRI and discuss imaging features detected by non-conventional MRI sequences that demonstrate promise in differentiating these two disorders.
Collapse
Affiliation(s)
- Ruth Geraldes
- NMO Service, Department of Neurology, Oxford University Hospitals, Oxford, UK.
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK.
- Wexham Park Hospital, Frimley Health Foundation Trust, Slough, UK.
| | - Georgina Arrambide
- Neurology-Neuroimmunology Department, Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Brenda Banwell
- Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Àlex Rovira
- Section of Neuroradiology, Department of Radiology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Rosa Cortese
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Silvia Messina
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
- Wexham Park Hospital, Frimley Health Foundation Trust, Slough, UK
| | - Mara Assunta Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Patrick Waters
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Declan Chard
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
- National Institute for Health Research (NIHR) University College London Hospitals (CLH) Biomedical Research Centre, London, UK
| | - Claudio Gasperini
- Multiple Sclerosis Centre, Department of Neurosciences, San Camillo-Forlanini Hospital, Rome, Italy
| | - Yael Hacohen
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Romina Mariano
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Christian Enzinger
- Department of Neurology, Medical University of Graz, Graz, Austria
- Division of Neuroradiology, Vascular and Interventional Radiology, Medical University of Graz, Graz, Austria
| | - Ludwig Kappos
- Research Center for Clinical Neuroimmunology and Neuroscience, University Hospital and University, Basel, Switzerland
| | - M Isabel Leite
- NMO Service, Department of Neurology, Oxford University Hospitals, Oxford, UK
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Jaume Sastre-Garriga
- Neurology-Neuroimmunology Department, Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Tarek Yousry
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Olga Ciccarelli
- Department of Neuroinflammation, Queen Square MS Centre, UCL Queen Square Institute of Neurology, London, UK
- University College London Hospitals (UCLH) National Institute for Health and Research (NIHR) Biomedical Research Centre (BRC), London, UK
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Jacqueline Palace
- NMO Service, Department of Neurology, Oxford University Hospitals, Oxford, UK.
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK.
| |
Collapse
|
3
|
Niedziela N, Nowak-Kiczmer M, Malciene L, Stasiołek M, Niedziela JT, Czuba ZP, Lis M, Sowa A, Adamczyk-Sowa M. Serum Vitamin D3 as a Potential Biomarker for Neuronal Damage in Smoldering Multiple Sclerosis. Int J Mol Sci 2024; 25:10502. [PMID: 39408830 PMCID: PMC11476431 DOI: 10.3390/ijms251910502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Permanent inflammatory demyelinating and neurodegenerative processes lead to neurological disability in patients with multiple sclerosis (MS). The anti-inflammatory properties of vitamin D3 (VitD) are well established, but its role in neurodegeneration is still uncertain. The usefulness of the serum concentration of VitD as a potential biomarker in evaluating brain injury in terms of recently known smoldering MS was under consideration. Methods: We assessed the concentrations of the parameters of brain injury (NF-H, GPAF, S100B, UCHL1) in the cerebrospinal fluid (CSF) of relapsing-remitting (RRMS, n = 123) and progressive MS (PMS, n = 88) patients in the group with normal levels of VitD (VitDn) and in the VitD deficiency group (VitDd). The levels of NF-H and UCHL1 were higher in the group of VitDd compared to VitDn. The higher serum levels of VitD were correlated with lower concentrations of GFAP, NF-H and S100B in the CSF of the whole group of MS patients and in women with MS as opposed to the levels of UCHL1. In men, there were noted negative correlations between the levels of serum VitD and GFAP and NF-H in CSF but not between VitD and S100B and UCHL1. The negative correlations were observed between VitD and the selected parameters of brain injury in MS patients, in women as well as in men. The concentrations of serum VitD together with selected parameters of brain injury in CSF seem to be promising biomarkers of neurodegeneration processes in smoldering MS.
Collapse
Affiliation(s)
- Natalia Niedziela
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3-go Maja 13-15, 41-800 Zabrze, Poland; (M.N.-K.); (A.S.); (M.A.-S.)
| | - Maria Nowak-Kiczmer
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3-go Maja 13-15, 41-800 Zabrze, Poland; (M.N.-K.); (A.S.); (M.A.-S.)
| | - Lina Malciene
- Klaipeda University Hospital, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| | - Mariusz Stasiołek
- Department of Neurology, Medical University of Lodz, ul. Kopcińskiego 22, 90-153 Lodz, Poland;
| | - Jacek T. Niedziela
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Silesian Centre for Heart Disease ul, M.C. Sklodowskiej 9, 41-800 Zabrze, Poland;
| | - Zenon P. Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. Jordana 19, 41-808 Zabrze, Poland
| | - Martyna Lis
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3-go Maja 13-15, 41-800 Zabrze, Poland; (M.N.-K.); (A.S.); (M.A.-S.)
| | - Agata Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3-go Maja 13-15, 41-800 Zabrze, Poland; (M.N.-K.); (A.S.); (M.A.-S.)
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3-go Maja 13-15, 41-800 Zabrze, Poland; (M.N.-K.); (A.S.); (M.A.-S.)
| |
Collapse
|
4
|
Sanabria-Castro A, Alape-Girón A, Flores-Díaz M, Echeverri-McCandless A, Parajeles-Vindas A. Oxidative stress involvement in the molecular pathogenesis and progression of multiple sclerosis: a literature review. Rev Neurosci 2024; 35:355-371. [PMID: 38163257 DOI: 10.1515/revneuro-2023-0091] [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: 08/16/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune debilitating disease of the central nervous system caused by a mosaic of interactions between genetic predisposition and environmental factors. The pathological hallmarks of MS are chronic inflammation, demyelination, and neurodegeneration. Oxidative stress, a state of imbalance between the production of reactive species and antioxidant defense mechanisms, is considered one of the key contributors in the pathophysiology of MS. This review is a comprehensive overview of the cellular and molecular mechanisms by which oxidant species contribute to the initiation and progression of MS including mitochondrial dysfunction, disruption of various signaling pathways, and autoimmune response activation. The detrimental effects of oxidative stress on neurons, oligodendrocytes, and astrocytes, as well as the role of oxidants in promoting and perpetuating inflammation, demyelination, and axonal damage, are discussed. Finally, this review also points out the therapeutic potential of various synthetic antioxidants that must be evaluated in clinical trials in patients with MS.
Collapse
Affiliation(s)
- Alfredo Sanabria-Castro
- Unidad de Investigación, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José, 10103, Costa Rica
- Departamento de Farmacología, Toxicología y Farmacodependencia, Facultad de Farmacia, Universidad de Costa Rica, San Pedro de Montes de Oca, 11501, Costa Rica
| | - Alberto Alape-Girón
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, Dulce Nombre Vázquez de Coronado, 11103, Costa Rica
| | - Marietta Flores-Díaz
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, Dulce Nombre Vázquez de Coronado, 11103, Costa Rica
| | - Ann Echeverri-McCandless
- Unidad de Investigación, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José, 10103, Costa Rica
| | - Alexander Parajeles-Vindas
- Servicio de Neurología, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José, 10103, Costa Rica
- Servicio de Neurología, Hospital Clínica Bíblica, San José, 10104, Costa Rica
| |
Collapse
|
5
|
Gharighnia S, Omidi A, Ragerdi Kashani I, Sepand MR, Pour Beiranvand S. Ameliorative effects of acetyl-L-carnitine on corpus callosum and functional recovery in demyelinated mouse model. Int J Neurosci 2024; 134:409-419. [PMID: 35912879 DOI: 10.1080/00207454.2022.2107515] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 10/16/2022]
Abstract
AIM Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disease of the central nervous system. Oxidative stress via distinct pathobiological pathways plays a pivotal role in the formation and persistence of MS lesions. Acetyl-L-carnitine (ALC) facilitates the uptake of acetyl coenzyme-A into the mitochondria by a fatty acid oxidation process. ALC could be a therapeutic antioxidant in the myelin repair process. This study explored the potential neuroprotective effects of ALC in cuprizone (CPZ) intoxicated mice. MATERIALS AND METHODS Thirty male C57BL/6 mice were divided into three groups. The control animals received a normal diet. The CPZ and CPZ + ALC groups were fed with a 0.2% cuprizone diet for 12 weeks. In the CPZ + ALC group, animals received ALC (300 mg/kg/day) from the 10th -12th weeks. Animals were evaluated functionally by beam walking test (BWT) weekly. Eventually, the corpus callosum (CC) was extracted for histological, biochemical, and molecular studies. RESULTS BWT data showed ALC significantly improves balance and gait in the demyelinating mouse model. Histological staining represented ALC effectively increased remyelination in the CC. Biochemical evaluations demonstrated ALC decreased the malondialdehyde level with a parallel increase in the reduced glutathione and catalase activity levels in the CC. Molecular analysis revealed that ALC significantly increased the expression of oligodendrocyte transcription-2 (Olig-2) and Poly lipoproteins (Plp) genes in the CC. CONCLUSIONS ALC improved balance and motor coordination in the demyelinated mouse model. It may be by reducing the levels of free radicals and increasing the expression of Olig-2 and Plp as myelin-related genes.
Collapse
Affiliation(s)
- Sanaz Gharighnia
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ameneh Omidi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Sepand
- Toxicology and Poisoning Research Centre, Department of Toxicology and Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Pour Beiranvand
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
6
|
Yang Z, Marcoci C, Öztürk HK, Giama E, Yenicelik AG, Slanař O, Linington C, Desai R, Smith KJ. Tissue Hypoxia and Associated Innate Immune Factors in Experimental Autoimmune Optic Neuritis. Int J Mol Sci 2024; 25:3077. [PMID: 38474322 DOI: 10.3390/ijms25053077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Visual loss in acute optic neuritis is typically attributed to axonal conduction block due to inflammatory demyelination, but the mechanisms remain unclear. Recent research has highlighted tissue hypoxia as an important cause of neurological deficits and tissue damage in both multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) and, here, we examine whether the optic nerves are hypoxic in experimental optic neuritis induced in Dark Agouti rats. At both the first and second peaks of disease expression, inflamed optic nerves labelled significantly for tissue hypoxia (namely, positive for hypoxia inducible factor-1α (HIF1α) and intravenously administered pimonidazole). Acutely inflamed nerves were also labelled significantly for innate markers of oxidative and nitrative stress and damage, including superoxide, nitric oxide and 3-nitrotyrosine. The density and diameter of capillaries were also increased. We conclude that in acute optic neuritis, the optic nerves are hypoxic and come under oxidative and nitrative stress and damage. Tissue hypoxia can cause mitochondrial failure and thus explains visual loss due to axonal conduction block. Tissue hypoxia can also induce a damaging oxidative and nitrative environment. The findings indicate that treatment to prevent tissue hypoxia in acute optic neuritis may help to restore vision and protect from damaging reactive oxygen and nitrogen species.
Collapse
Affiliation(s)
- Zhiyuan Yang
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Cristina Marcoci
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Hatice Kübra Öztürk
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, 12800 Prague, Czech Republic
| | - Eleni Giama
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Ayse Gertrude Yenicelik
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, 12800 Prague, Czech Republic
| | - Christopher Linington
- School of Infection and Immunity, The Sir Graeme Davies Building, Glasgow G12 8TA, UK
| | - Roshni Desai
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, London WC1N 1PJ, UK
| |
Collapse
|
7
|
Pukoli D, Vécsei L. Smouldering Lesion in MS: Microglia, Lymphocytes and Pathobiochemical Mechanisms. Int J Mol Sci 2023; 24:12631. [PMID: 37628811 PMCID: PMC10454160 DOI: 10.3390/ijms241612631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated, chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS). Immune cell infiltration can lead to permanent activation of macrophages and microglia in the parenchyma, resulting in demyelination and neurodegeneration. Thus, neurodegeneration that begins with acute lymphocytic inflammation may progress to chronic inflammation. This chronic inflammation is thought to underlie the development of so-called smouldering lesions. These lesions evolve from acute inflammatory lesions and are associated with continuous low-grade demyelination and neurodegeneration over many years. Their presence is associated with poor disease prognosis and promotes the transition to progressive MS, which may later manifest clinically as progressive MS when neurodegeneration exceeds the upper limit of functional compensation. In smouldering lesions, in the presence of only moderate inflammatory activity, a toxic environment is clearly identifiable and contributes to the progressive degeneration of neurons, axons, and oligodendrocytes and, thus, to clinical disease progression. In addition to the cells of the immune system, the development of oxidative stress in MS lesions, mitochondrial damage, and hypoxia caused by the resulting energy deficit and iron accumulation are thought to play a role in this process. In addition to classical immune mediators, this chronic toxic environment contains high concentrations of oxidants and iron ions, as well as the excitatory neurotransmitter glutamate. In this review, we will discuss how these pathobiochemical markers and mechanisms, alone or in combination, lead to neuronal, axonal, and glial cell death and ultimately to the process of neuroinflammation and neurodegeneration, and then discuss the concepts and conclusions that emerge from these findings. Understanding the role of these pathobiochemical markers would be important to gain a better insight into the relationship between the clinical classification and the pathomechanism of MS.
Collapse
Affiliation(s)
- Dániel Pukoli
- Department of Neurology, Esztergomi Vaszary Kolos Hospital, 2500 Esztergom, Hungary;
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, University of Szeged, Semmelweis u. 6., H-6725 Szeged, Hungary
- Danube Neuroscience Research Laboratory, ELKH-SZTE Neuroscience Research Group, Eötvös Loránd Research Network, University of Szeged (ELKH-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| |
Collapse
|
8
|
Pietroboni AM, Colombi A, Contarino VE, Russo FML, Conte G, Morabito A, Siggillino S, Carandini T, Fenoglio C, Arighi A, De Riz MA, Arcaro M, Sacchi L, Fumagalli GG, Bianchi AM, Triulzi F, Scarpini E, Galimberti D. Quantitative susceptibility mapping of the normal-appearing white matter as a potential new marker of disability progression in multiple sclerosis. Eur Radiol 2023; 33:5368-5377. [PMID: 36562783 DOI: 10.1007/s00330-022-09338-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/03/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To investigate the normal-appearing white matter (NAWM) susceptibility in a cohort of newly diagnosed multiple sclerosis (MS) patients and to evaluate possible correlations between NAWM susceptibility and disability progression. METHODS Fifty-nine patients with a diagnosis of MS (n = 53) or clinically isolated syndrome (CIS) (n = 6) were recruited and followed up. All participants underwent neurological examination, blood sampling for serum neurofilament light chain (sNfL) level assessment, lumbar puncture for the quantification of cerebrospinal fluid (CSF) β-amyloid1-42 (Aβ) levels, and brain MRI. T2-weighted scans were used to quantify white matter (WM) lesion loads. For each scan, we derived the NAWM volume fraction and the WM lesion volume fraction. Quantitative susceptibility mapping (QSM) of the NAWM was calculated using the susceptibility tensor imaging (STI) suite. Susceptibility maps were computed with the STAR algorithm. RESULTS Primary progressive patients (n = 9) showed a higher mean susceptibility value in the NAWM than relapsing-remitting (n = 44) and CIS (n = 6) (p = 0.01 and p = 0.02). Patients with a higher susceptibility in the NAWM showed increased sNfL concentration (ρ = 0.38, p = 0.004) and lower CSF Aβ levels (ρ = -0.34, p = 0.009). Mean NAWM susceptibility turned out to be a predictor of the expanded disability status scale (EDSS) worsening at follow-up (β = 0.41, t = 2.66, p = 0.01) and of the MS severity scale (MSSS) (β = 0.38, t = 2.43, p = 0.019). CONCLUSIONS QSM in the NAWM seems to predict the EDSS increment over time. This finding might provide evidence on the role of QSM in identifying patients with an increased risk of early disability progression. KEY POINTS • NAWM-QSM is higher in PPMS patients than in RRMS. • NAWM-QSM seems to be a predictor of EDSS worsening over time. • Patients with higher NAWM-QSM show increased sNfL concentration and lower CSF Aβ levels.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy.
| | - Annalisa Colombi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Valeria E Contarino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Francesco Maria Lo Russo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Giorgio Conte
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
- University of Milan, Milan, Italy
| | - Aurelia Morabito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Silvia Siggillino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | | | - Andrea Arighi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Milena A De Riz
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Marina Arcaro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | | | - Giorgio G Fumagalli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | | | - Fabio Triulzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
- University of Milan, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milan, Italy
- University of Milan, Milan, Italy
| |
Collapse
|
9
|
Berek K, Bauer A, Rudzki D, Auer M, Barket R, Zinganell A, Lerch M, Hofer L, Grams A, Poskaite P, Wurth S, Berger T, Di Pauli F, Deisenhammer F, Hegen H, Reindl M. Immune profiling in multiple sclerosis: a single-center study of 65 cytokines, chemokines, and related molecules in cerebrospinal fluid and serum. Front Immunol 2023; 14:1200146. [PMID: 37383229 PMCID: PMC10294231 DOI: 10.3389/fimmu.2023.1200146] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction The understanding of the pathophysiology of multiple sclerosis (MS) has evolved alongside the characterization of cytokines and chemokines in cerebrospinal fluid (CSF) and serum. However, the complex interplay of pro- and anti-inflammatory cytokines and chemokines in different body fluids in people with MS (pwMS) and their association with disease progression is still not well understood and needs further investigation. Therefore, the aim of this study was to profile a total of 65 cytokines, chemokines, and related molecules in paired serum and CSF samples of pwMS at disease onset. Methods Multiplex bead-based assays were performed and baseline routine laboratory diagnostics, magnetic resonance imaging (MRI), and clinical characteristics were assessed. Of 44 participants included, 40 had a relapsing-remitting disease course and four a primary progressive MS. Results There were 29 cytokines and chemokines that were significantly higher in CSF and 15 in serum. Statistically significant associations with moderate effect sizes were found for 34 of 65 analytes with sex, age, CSF, and MRI parameters and disease progression. Discussion In conclusion, this study provides data on the distribution of 65 different cytokines, chemokines, and related molecules in CSF and serum in newly diagnosed pwMS.
Collapse
Affiliation(s)
- Klaus Berek
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Angelika Bauer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Dagmar Rudzki
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Michael Auer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Barket
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anne Zinganell
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Lerch
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Livia Hofer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Astrid Grams
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Paulina Poskaite
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Wurth
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Franziska Di Pauli
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Harald Hegen
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Reindl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
10
|
Treatment with the Olive Secoiridoid Oleacein Protects against the Intestinal Alterations Associated with EAE. Int J Mol Sci 2023; 24:ijms24054977. [PMID: 36902407 PMCID: PMC10003427 DOI: 10.3390/ijms24054977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Multiple sclerosis (MS) is a CNS inflammatory demyelinating disease. Recent investigations highlight the gut-brain axis as a communication network with crucial implications in neurological diseases. Thus, disrupted intestinal integrity allows the translocation of luminal molecules into systemic circulation, promoting systemic/brain immune-inflammatory responses. In both, MS and its preclinical model, the experimental autoimmune encephalomyelitis (EAE) gastrointestinal symptoms including "leaky gut" have been reported. Oleacein (OLE), a phenolic compound from extra virgin olive oil or olive leaves, harbors a wide range of therapeutic properties. Previously, we showed OLE effectiveness preventing motor defects and inflammatory damage of CNS tissues on EAE mice. The current studies examine its potential protective effects on intestinal barrier dysfunction using MOG35-55-induced EAE in C57BL/6 mice. OLE decreased EAE-induced inflammation and oxidative stress in the intestine, preventing tissue injury and permeability alterations. OLE protected from EAE-induced superoxide anion and accumulation of protein and lipid oxidation products in colon, also enhancing its antioxidant capacity. These effects were accompanied by reduced colonic IL-1β and TNFα levels in OLE-treated EAE mice, whereas the immunoregulatory cytokines IL-25 and IL-33 remained unchanged. Moreover, OLE protected the mucin-containing goblet cells in colon and the serum levels of iFABP and sCD14, markers that reflect loss of intestinal epithelial barrier integrity and low-grade systemic inflammation, were significantly reduced. These effects on intestinal permeability did not draw significant differences on the abundance and diversity of gut microbiota. However, OLE induced an EAE-independent raise in the abundance of Akkermansiaceae family. Consistently, using Caco-2 cells as an in vitro model, we confirmed that OLE protected against intestinal barrier dysfunction induced by harmful mediators present in both EAE and MS. This study proves that the protective effect of OLE in EAE also involves normalizing the gut alterations associated to the disease.
Collapse
|
11
|
Shi J, Xiao Y, Zhang N, Jiao M, Tang X, Dai C, Wang C, Xu Y, Tan Z, Gong F, Zheng F. HMGB1 from Astrocytes Promotes EAE by Influencing the Immune Cell Infiltration-Associated Functions of BMECs in Mice. Neurosci Bull 2022; 38:1303-1314. [PMID: 35697993 PMCID: PMC9672173 DOI: 10.1007/s12264-022-00890-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
High mobility group box 1 (HMGB1) has been reported to play an important role in experimental autoimmune encephalomyelitis (EAE). Astrocytes are important components of neurovascular units and tightly appose the endothelial cells of microvessels by their perivascular endfeet and directly regulate the functions of the blood-brain barrier. Astrocytes express more HMGB1 during EAE while the exact roles of astrocytic HMGB1 in EAE have not been well elucidated. Here, using conditional-knockout mice, we found that astrocytic HMGB1 depletion decreased morbidity, delayed the onset time, and reduced the disease score and demyelination of EAE. Meanwhile, there were fewer immune cells, especially pathogenic T cells infiltration in the central nervous system of astrocytic HMGB1 conditional-knockout EAE mice, accompanied by up-regulated expression of the tight-junction protein Claudin5 and down-regulated expression of the cell adhesion molecules ICAM1 and VCAM1 in vivo. In vitro, HMGB1 released from astrocytes decreased Claudin5 while increased ICAM1 and VCAM1 expressed by brain microvascular endothelial cells (BMECs) through TLR4 or RAGE. Taken together, our results demonstrate that HMGB1 derived from astrocytes aggravates EAE by directly influencing the immune cell infiltration-associated functions of BMECs.
Collapse
Affiliation(s)
- Junyu Shi
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yifan Xiao
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Na Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chan Dai
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chenchen Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yong Xu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zheng Tan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, 430030, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, 430030, China.
| |
Collapse
|
12
|
The Efficacy and Safety of Anti-CD20 Antibody Treatments in Relapsing Multiple Sclerosis: A Systematic Review and Network Meta-analysis. CNS Drugs 2022; 36:1155-1170. [PMID: 36245023 DOI: 10.1007/s40263-022-00961-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Several large randomized controlled trials of anti-CD20 antibodies have been successfully conducted for the treatment of relapsing multiple sclerosis. Despite this, there are few systematic comparisons of different anti-CD20 antibodies and a comprehensive evaluation of their efficacy and safety is yet to be carried out. OBJECTIVE The objective of this systematic review and network meta-analysis was to evaluate the efficacy and safety of the three approved anti-CD20 antibodies for the treatment of relapsing multiple sclerosis and to aid clinicians in choosing medications. METHODS MEDLINE, EMBASE, Cochrane Library, and clinicaltrials.gov were all searched for randomized controlled trials conducted to evaluate anti-CD20 antibodies (rituximab, ocrelizumab, ofatumumab) and corresponding controls up to 31 May, 2022. Review Manager 5.3 and R 3.5.2 software were used to assess the data. The risk ratio and mean difference were analyzed and calculated with a random-effects model. RESULTS We pooled 4181 patients from ten randomized controlled trials. Without increasing the risk of adverse events and serious adverse events, anti-CD20 antibodies were superior to the active control group in all efficacy outcomes (both p < 0.005, certainty of evidence, very low to high). For the comparison between anti-CD20 groups, rituximab was found to be able to significantly increase the number of patients free of relapse more effectively than the other two interventions; however, the surface under curve ranking area values for serious adverse events were also the highest (84.8%). At the same time, ocrelizumab and ofatumumab exhibited satisfactory efficacy without showing a worse safety than any other interventions. CONCLUSIONS Overall, anti-CD20 antibody treatment is superior to a corresponding control in efficacy and safety measures and ocrelizumab and ofatumumab may be the most suitable anti-CD20 antibodies for treating relapsing multiple sclerosis. Additional large-scale and high-quality studies are still needed to further explore the safety of these therapies.
Collapse
|
13
|
Naeem AG, El-Naga RN, Michel HE. Nebivolol elicits a neuroprotective effect in the cuprizone model of multiple sclerosis in mice: emphasis on M1/M2 polarization and inhibition of NLRP3 inflammasome activation. Inflammopharmacology 2022; 30:2197-2209. [PMID: 35948811 DOI: 10.1007/s10787-022-01045-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/23/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND AIM Multiple sclerosis (MS) is a demyelinating neurodegenerative inflammatory disease affecting mainly young adults. Microgliosis-derived neuroinflammation represents a key hallmark in MS pathology and progression. Nebivolol (Neb) demonstrated antioxidant, anti-inflammatory and neuroprotective properties in several brain pathologies. This study was conducted to investigate the potential neuroprotective effect of Neb in the cuprizone (Cup) model of MS. METHODS C57Bl/6 mice were fed 0.2% Cup mixed into rodent chow for 5 weeks. Neb (5 and 10 mg/kg/day) was administered by oral gavage during the last 2 weeks. RESULTS Neb prevented Cup-induced weight loss and motor deficits as evidenced by increased latency to fall in the rotarod test and enhanced locomotor activity as compared to Cup-intoxicated mice. Neb reversed Cup-induced demyelination as confirmed by Luxol fast blue staining and myelin basic protein western blotting. Administration of Neb modulated microglial activation status by suppressing M1 markers (Iba-1, CD86, iNOS, NO and TNF-α) and increasing M2 markers (Arg-1 and IL-10) as compared to Cup-fed mice. Furthermore, Neb hindered NLRP3/caspase-1/IL-18 inflammatory cascade and alleviated oxidative stress by reducing lipid peroxidation, as well as increasing catalase and superoxide dismutase activities. CONCLUSION These findings suggest the potential neuroprotective effect of Neb in the Cup-induced model of MS in mice, at least partially by virtue of shifting microglia towards M2 phenotype, mitigation of NLRP3 inflammasome activation and alleviation of oxidative stress.
Collapse
Affiliation(s)
- Antoinette G Naeem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
14
|
Galoppin M, Kari S, Soldati S, Pal A, Rival M, Engelhardt B, Astier A, Thouvenot E. Full spectrum of vitamin D immunomodulation in multiple sclerosis: mechanisms and therapeutic implications. Brain Commun 2022; 4:fcac171. [PMID: 35813882 PMCID: PMC9260308 DOI: 10.1093/braincomms/fcac171] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/03/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Vitamin D deficiency has been associated with the risk of multiple sclerosis, disease activity and progression. Results from in vitro experiments, animal models and analysis of human samples from randomized controlled trials provide comprehensive data illustrating the pleiotropic actions of Vitamin D on the immune system. They globally result in immunomodulation by decreasing differentiation of effector T and B cells while promoting regulatory subsets. Vitamin D also modulates innate immune cells such as macrophages, monocytes and dendritic cells, and acts at the level of the blood–brain barrier reducing immune cell trafficking. Vitamin D exerts additional activity within the central nervous system reducing microglial and astrocytic activation. The immunomodulatory role of Vitamin D detected in animal models of multiple sclerosis has suggested its potential therapeutic use for treating multiple sclerosis. In this review, we focus on recent published data describing the biological effects of Vitamin D in animal models of multiple sclerosis on immune cells, blood–brain barrier function, activation of glial cells and its potential neuroprotective effects. Based on the current knowledge, we also discuss optimization of therapeutic interventions with Vitamin D in patients with multiple sclerosis, as well as new technologies allowing in-depth analysis of immune cell regulations by vitamin D.
Collapse
Affiliation(s)
- Manon Galoppin
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
| | - Saniya Kari
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Arindam Pal
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Manon Rival
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| | | | - Anne Astier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Eric Thouvenot
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| |
Collapse
|
15
|
Treatment of experimental autoimmune encephalomyelitis using AAV gene therapy by blocking T cell costimulatory pathways. Mol Ther Methods Clin Dev 2022; 25:461-475. [PMID: 35615707 PMCID: PMC9118358 DOI: 10.1016/j.omtm.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/25/2022] [Indexed: 11/21/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS), characterized by inflammation and demyelination. Presently, repeated relapses of MS necessitate long-term immune-regulatory therapy. Blocking the CD28-B7 and CD40-CD40L costimulatory pathways is an effective and synergistic method for the prevention and amelioration of clinical symptoms of experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. In this study, to explore the efficacy and safety of MS gene therapy, we used adeno-associated virus (AAV) as a vector to deliver CTLA4-immunoglobulin (Ig) or CD40-Ig on the EAE induced by myelin oligodendrocyte glycoprotein (MOG). Our results showed that a single administration of AAV8-CTLA4-Ig, either alone or with AAV8-CD40-Ig, protected mice from EAE and reversed disease progression. Decreased CD4+ and CD8+ T cell infiltration, inhibition of MOG antibody response, and downregulation of neuroinflammation were observed in mice receiving AAV, suggesting that autoimmunity was suppressed in EAE pathology. Moreover, no hematological or hepatic toxicity was observed in AAV-treated mice. Thus, compared with treatment with recombinant CTLA4-Ig (belatacept), AAV gene therapy could effectively control clinical symptoms and suppress autoimmunity in the long term. In summary, our study provides a potential therapeutic method for blocking T cell costimulation for the treatment of MS via gene therapy.
Collapse
|
16
|
Periventricular magnetisation transfer abnormalities in early multiple sclerosis. Neuroimage Clin 2022; 34:103012. [PMID: 35487133 PMCID: PMC9125781 DOI: 10.1016/j.nicl.2022.103012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 11/30/2022]
Abstract
Periventricular-MTR gradients are present from the earliest stage of MS and become steeper in advanced disease. Lower MTR in periventricular-NAWM was positively associated with reduced cortical-mean-thickness. MTR in periventricular-lesions scaled with cortical-mean-thickness, while non-periventricular lesions were unrelated. These findings suggest a common pathophysiologic mechanism between CSF-adjacent cortical and periventricular areas.
Objective Recent studies suggested that CSF-mediated factors contribute to periventricular (PV) T2-hyperintense lesion formation in multiple sclerosis (MS) and this in turn correlates with cortical damage. We thus investigated if such PV-changes are observable microstructurally in early-MS and if they correlate with cortical damage. Methods We assessed the magnetisation transfer ratio (MTR) in PV normal-appearing white matter (NAWM) and in MS lesions in 44 patients with a clinically isolated syndrome (CIS) suggestive of MS and 73 relapsing-remitting MS (RRMS) patients. Band-wise MTR values were related to cortical mean thickness (CMT) and compared with 49 healthy controls (HCs). For each band, MTR changes were assessed relative to the average MTR values of all HCs. Results Relative to HCs, PV-MTR was significantly reduced up to 2.63% in CIS and 5.37% in RRMS (p < 0.0001). The MTR decreased towards the lateral ventricles with 0.18%/mm in CIS and 0.31%/mm in RRMS patients, relative to HCs. In RRMS, MTR-values adjacent to the ventricle and in PV-lesions correlated positively with CMT and negatively with EDSS. Conclusion PV-MTR gradients are present from the earliest stage of MS, consistent with more pronounced microstructural WM-damage closer to the ventricles. The positive association between reduced CMT and lower MTR in PV-NAWM suggests a common pathophysiologic mechanism. Together, these findings indicate the potential use of multimodal MRI as refined marker for MS-related tissue changes.
Collapse
|
17
|
2-Chlorodeoxyadenosine (Cladribine) preferentially inhibits the biological activity of microglial cells. Int Immunopharmacol 2022; 105:108571. [DOI: 10.1016/j.intimp.2022.108571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 02/05/2023]
|
18
|
Scalabrino G. Newly Identified Deficiencies in the Multiple Sclerosis Central Nervous System and Their Impact on the Remyelination Failure. Biomedicines 2022; 10:biomedicines10040815. [PMID: 35453565 PMCID: PMC9026986 DOI: 10.3390/biomedicines10040815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of multiple sclerosis (MS) remains enigmatic and controversial. Myelin sheaths in the central nervous system (CNS) insulate axons and allow saltatory nerve conduction. MS brings about the destruction of myelin sheaths and the myelin-producing oligodendrocytes (ODCs). The conundrum of remyelination failure is, therefore, crucial in MS. In this review, the roles of epidermal growth factor (EGF), normal prions, and cobalamin in CNS myelinogenesis are briefly summarized. Thereafter, some findings of other authors and ourselves on MS and MS-like models are recapitulated, because they have shown that: (a) EGF is significantly decreased in the CNS of living or deceased MS patients; (b) its repeated administration to mice in various MS-models prevents demyelination and inflammatory reaction; (c) as was the case for EGF, normal prion levels are decreased in the MS CNS, with a strong correspondence between liquid and tissue levels; and (d) MS cobalamin levels are increased in the cerebrospinal fluid, but decreased in the spinal cord. In fact, no remyelination can occur in MS if these molecules (essential for any form of CNS myelination) are lacking. Lastly, other non-immunological MS abnormalities are reviewed. Together, these results have led to a critical reassessment of MS pathogenesis, partly because EGF has little or no role in immunology.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| |
Collapse
|
19
|
Pathophysiology of neurodegenerative diseases: An interplay among axonal transport failure, oxidative stress, and inflammation? Semin Immunol 2022; 59:101628. [PMID: 35779975 PMCID: PMC9807734 DOI: 10.1016/j.smim.2022.101628] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Neurodegenerative diseases (NDs) are heterogeneous neurological disorders characterized by a progressive loss of selected neuronal populations. A significant risk factor for most NDs is aging. Considering the constant increase in life expectancy, NDs represent a global public health burden. Axonal transport (AT) is a central cellular process underlying the generation and maintenance of neuronal architecture and connectivity. Deficits in AT appear to be a common thread for most, if not all, NDs. Neuroinflammation has been notoriously difficult to define in relation to NDs. Inflammation is a complex multifactorial process in the CNS, which varies depending on the disease stage. Several lines of evidence suggest that AT defect, axonopathy and neuroinflammation are tightly interlaced. However, whether these impairments play a causative role in NDs or are merely a downstream effect of neuronal degeneration remains unsettled. We still lack reliable information on the temporal relationship between these pathogenic mechanisms, although several findings suggest that they may occur early during ND pathophysiology. This article will review the latest evidence emerging on whether the interplay between AT perturbations and some aspects of CNS inflammation can participate in ND etiology, analyze their potential as therapeutic targets, and the urge to identify early surrogate biomarkers.
Collapse
|
20
|
The Efficacy of Fecal Microbiota Transplantation in Experimental Autoimmune Encephalomyelitis: Transcriptome and Gut Microbiota Profiling. J Immunol Res 2021; 2021:4400428. [PMID: 34938813 PMCID: PMC8687821 DOI: 10.1155/2021/4400428] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Objective To study the protective effect of fecal microbiota transplantation (FMT) on experimental autoimmune encephalomyelitis (EAE) and reveal its potential intestinal microflora-dependent mechanism through analyses of the intestinal microbiota and spinal cord transcriptome in mice. Method We measured the severity of disease by clinical EAE scores and H&E staining. Gut microbiota alteration in the gut and differentially expressed genes (DEGs) in the spinal cord were analyzed through 16S rRNA and transcriptome sequencing. Finally, we analyzed associations between the relative abundance of intestinal microbiota constituents and DEGs. Results We observed that clinical EAE scores were lower in the EAE+FMT group than in the EAE group. Meanwhile, mice in the EAE+FMT group also had a lower number of infiltrating cells. The results of 16S rRNA sequence analysis showed that FMT increased the relative abundance of Firmicutes and Proteobacteria and reduced the abundance of Bacteroides and Actinobacteria. Meanwhile, FMT could modulate gut microbiota balance, especially via increasing the relative abundance of g_Adlercreutzia, g_Sutterella, g_Prevotella_9, and g_Tyzzerella_3 and decreasing the relative abundance of g_Turicibacter. Next, we analyzed the transcriptome of mouse spinal cord tissue and found that 1476 genes were differentially expressed between the EAE and FMT groups. The analysis of these genes showed that FMT mainly participated in the inflammatory response. Correlation analysis between gut microbes and transcriptome revealed that the relative abundance of Adlercreutzia was correlated with the expression of inflammation-related genes negatively, including Casp6, IL1RL2 (IL-36R), IL-17RA, TNF, CCL3, CCR5, and CCL8, and correlated with the expression of neuroprotection-related genes positively, including Snap25, Edil3, Nrn1, Cpeb3, and Gpr37. Conclusion Altogether, FMT may selectively regulate gene expression to improve inflammation and maintain the stability of the intestinal environment in a gut microbiota-dependent manner.
Collapse
|
21
|
Reyes-Mata PM, Rojas-Mayorquín AE, Carrera-Quintanar L, González-Castillo C, Mireles-Ramírez MA, Guerrero-García JDJ, Ortuño-Sahagún D. Pleiotrophin serum level is increased in Relapsing-Remitting Multiple Sclerosis and correlates with sex, BMI and treatment. Arch Med Res 2021; 53:59-68. [PMID: 34247888 DOI: 10.1016/j.arcmed.2021.06.005] [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: 01/11/2021] [Revised: 05/02/2021] [Accepted: 06/15/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Multiple Sclerosis (MS) is an immune-mediated demyelinating disease mainly affecting the Central Nervous System (CNS). 80% of MS patients present the Relapsing-Remitting form (RRMS). Pleiotrophin (PTN), a cytokine previously associated with other autoimmune and neurological diseases, could play a role in the pathophysiology of RRMS due to its neuro and immunomodulatory effect. However, PTN has never been explored in RRMS patients. AIM OF THE STUDY To determine PTN serum levels in patients with RRMS, treated with Glatiramer acetate (GA) or Interferon-beta (IFN-β), as well as in non-treated patients and healthy controls as a first attempt to explore PTN in RRMS. METHODS PTN serum levels were quantified by ELISA in 57 patients and 18 controls. RESULTS We demonstrated that PTN serum levels are significantly higher in RRMS patients. In IFN-β treated patients alone, PTN correlated positively with time of disease evolution and time of IFN-β use and correlated negatively with the MS severity score (MSSS). When comparing groups according to weight status, we observed that PTN is statistically increased in overweight female patients and that weight does not affect male patients. The Area Under the Curve (AUC) of the Receiver Operating Characteristic (ROC) curve analysis was higher for males compared to females. CONCLUSION PTN serum level is higher in RRMS patients and that is associated with sex, BMI and IFN-β treatment. Therefore, we propose that PTN could be playing a role in MS. Further studies must be performed to identify the exact role of PTN in this pathology.
Collapse
Affiliation(s)
- Paulina María Reyes-Mata
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | - Argelia Esperanza Rojas-Mayorquín
- Departamento de Ciencias Ambientales, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Jalisco, México
| | - Lucrecia Carrera-Quintanar
- Laboratorio de Ciencias de los Alimentos, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | | | - Mario Alberto Mireles-Ramírez
- Unidad Médica de Alta Especialidad, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano de Seguro Social, Guadalajara, Jalisco, México
| | - José de Jesús Guerrero-García
- Banco de Sangre Central, Unidad Médica de Alta Especialidad, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano de Seguro Social, Guadalajara, Jalisco, México
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México.
| |
Collapse
|
22
|
Cree BAC, Arnold DL, Chataway J, Chitnis T, Fox RJ, Pozo Ramajo A, Murphy N, Lassmann H. Secondary Progressive Multiple Sclerosis: New Insights. Neurology 2021; 97:378-388. [PMID: 34088878 PMCID: PMC8397587 DOI: 10.1212/wnl.0000000000012323] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/13/2021] [Indexed: 01/01/2023] Open
Abstract
In most cases, multiple sclerosis (MS) begins with a relapsing-remitting course followed by insidious disability worsening that is independent from clinically apparent relapses and is termed secondary progressive MS (SMPS). Major differences exist between relapsing-remitting MS (RRMS) and SPMS, especially regarding therapeutic response to treatment. This review provides an overview of the pathology, differentiation, and challenges in the diagnosis and treatment of SPMS. We emphasize the criticality of conversion from a relapsing-remitting to a secondary progressive disease course not only because such conversion is evidence of disability progression, but also because, until recently, treatments that effectively reduced disability progression in relapsing MS were not proven to be effective in SPMS. Clear clinical, imaging, immunologic, or pathologic criteria marking the transition from RRMS to SPMS have not yet been established. Early identification of SPMS will require tools that, together with the use of appropriate treatments, may result in better long-term outcomes for the population of patients with SPMS.
Collapse
Affiliation(s)
- Bruce A C Cree
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria.
| | - Douglas L Arnold
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria
| | - Jeremy Chataway
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria
| | - Tanuja Chitnis
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria
| | - Robert J Fox
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria
| | - Angela Pozo Ramajo
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria
| | - Niamh Murphy
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria
| | - Hans Lassmann
- From the UCSF Weill Institute for Neurosciences, Department of Neurology (B.A.C.C.), University of California San Francisco; NeuroRx Research (D.L.A.), Montreal; Brain Imaging Centre (D.L.A.), Montreal Neurological Institute, McGill University, Canada; Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation (J.C.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London; National Institute for Health Research (J.C.), University College London Hospitals, Biomedical Research Centre, UK; Brigham Multiple Sclerosis Center (T.C.), Brigham and Women's Hospital, Boston, MA; Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute (R.J.F.), Cleveland Clinic, OH; Oxford PharmaGenesis (A.P.R.), UK; Novartis Pharma AG (N.M.), Basel, Switzerland; and Center for Brain Research (H.L.), Medical University of Vienna, Austria
| |
Collapse
|
23
|
Zhang N, Liu C, Zhang R, Jin L, Yin X, Zheng X, Siebert HC, Li Y, Wang Z, Loers G, Petridis AK. Amelioration of clinical course and demyelination in the cuprizone mouse model in relation to ketogenic diet. Food Funct 2021; 11:5647-5663. [PMID: 32539054 DOI: 10.1039/c9fo02944c] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ketogenic diet (KD) is defined as a high-fat, low-carbohydrate diet with appropriate amounts of protein, which has broad neuroprotective effects. However, the mechanisms of ameliorating the demyelination and of the neuroprotective effects of KD have not yet been completely elucidated. Therefore, the present study investigated the protection mechanism of KD treatment in the cuprizone (bis-cyclohexanone oxalydihydrazone, CPZ)-induced demyelination mice model, with special emphasis on neuroinflammation. After the KD treatment, an increased ketone body level in the blood of mice was detected, and a significant increase in the distance traveled within the central area was observed in the open field test, which reflected the increased exploration and decreased anxiety of mice that received CPZ. The results of Luxol fast blue and myelin basic protein (MBP) immunohistochemistry staining for the evaluation of the myelin content within the corpus callosum revealed a noticeable increase in the number of myelinated fibers and myelin score after KD administration in these animals. Concomitant, the protein expressions of glial fibrillary acidic protein (GFAP, an astrocyte marker), ionized calcium-binding adaptor molecule 1 (Iba-1, a microglial marker), CD68 (an activated microglia marker) and CD16/32 (a M1 microglial marker) were down-regulated, while the expression of oligodendrocyte lineage transcription factor 2 (OLIG2, an oligodendrocyte precursor cells marker) was up-regulated by the KD treatment. In addition, the KD treatment not only reduced the level of the C-X-C motif chemokine 10 (CXCL10), which is correlated to the recruitment of activated microglia, but also inhibited the production of proinflammatory cytokines, including interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α), which are closely correlated to the M1 phenotype microglia. It is noteworthy, that the expression levels of histone deacetylase 3 (HADC3) and nod-like receptor pyrin domain containing 3 (NLRP3) significantly decreased after KD administration. In conclusion, these data demonstrate that KD decreased the reactive astrocytes and activated the microglia in the corpus callosum, and that KD inhibited the HADC3 and NLRP3 inflammasome signaling pathway in CPZ-treated mice. This suggests that the inhibition of the HADC3 and NLRP3 signaling pathway may be a novel mechanism by which KD exerts its protective actions for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Chunhong Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Li Jin
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Xiaohan Yin
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Xuexing Zheng
- Department of Virology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Hans-Christian Siebert
- RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Schauenburgerstr. 116, 24118 Kiel, Germany
| | - Yubao Li
- College of agriculture, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Falkenried 94, 20251 Hamburg, Germany
| | - Athanasios K Petridis
- Neurosurgical Department, Heinrich Heine University of Düsseldorf, Moorenstraße 5, 40255 Düsseldorf, Germany
| |
Collapse
|
24
|
Sherafat A, Pfeiffer F, Reiss AM, Wood WM, Nishiyama A. Microglial neuropilin-1 promotes oligodendrocyte expansion during development and remyelination by trans-activating platelet-derived growth factor receptor. Nat Commun 2021; 12:2265. [PMID: 33859199 PMCID: PMC8050320 DOI: 10.1038/s41467-021-22532-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/08/2021] [Indexed: 02/02/2023] Open
Abstract
Nerve-glia (NG2) glia or oligodendrocyte precursor cells (OPCs) are distributed throughout the gray and white matter and generate myelinating cells. OPCs in white matter proliferate more than those in gray matter in response to platelet-derived growth factor AA (PDGF AA), despite similar levels of its alpha receptor (PDGFRα) on their surface. Here we show that the type 1 integral membrane protein neuropilin-1 (Nrp1) is expressed not on OPCs but on amoeboid and activated microglia in white but not gray matter in an age- and activity-dependent manner. Microglia-specific deletion of Nrp1 compromised developmental OPC proliferation in white matter as well as OPC expansion and subsequent myelin repair after acute demyelination. Exogenous Nrp1 increased PDGF AA-induced OPC proliferation and PDGFRα phosphorylation on dissociated OPCs, most prominently in the presence of suboptimum concentrations of PDGF AA. These findings uncover a mechanism of regulating oligodendrocyte lineage cell density that involves trans-activation of PDGFRα on OPCs via Nrp1 expressed by adjacent microglia.
Collapse
Affiliation(s)
- Amin Sherafat
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Friederike Pfeiffer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Alexander M Reiss
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - William M Wood
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA.
- The Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
25
|
Simkins TJ, Duncan GJ, Bourdette D. Chronic Demyelination and Axonal Degeneration in Multiple Sclerosis: Pathogenesis and Therapeutic Implications. Curr Neurol Neurosci Rep 2021; 21:26. [PMID: 33835275 DOI: 10.1007/s11910-021-01110-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system (CNS). Inflammatory attacks in MS lead to both demyelination and axonal damage. However, due to incomplete remyelination most MS lesions remain chronically demyelinated. In parallel, there is axonal degeneration in the CNS of MS patients, contributing to progressive disability. There are currently no approved therapies that adequately restore myelin or protect axons from degeneration. In this review, we will discuss the pathophysiology of axonal loss and chronic demyelination in MS and how understanding this pathophysiology is leading to the development of new MS therapeutics. RECENT FINDINGS Ongoing research into the function of oligodendrocytes and myelin has revealed the importance of their relationship with neuronal health. Demyelination in MS leads to a number of pathophysiologic changes contributing to axonal generation. Among these are mitochondrial dysfunction, persistent neuroinflammation, and the effects of reactive oxygen and nitrogen species. With this information, we review currently approved and investigational therapies designed to restore lost or damaged myelin and protect against neuronal degeneration. The development of therapies to restore lost myelin and protect neurons is a promising avenue of investigation for the benefit of patients with MS.
Collapse
Affiliation(s)
- Tyrell J Simkins
- Department of Neurology, Oregon Health and Science University, 3181S W Sam Jackson Rd L226, Portland, OR, 97239, USA. .,Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA. .,Department of Neurology, Portland VA Medical Center, Portland, OR, USA.
| | - Greg J Duncan
- Department of Neurology, Oregon Health and Science University, 3181S W Sam Jackson Rd L226, Portland, OR, 97239, USA.,Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| | - Dennis Bourdette
- Department of Neurology, Oregon Health and Science University, 3181S W Sam Jackson Rd L226, Portland, OR, 97239, USA.,Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
26
|
Tian H, Cheng Y, Zhang Y, Bai X, Jiang Y, Li J, Fan S, Ding H. 18β-Glycyrrhetinic acid alleviates demyelination by modulating the microglial M1/M2 phenotype in a mouse model of cuprizone-induced demyelination. Neurosci Lett 2021; 755:135871. [PMID: 33812928 DOI: 10.1016/j.neulet.2021.135871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
This research aimed to examine the nutritious supplementary function of 18β-Glycyrrhetinic acid (18β-GA) in moderating the myelin sheath destruction and behavioral impairments observed in the cuprizone model of demyelination. Mice were fed daily on food containing cuprizone (0.3 %) and given doses of 18β-GA (5 or 1 mg/kg) for a period of five weeks. The groups treated with 18β-GA exhibited improvements in exploratory behavior, locomotive activity, and weight. As assessed using luxol-fast blue and myelin basic protein (MBP) staining, which were used to detect demyelination in the brain, 18β-GA both reduced and prevented instances of cuprizone-induced demyelinating lesions; treatment with 18β-GA also caused the MBP level in the corpus callosum to increase. Furthermore, alongside these positive results following 18β-GA treatment, microglial polarisation was also observed to shift towards the beneficial M2 phenotype. The results of this research thus indicate the potential clinical application of 18β-GA for the prevention of myelin damage and behavioral dysfunction.
Collapse
Affiliation(s)
- Hui Tian
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Yahong Cheng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Yiyuan Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Xinying Bai
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Yuan Jiang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Jinjin Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Shiqi Fan
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Hong Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, PR China.
| |
Collapse
|
27
|
Charvet B, Pierquin J, Brunel J, Gorter R, Quétard C, Horvat B, Amor S, Portoukalian J, Perron H. Human Endogenous Retrovirus Type W Envelope from Multiple Sclerosis Demyelinating Lesions Shows Unique Solubility and Antigenic Characteristics. Virol Sin 2021; 36:1006-1026. [PMID: 33770381 PMCID: PMC8558138 DOI: 10.1007/s12250-021-00372-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
In multiple sclerosis (MS), human endogenous retrovirus W family (HERV-W) envelope protein, pHERV-W ENV, limits remyelination and induces microglia-mediated neurodegeneration. To better understand its role, we examined the soluble pHERV-W antigen from MS brain lesions detected by specific antibodies. Physico-chemical and antigenic characteristics confirmed differences between pHERV-W ENV and syncytin-1. pHERV-W ENV monomers and trimers remained associated with membranes, while hexamers self-assembled from monomers into a soluble macrostructure involving sulfatides in MS brain. Extracellular hexamers are stabilized by internal hydrophobic bonds and external hydrophilic moieties. HERV-W studies in MS also suggest that this diffusible antigen may correspond to a previously described high-molecular-weight neurotoxic factor secreted by MS B-cells and thus represents a major agonist in MS pathogenesis. Adapted methods are now needed to identify encoding HERV provirus(es) in affected cells DNA. The properties and origin of MS brain pHERV-W ENV soluble antigen will allow a better understanding of the role of HERVs in MS pathogenesis. The present results anyhow pave the way to an accurate detection of the different forms of pHERV-W ENV antigen with appropriate conditions that remained unseen until now.
Collapse
Affiliation(s)
- Benjamin Charvet
- GeNeuro Innovation, Lyon, 69008, France. .,CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, University of Lyon, ENS Lyon, France. .,Université Claude Bernard Lyon 1, Lyon, 69000, France.
| | | | - Joanna Brunel
- GeNeuro Innovation, Lyon, 69008, France.,CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, University of Lyon, ENS Lyon, France.,Université Claude Bernard Lyon 1, Lyon, 69000, France
| | - Rianne Gorter
- Department of Pathology, Amsterdam UMC, Location VUMC, 1007 MB, Amsterdam, The Netherlands
| | | | - Branka Horvat
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, University of Lyon, ENS Lyon, France.,Université Claude Bernard Lyon 1, Lyon, 69000, France
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, Location VUMC, 1007 MB, Amsterdam, The Netherlands.,Centre for Neuroscience and Trauma, Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | | | - Hervé Perron
- GeNeuro Innovation, Lyon, 69008, France. .,Université Claude Bernard Lyon 1, Lyon, 69000, France.
| |
Collapse
|
28
|
Golan M, Krivitsky A, Mausner-Fainberg K, Benhamou M, Vigiser I, Regev K, Kolb H, Karni A. Increased Expression of Ephrins on Immune Cells of Patients with Relapsing Remitting Multiple Sclerosis Affects Oligodendrocyte Differentiation. Int J Mol Sci 2021; 22:ijms22042182. [PMID: 33671716 PMCID: PMC7927032 DOI: 10.3390/ijms22042182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/02/2022] Open
Abstract
The effect of the inflammatory response on regenerative processes in the brain is complex. This complexity is even greater when the cause of the tissue damage is an autoimmune response. Multiple sclerosis (MS) is an immune-mediated disease in which demyelination foci are formed in the central nervous system. The degree of repair through oligodendrocyte regeneration and remyelination is insufficient. Ephrins are membrane-bound ligands activating tyrosine kinase signaling proteins that are known to have an inhibitory effect on oligodendrocyte regeneration. In this study, we examined the expression of ephrins on immune cells of 43 patients with relapsing-remitting (RR) MS compared to 27 matched healthy controls (HC). We found an increased expression of ephrin-A2, -A3 and -B3, especially on T cell subpopulations. We also showed overexpression of ephrins on immune cells of patients with RR-MS that increases the forward signaling pathway and that expression of ephrins on immune cells has an inhibitory effect on the differentiation of oligodendrocyte precursor cells (OPCs) in vitro. Our study findings support the concept that the immune activity of T cells in patients with RR-MS has an inhibitory effect on the differentiation capacity of OPCs through the expression and forward signaling of ephrins.
Collapse
Affiliation(s)
- Maya Golan
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Avivit Krivitsky
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Karin Mausner-Fainberg
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Moshe Benhamou
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ifat Vigiser
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Keren Regev
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Hadar Kolb
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Arnon Karni
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
29
|
das Neves SP, Sousa JC, Sousa N, Cerqueira JJ, Marques F. Altered astrocytic function in experimental neuroinflammation and multiple sclerosis. Glia 2020; 69:1341-1368. [PMID: 33247866 DOI: 10.1002/glia.23940] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that affects about 2.5 million people worldwide. In MS, the patients' immune system starts to attack the myelin sheath, leading to demyelination, neurodegeneration, and, ultimately, loss of vital neurological functions such as walking. There is currently no cure for MS and the available treatments only slow the initial phases of the disease. The later-disease mechanisms are poorly understood and do not directly correlate with the activity of immune system cells, the main target of the available treatments. Instead, evidence suggests that disease progression and disability are better correlated with the maintenance of a persistent low-grade inflammation inside the CNS, driven by local glial cells, like astrocytes and microglia. Depending on the context, astrocytes can (a) exacerbate inflammation or (b) promote immunosuppression and tissue repair. In this review, we will address the present knowledge that exists regarding the role of astrocytes in MS and experimental animal models of the disease.
Collapse
Affiliation(s)
- Sofia Pereira das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - João Carlos Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal.,Clinical Academic Center, Braga, Portugal
| | - João José Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal.,Clinical Academic Center, Braga, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| |
Collapse
|
30
|
Localised Grey Matter Atrophy in Multiple Sclerosis and Clinically Isolated Syndrome-A Coordinate-Based Meta-Analysis, Meta-Analysis of Networks, and Meta-Regression of Voxel-Based Morphometry Studies. Brain Sci 2020; 10:brainsci10110798. [PMID: 33143012 PMCID: PMC7693631 DOI: 10.3390/brainsci10110798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 01/04/2023] Open
Abstract
Background: Atrophy of grey matter (GM) is observed in the earliest stages of multiple sclerosis (MS) and is associated with cognitive decline and physical disability. Localised GM atrophy in MS can be explored and better understood using magnetic resonance imaging and voxel-based morphometry (VBM). However, results are difficult to interpret due to methodological differences between studies. Methods: Coordinate-based analysis is a way to find the reliably observable results across multiple independent VBM studies. This work uses coordinate-based meta-analysis, meta-analysis of networks, and meta-regression to summarise the evidence from voxel-based morphometry of regional GM hanges in patients with MS and clinically isolated syndrome (CIS), and whether these measured changes are relatable to clinical features. Results: Thirty-four published articles reporting forty-four independent experiments using VBM for the assessment of GM atrophy between MS or CIS patients and healthy controls were identified. Analysis identified eight clusters of consistent cross-study reporting of localised GM atrophy involving both cortical and subcortical regions. Meta-network analysis identified a network-like pattern indicating that GM loss occurs with some symmetry between hemispheres. Meta-regression analysis indicates a relationship between disease duration or age and the magnitude of reported statistical effect in some deep GM structures. Conclusions: These results suggest consistency in MRI-detectible regional GM loss across multiple MS studies, and the estimated effect sizes and symmetries can help design prospective studies to test specific hypotheses.
Collapse
|
31
|
Hauptmann J, Johann L, Marini F, Kitic M, Colombo E, Mufazalov IA, Krueger M, Karram K, Moos S, Wanke F, Kurschus FC, Klein M, Cardoso S, Strauß J, Bolisetty S, Lühder F, Schwaninger M, Binder H, Bechman I, Bopp T, Agarwal A, Soares MP, Regen T, Waisman A. Interleukin-1 promotes autoimmune neuroinflammation by suppressing endothelial heme oxygenase-1 at the blood-brain barrier. Acta Neuropathol 2020; 140:549-567. [PMID: 32651669 PMCID: PMC7498485 DOI: 10.1007/s00401-020-02187-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/05/2020] [Accepted: 06/26/2020] [Indexed: 12/18/2022]
Abstract
The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood-brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Judith Hauptmann
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lisa Johann
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Federico Marini
- Center of Thrombosis and Hemostasis Mainz (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Maja Kitic
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisa Colombo
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ilgiz A Mufazalov
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Martin Krueger
- Anatomical Institute, University of Leipzig, Leipzig, Germany
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sonja Moos
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Dermatology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area Roche Innovation Center, Basel, Switzerland
| | - Florian C Kurschus
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Dermatology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Judith Strauß
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Subhashini Bolisetty
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Harald Binder
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Ingo Bechman
- Anatomical Institute, University of Leipzig, Leipzig, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anupam Agarwal
- Nephrology Research and Training Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
32
|
Vitamin B1 Intake in Multiple Sclerosis Patients and its Impact on Depression Presence: A Pilot Study. Nutrients 2020; 12:nu12092655. [PMID: 32878159 PMCID: PMC7551277 DOI: 10.3390/nu12092655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Vitamin B1, or thiamine, is one of the most relevant vitamins in obtaining energy for the nervous system. Thiamine deficiency or lack of activity causes neurological manifestations, especially symptoms of depression, intrinsic to multiple sclerosis (MS) and related to its pathogenesis. On this basis, the aim of this study was to determine the possible relationship between the nutritional habits of patients with MS and the presence of depression. Therefore, a cross-sectional and observational descriptive study was conducted. An analysis of dietary habits and vitamin B1 consumption in a Spanish population of 51 MS patients was performed by recording the frequency of food consumption. Results showed a vitamin B1 intake within the established range, mainly provided by the consumption of ultra-processed products such as cold meats or pastries, and a total carbohydrate consumption lower than recommended, which stands out for its high content of simple carbohydrates deriving from processed foods such as dairy desserts, juice, snacks, pastries, chocolate bars, soft drinks and fermented alcohol. In addition, a significant negative correlation between depression and the intake of thiamine and total carbohydrates was observed. These findings could explain the influence of MS patients' eating habits, and consequently vitamin B1 activity, on depression levels.
Collapse
|
33
|
Huss A, Otto M, Senel M, Ludolph AC, Abdelhak A, Tumani H. A Score Based on NfL and Glial Markers May Differentiate Between Relapsing-Remitting and Progressive MS Course. Front Neurol 2020; 11:608. [PMID: 32765393 PMCID: PMC7378743 DOI: 10.3389/fneur.2020.00608] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 05/25/2020] [Indexed: 01/22/2023] Open
Abstract
Background: The diagnostic use of biomarkers in body fluids of multiple sclerosis (MS) patients allows the monitoring of different pathophysiological aspects of the disease. We previously reported elevated cerebrospinal fluid (CSF) and serum levels of glial fibrillary acidic protein (GFAP) but not neurofilament light chain (NfL) in progressive (PMS) compared to relapsing–remitting MS (RRMS) patients. Objectives: We analyzed the glial marker chitinase-3-like protein 1 (CHI3L1) in the CSF and serum of PMS and RRMS patients. To capture the extent of glial processes in relation to axonal damage in each individual patient, we established a score based on CHI3L1, GFAP, and NfL and compared this score between RRMS and PMS patients and its association with the extended disability status scale (EDSS). Methods: For this retrospective study, we included 86 MS patients (47 RRMS and 39 PMS) and 20 patients with other non-inflammatory neurological diseases (OND) as controls. NfL and GFAP levels were determined by the single-molecule array (Simoa). CHI3L1 levels were measured with classical enzyme-linked immunosorbent assay. A score was calculated based on glial to axonal markers (CHI3L1*GFAP/NfL, referred to as “Glia score”). Results: CHI3L1 showed higher CSF levels in PMS vs. RRMS and controls (p < 0.001 and p < 0.0001, respectively), RMS vs. controls (p < 0.01), and higher serum levels for PMS vs. RRMS (p < 0.05). The Glia score was higher in the CSF of PMS compared to RRMS patients (p < 0.0001) and in the serum of PMS patients compared to RRMS (p < 0.01). Furthermore, the Glia score and CHI3L1 in serum but not in CSF correlated with the disability as determined by EDSS in the PMS group but not in the RRMS group (Spearman ρ = 0.46 and 0.45, p = 0.003 and 0.004, respectively). Discussion: Our data indicate the involvement of glial mechanisms during the pathogenesis of PMS. Moreover, a calculated score may help to differentiate between PMS and RMS in the CSF and monitor disease progression in the serum of PMS patients.
Collapse
Affiliation(s)
- André Huss
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Markus Otto
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Makbule Senel
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Albert C Ludolph
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Ahmed Abdelhak
- Department of Neurology, University Hospital of Ulm, Ulm, Germany.,Department of Neurology and Stroke, University Hospital of Tuebingen, Tübingen, Germany
| | - Hayrettin Tumani
- Department of Neurology, University Hospital of Ulm, Ulm, Germany.,Speciality Clinic of Neurology Dietenbronn, Schwendi, Germany
| |
Collapse
|
34
|
Guerrero-García J. The role of astrocytes in multiple sclerosis pathogenesis. NEUROLOGÍA (ENGLISH EDITION) 2020. [DOI: 10.1016/j.nrleng.2017.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
35
|
Szilagyi GT, Nawrocki AM, Eros K, Schmidt J, Fekete K, Elkjaer ML, Hyrlov KH, Larsen MR, Illes Z, Gallyas F. Proteomic changes during experimental de- and remyelination in the corpus callosum. PLoS One 2020; 15:e0230249. [PMID: 32272486 PMCID: PMC7145428 DOI: 10.1371/journal.pone.0230249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 02/25/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND In the cuprizone model of multiple sclerosis, de- and remyelination can be studied without major interference from the adaptive immune responses. Since previous proteomic studies did not focus on the corpus callosum, where cuprizone causes the most pronounced demyelination, we performed a bottom up proteomic analysis on this brain region. METHODS Eight week-old mice treated with 0.2% cuprizone, for 4 weeks and controls (C) were sacrificed after termination of the treatment (4wD), and 2 (2dR) or 14 (2wR) days later. Homogenates of dissected corpus callosum were analysed by quantitative proteomics. For data processing, clustering, gene ontology analysis, and regulatory network prediction, we used Perseus, PANTHER and Ingenuity Pathway Analysis softwares, respectively. RESULTS We identified 4886 unmodified, single- or multi phosphorylated and/or gycosylated (PTM) proteins. Out of them, 191 proteins were differentially regulated in at least one experimental group. We found 57 proteins specific for demyelination, 27 for early- and 57 for late remyelinationwhile 36 proteins were affected in two, and 23 proteins in all three groups. Phosphorylation represented 92% of the post translational modifications among differentially regulated modified (PTM) proteins with decreased level, while it was only 30% of the PTM proteins with increased level. Gene ontology analysis could not classify the demyelination specific proteins into any biological process category, while allocated the remyelination specific ones to nervous system development and myelination as the most specific subcategory. We also identified a protein network in experimental remyelination, and the gene orthologues of the network were differentially expressed in remyelinating multiple sclerosis brain lesions consistent with an early remyelination pattern. CONCLUSION Proteomic analysis seems more informative for remyelination than demyelination in the cuprizone model.
Collapse
Affiliation(s)
- Gabor T. Szilagyi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Arkadiusz M. Nawrocki
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Krisztian Eros
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
- Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Janos Schmidt
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Maria L. Elkjaer
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Kirsten H. Hyrlov
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Martin R. Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, BRIDGE University of Southern Denmark, Odense, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
- Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail:
| |
Collapse
|
36
|
Amini R, Karampoor S, Zahednasab H, Keyvani H, Gheiasian M, Jalilian FA. Serum levels of matrix metalloproteinase-2, -9, and vitamin D in patients with multiple sclerosis with or without herpesvirus-6 seropositivity. Braz J Infect Dis 2020; 24:144-149. [PMID: 32243867 PMCID: PMC9392051 DOI: 10.1016/j.bjid.2020.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 02/10/2020] [Accepted: 02/24/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Razieh Amini
- Hamadan University of Medical Sciences, Faculty of Medicine, Department of Molecular Medicine and Genetics, Hamadan, Iran
| | - Sajad Karampoor
- Iran University of Medical Sciences, School of Medicine, Department of Virology, Tehran, Iran.
| | - Hamid Zahednasab
- University of Tehran, Institute of Biochemistry and Biophysics, Tehran, Iran
| | - Hossein Keyvani
- Iran University of Medical Sciences, School of Medicine, Department of Virology, Tehran, Iran
| | - Masoud Gheiasian
- Hamadan University of Medical Sciences, Faculty of Medicine, Department of Neurology, Hamadan, Iran
| | - Farid Azizi Jalilian
- Hamadan University of Medical Sciences, Faculty of Medicine Hamadan, Department of Medical Virology, Hamadan, Iran.
| |
Collapse
|
37
|
Dixon GA, Pérez CA. Multiple Sclerosis and the Choroid Plexus: Emerging Concepts of Disease Immunopathophysiology. Pediatr Neurol 2020; 103:65-75. [PMID: 31780202 DOI: 10.1016/j.pediatrneurol.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/11/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The coexistence of multiple sclerosis and intracranial neoplasms is very rare, and whether this occurrence can be explained by a causal relationship or by coincidence remains a matter of debate. Possible roles of the choroid plexus as a site of tumor cell invasion and lymphocyte infiltration into the central nervous system have been hypothesized in recent studies. METHODS We describe a 13-year-old boy with concurrent multiple sclerosis and choroid plexus papilloma, then review the published literature with a focus on the pathophysiologic mechanisms of neuroinflammation in multiple sclerosis and the potential role of the choroid plexus in this process. RESULTS A growing body of evidence suggests that both physical and functional dysregulation of the choroid plexus may be a common mechanism underlying the pathophysiology of central nervous system inflammation. CONCLUSIONS In multiple sclerosis, the choroid plexus could act as a gateway for lymphocyte entry from the peripheral blood into the central nervous system at its earlier stages. However, future studies are needed to identify whether structural alterations of the choroid plexus play a role in the pathophysiology of multiple sclerosis and to provide suitable models to determine their consequences.
Collapse
Affiliation(s)
- Grant A Dixon
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Carlos A Pérez
- Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
38
|
Abstract
Emerging data point to important contributions of both autoimmune inflammation and progressive degeneration in the pathophysiology of multiple sclerosis (MS). Unfortunately, after decades of intensive investigation, the fundamental cause remains unknown. A large body of research on the immunobiology of MS has resulted in a variety of anti-inflammatory therapies that are highly effective at reducing brain inflammation and clinical/radiological relapses. However, despite potent suppression of inflammation, benefit in the more important and disabling progressive phase is extremely limited; thus, progressive MS has emerged as the greatest challenge for the MS research and clinical communities. Data obtained over the years point to a complex interplay between environment (e.g., the near-absolute requirement of Epstein-Barr virus exposure), immunogenetics (strong associations with a large number of immune genes), and an ever more convincing role of an underlying degenerative process resulting in demyelination (in both white and grey matter regions), axonal and neuro-synaptic injury, and a persistent innate inflammatory response with a seemingly diminishing role of T cell-mediated autoimmunity as the disease progresses. Together, these observations point toward a primary degenerative process, one whose cause remains unknown but one that entrains a nearly ubiquitous secondary autoimmune response, as a likely sequence of events underpinning this disease. Here, we briefly review what is known about the potential pathophysiological mechanisms, focus on progressive MS, and discuss the two main hypotheses of MS pathogenesis that are the topic of vigorous debate in the field: whether primary autoimmunity or degeneration lies at the foundation. Unravelling this controversy will be critically important for developing effective new therapies for the most disabling later phases of this disease.
Collapse
Affiliation(s)
- Peter K. Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Medicine University of Calgary, Calgary, Alberta, Canada
| | - Shigeki Tsutsui
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Medicine University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
39
|
Elkjaer ML, Frisch T, Reynolds R, Kacprowski T, Burton M, Kruse TA, Thomassen M, Baumbach J, Illes Z. Molecular signature of different lesion types in the brain white matter of patients with progressive multiple sclerosis. Acta Neuropathol Commun 2019; 7:205. [PMID: 31829262 PMCID: PMC6907342 DOI: 10.1186/s40478-019-0855-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022] Open
Abstract
To identify pathogenetic markers and potential drivers of different lesion types in the white matter (WM) of patients with progressive multiple sclerosis (PMS), we sequenced RNA from 73 different WM areas. Compared to 25 WM controls, 6713 out of 18,609 genes were significantly differentially expressed in MS tissues (FDR < 0.05). A computational systems medicine analysis was performed to describe the MS lesion endophenotypes. The cellular source of specific molecules was examined by RNAscope, immunohistochemistry, and immunofluorescence. To examine common lesion specific mechanisms, we performed de novo network enrichment based on shared differentially expressed genes (DEGs), and found TGFβ-R2 as a central hub. RNAscope revealed astrocytes as the cellular source of TGFβ-R2 in remyelinating lesions. Since lesion-specific unique DEGs were more common than shared signatures, we examined lesion-specific pathways and de novo networks enriched with unique DEGs. Such network analysis indicated classic inflammatory responses in active lesions; catabolic and heat shock protein responses in inactive lesions; neuronal/axonal specific processes in chronic active lesions. In remyelinating lesions, de novo analyses identified axonal transport responses and adaptive immune markers, which was also supported by the most heterogeneous immunoglobulin gene expression. The signature of the normal-appearing white matter (NAWM) was more similar to control WM than to lesions: only 465 DEGs differentiated NAWM from controls, and 16 were unique. The upregulated marker CD26/DPP4 was expressed by microglia in the NAWM but by mononuclear cells in active lesions, which may indicate a special subset of microglia before the lesion develops, but also emphasizes that omics related to MS lesions should be interpreted in the context of different lesions types. While chronic active lesions were the most distinct from control WM based on the highest number of unique DEGs (n = 2213), remyelinating lesions had the highest gene expression levels, and the most different molecular map from chronic active lesions. This may suggest that these two lesion types represent two ends of the spectrum of lesion evolution in PMS. The profound changes in chronic active lesions, the predominance of synaptic/neural/axonal signatures coupled with minor inflammation may indicate end-stage irreversible molecular events responsible for this less treatable phase.
Collapse
|
40
|
Abstract
Fingolimod is the first orally administered drug approved for the treatment of relapsing-remitting multiple sclerosis (MS). This drug, modulating sphingosine receptors, regulates the trafficking of lymphocytes between primary and secondary lymphoid organs, trapping naïve T cells and central memory T cells in secondary lymphoid organs, without affecting effector memory T cells and therefore without compromising immunosurveillance. Additionally, fingolimod inhibits expression of Th1 and Th17 cytokines and enhances regulatory T-cell differentiation. It also acts on the B arm of immunity through an increased ratio of naïve to memory B cells, higher percentage of plasma cells, and highly increased proportion of transitional B cells as well as additional regulatory subsets. Fingolimod treatment enhances the capacity of regulatory B cells to transmigrate across brain endothelial cells. In fact, patients treated with fingolimod have increased regulatory B-cell frequency in the cerebrospinal fluid. These findings suggest a novel role for fingolimod in MS, by both direct effects and indirect partitioning effects on lymphocytes.
Collapse
Affiliation(s)
| | - Diego Centonze
- Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy.
- Department of Systems Medicine, Tor Vergata University, Rome, Italy.
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| |
Collapse
|
41
|
Morshedi M, Hashemi R, Moazzen S, Sahebkar A, Hosseinifard ES. Immunomodulatory and anti-inflammatory effects of probiotics in multiple sclerosis: a systematic review. J Neuroinflammation 2019; 16:231. [PMID: 31752913 PMCID: PMC6868771 DOI: 10.1186/s12974-019-1611-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and autoimmune neurological disorder which leads to demyelination. Although the etiology of MS is yet to be known, it appears that regulating the immune system and suppressing inflammatory pathways may possibly have a favorable effect on the healing of this disease. Evidence suggests that probiotics consumption via gut microbiome alteration devises beneficial effects in improving immune and inflammatory responses in MS. All articles were systematically searched (in the main databases) for this paper. Two investigators independently scrutinized full texts of the potentially eligible articles. The quality of the study was evaluated using standardized tools. The methodological quality of seven studies included in this review ranged from fair to good. The findings illustrated that there were statistically significant improvements in the static and dynamic balance in patients and animals with MS. In the paper in hand, the effects of probiotics administration on immune and inflammatory markers in MS disease are evaluated. In addition, the limitations and knowledge gaps were reported while proposing a possible mechanism of probiotics therapy in modulating immune and inflammatory responses. This systematic review indicated that the probiotics could improve immune and inflammatory parameters, the cytokines and cells in MS disease. Probiotics may have efficient effects in management and treatment of MS. More studies are required to clarify the effect of supplementation with probiotics and their mechanisms in MS disease.
Collapse
Affiliation(s)
- Mohammad Morshedi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Hashemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Moazzen
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, 9713, GZ, the Netherlands
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
42
|
Oki S. Eomes-expressing T-helper cells as potential target of therapy in chronic neuroinflammation. Neurochem Int 2019; 130:104348. [DOI: 10.1016/j.neuint.2018.11.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 11/29/2022]
|
43
|
Mohtashami L, Shakeri A, Javadi B. Neuroprotective natural products against experimental autoimmune encephalomyelitis: A review. Neurochem Int 2019; 129:104516. [DOI: 10.1016/j.neuint.2019.104516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
|
44
|
Azevedo CJ, Cen SY, Jaberzadeh A, Zheng L, Hauser SL, Pelletier D. Contribution of normal aging to brain atrophy in MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:6/6/e616. [PMID: 32330116 PMCID: PMC6807662 DOI: 10.1212/nxi.0000000000000616] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 07/01/2019] [Indexed: 12/21/2022]
Abstract
Objective To identify the top brain regions affected by MS-specific atrophy (i.e., atrophy in excess of normal aging) and to test whether normal aging and MS-specific atrophy increase or decrease in these regions with age. Methods Six hundred fifty subjects (2,790 MRI time points) were analyzed: 520 subjects with relapse-onset MS from a 5-year prospective cohort with annual standardized 1-mm 3D T1-weighted images (3DT1s; 2,483 MRIs) and 130 healthy controls with longitudinal 3DT1s (307 MRIs). Rates of change in all FreeSurfer regions (v5.3) and Structural Image Evaluation Using Normalization of Atrophy (SIENA) were estimated with mixed-effects models. All FreeSurfer regions were ranked by the MS-specific atrophy slope/standard error ratio (βMS × time/SEβMS × time). In the top regions, age was added as an effect modifier to test whether MS-specific atrophy varied by age. Results The top-ranked regions were all gray matter structures. For SIENA, normal aging increased from 0.01%/y at age 30 years to −0.31%/y at age 60 years (−0.11% ± 0.032%/decade, p < 0.01), whereas MS-specific atrophy decreased from −0.38%/y at age 30 years to −0.12%/y at age 60 years (0.09% ± 0.035%/decade, p = 0.01). Similarly, in the thalamus, normal aging increased from −0.15%/y at age 30 years to −0.62%/y at age 60 years (−0.16% ± 0.079%/decade, p < 0.05), and MS-specific atrophy decreased from −0.59%/y at age 30 years to −0.05%/y at age 60 years (0.18% ± 0.08%/decade, p < 0.05). In the putamen and caudate, normal aging and MS-specific atrophy did not vary by age. Conclusions For SIENA and thalamic atrophy, the contribution of normal aging increases with age, but does not change in the putamen and caudate. This may have substantial implications to understand the biology of brain atrophy in MS.
Collapse
Affiliation(s)
- Christina J Azevedo
- From the Department of Neurology (C.J.A., S.Y.C., A.J., L.Z., D.P.), University of Southern California, Los Angeles; and Department of Neurology (S.L.H.), University of California, San Francisco
| | - Steven Y Cen
- From the Department of Neurology (C.J.A., S.Y.C., A.J., L.Z., D.P.), University of Southern California, Los Angeles; and Department of Neurology (S.L.H.), University of California, San Francisco
| | - Amir Jaberzadeh
- From the Department of Neurology (C.J.A., S.Y.C., A.J., L.Z., D.P.), University of Southern California, Los Angeles; and Department of Neurology (S.L.H.), University of California, San Francisco
| | - Ling Zheng
- From the Department of Neurology (C.J.A., S.Y.C., A.J., L.Z., D.P.), University of Southern California, Los Angeles; and Department of Neurology (S.L.H.), University of California, San Francisco
| | - Stephen L Hauser
- From the Department of Neurology (C.J.A., S.Y.C., A.J., L.Z., D.P.), University of Southern California, Los Angeles; and Department of Neurology (S.L.H.), University of California, San Francisco
| | - Daniel Pelletier
- From the Department of Neurology (C.J.A., S.Y.C., A.J., L.Z., D.P.), University of Southern California, Los Angeles; and Department of Neurology (S.L.H.), University of California, San Francisco.
| |
Collapse
|
45
|
Differential Modulation of NF- κB in Neurons and Astrocytes Underlies Neuroprotection and Antigliosis Activity of Natural Antioxidant Molecules. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8056904. [PMID: 31485299 PMCID: PMC6710787 DOI: 10.1155/2019/8056904] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 06/20/2019] [Indexed: 02/02/2023]
Abstract
Neuroinflammation, a hallmark of chronic neurodegenerative disorders, is characterized by sustained glial activation and the generation of an inflammatory loop, through the release of cytokines and other neurotoxic mediators that cause oxidative stress and limit functional repair of brain parenchyma. Dietary antioxidants may protect against neurodegenerative diseases by counteracting chronic neuroinflammation and reducing oxidative stress. Here, we describe the effects of a number of natural antioxidants (polyphenols, carotenoids, and thiolic molecules) in rescuing astrocytic function and neuronal viability following glial activation by reducing astrocyte proliferation and restoring astrocytic and neuronal survival and basal levels of reactive oxygen species (ROS). All antioxidant molecules are also effective under conditions of oxidative stress and glutamate toxicity, two maladaptive components of neuroinflammatory processes. Moreover, it is remarkable that their antioxidant and anti-inflammatory activity occurs through differential modulation of NF-κB binding activity in neurons and astrocytes. In fact, we show that inflammatory stimuli promote a significant induction of NF-κB binding activity in astrocytes and its concomitant reduction in neurons. These changes are prevented in astrocytes and neurons pretreated with the antioxidant molecules, suggesting that NF-κB plays a key role in the modulation of survival and anti-inflammatory responses. Finally, we newly demonstrate that effective antigliosis and neuroprotective activity is achieved with a defined cocktail of four natural antioxidants at very low concentrations, suggesting a promising strategy to reduce inflammatory and oxidative damage in neurodegenerative diseases with limited side effects.
Collapse
|
46
|
Elkjaer ML, Frisch T, Reynolds R, Kacprowski T, Burton M, Kruse TA, Thomassen M, Baumbach J, Illes Z. Unique RNA signature of different lesion types in the brain white matter in progressive multiple sclerosis. Acta Neuropathol Commun 2019; 7:58. [PMID: 31023379 PMCID: PMC6482546 DOI: 10.1186/s40478-019-0709-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 01/18/2023] Open
Abstract
The heterogeneity of multiple sclerosis is reflected by dynamic changes of different lesion types in the brain white matter (WM). To identify potential drivers of this process, we RNA-sequenced 73 WM areas from patients with progressive MS (PMS) and 25 control WM. Lesion endophenotypes were described by a computational systems medicine analysis combined with RNAscope, immunohistochemistry, and immunofluorescence. The signature of the normal-appearing WM (NAWM) was more similar to control WM than to lesions: one of the six upregulated genes in NAWM was CD26/DPP4 expressed by microglia. Chronic active lesions that become prominent in PMS had a signature that were different from all other lesion types, and were differentiated from them by two clusters of 62 differentially expressed genes (DEGs). An upcoming MS biomarker, CHI3L1 was among the top ten upregulated genes in chronic active lesions expressed by astrocytes in the rim. TGFβ-R2 was the central hub in a remyelination-related protein interaction network, and was expressed there by astrocytes. We used de novo networks enriched by unique DEGs to determine lesion-specific pathway regulation, i.e. cellular trafficking and activation in active lesions; healing and immune responses in remyelinating lesions characterized by the most heterogeneous immunoglobulin gene expression; coagulation and ion balance in inactive lesions; and metabolic changes in chronic active lesions. Because we found inverse differential regulation of particular genes among different lesion types, our data emphasize that omics related to MS lesions should be interpreted in the context of lesion pathology. Our data indicate that the impact of molecular pathways is substantially changing as different lesions develop. This was also reflected by the high number of unique DEGs that were more common than shared signatures. A special microglia subset characterized by CD26 may play a role in early lesion development, while astrocyte-derived TGFβ-R2 and TGFβ pathways may be drivers of repair in contrast to chronic tissue damage. The highly specific mechanistic signature of chronic active lesions indicates that as these lesions develop in PMS, the molecular changes are substantially skewed: the unique mitochondrial/metabolic changes and specific downregulation of molecules involved in tissue repair may reflect a stage of exhaustion.
Collapse
|
47
|
Abdelhak A, Hottenrott T, Morenas-Rodríguez E, Suárez-Calvet M, Zettl UK, Haass C, Meuth SG, Rauer S, Otto M, Tumani H, Huss A. Glial Activation Markers in CSF and Serum From Patients With Primary Progressive Multiple Sclerosis: Potential of Serum GFAP as Disease Severity Marker? Front Neurol 2019; 10:280. [PMID: 30972011 PMCID: PMC6443875 DOI: 10.3389/fneur.2019.00280] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
Background: In progressive multiple sclerosis (MS), glial activation is thought to be a relevant mechanism of disability progression. Therefore, in vivo assessment of the glial cell activity is, in the emerging treatment era of primary progressive MS (PPMS), more important than ever. Objectives: To test the association of cerebrospinal fluid (CSF) and serum markers of glial activation in PPMS patients; including glial fibrillary acidic protein (GFAP), chitinase-3-like protein 1 (CHI3L1), soluble variant of triggering receptor expressed on myeloid cells 2 (sTREM2), and marker of neuroaxonal damage (Neurofilament light chain, NfL) as well as clinical severity. Methods: CSF and serum samples from PPMS patients were collected in the MS-centers at Universities of Freiburg (n = 49), Ulm (n = 27), Muenster (n = 11), and Rostock (n = 6). sTREM2 and CHI3L1 levels were measured using the previously reported ELISA assays, while NfL and GFAP were measured using SIMOA assays. Clinical data included age, gender, disease duration, treatment status, and Expanded Disability Status Scale (EDSS). Results: 93 CSF samples and 71 matching serum samples were analyzed. The median age of patients was 49 years and disease duration 4.5 years. GFAPserum correlated with EDSS after correction for age (β = 0.3, p = 0.001). Furthermore, EDSS was higher in patients with a GFAPserum level ≥ 151.7 pg/ml compared to patients with GFAPserum below this cut-off (5.5 vs. 4.0, p = 0.009). Other markers did not correlate with the clinical severity. Moreover, we found a correlation between NfLCSF and GFAPCSF, sTREM2 and CHI3L1 (ρ = 0.4 for GFAPCSF and sTREM2, ρ = 0.3 for CHI3L1, p < 0.01 for sTREM2 and CHI3L1 and <0.001 for GFAPCSF). CHI3L1 did not correlate with GFAPCSF but with sTREM2 (ρ = 0.4, p < 0.01). Discussion: The correlation between the glial activation markers in CSF with the markers of neuroaxonal demise supports the notion of the glial involvement in PPMS. The positive correlation between GFAPCSF with disease duration and GFAPserum with the clinical severity of the disease may highlight a particular role of the astrocytes in PPMS and mark the potential of GFAPserum as a disease severity marker.
Collapse
Affiliation(s)
- Ahmed Abdelhak
- Department of Neurology, University Hospital of Tuebingen, Tuebingen, Germany.,Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Tilman Hottenrott
- Department of Neurology, University Hospital of Freiburg, Freiburg, Germany
| | - Estrella Morenas-Rodríguez
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,BioMedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Uwe K Zettl
- Neuroimmunological Section, Department of Neurology, University Hospital of Rostock, Rostock, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,BioMedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sven G Meuth
- Department of Neurology, University Hospital of Muenster, Münster, Germany
| | - Sebastian Rauer
- Department of Neurology, University Hospital of Freiburg, Freiburg, Germany
| | - Markus Otto
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, University Hospital of Ulm, Ulm, Germany.,Specialty Hospital Dietenbronn, Schwendi, Germany
| | - André Huss
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
48
|
Correale J, Marrodan M, Ysrraelit MC. Mechanisms of Neurodegeneration and Axonal Dysfunction in Progressive Multiple Sclerosis. Biomedicines 2019; 7:biomedicines7010014. [PMID: 30791637 PMCID: PMC6466454 DOI: 10.3390/biomedicines7010014] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/14/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Multiple Sclerosis (MS) is a major cause of neurological disability, which increases predominantly during disease progression as a result of cortical and grey matter structures involvement. The gradual accumulation of disability characteristic of the disease seems to also result from a different set of mechanisms, including in particular immune reactions confined to the Central Nervous System such as: (a) B-cell dysregulation, (b) CD8+ T cells causing demyelination or axonal/neuronal damage, and (c) microglial cell activation associated with neuritic transection found in cortical demyelinating lesions. Other potential drivers of neurodegeneration are generation of oxygen and nitrogen reactive species, and mitochondrial damage, inducing impaired energy production, and intra-axonal accumulation of Ca2+, which in turn activates a variety of catabolic enzymes ultimately leading to progressive proteolytic degradation of cytoskeleton proteins. Loss of axon energy provided by oligodendrocytes determines further axonal degeneration and neuronal loss. Clearly, these different mechanisms are not mutually exclusive and could act in combination. Given the multifactorial pathophysiology of progressive MS, many potential therapeutic targets could be investigated in the future. This remains however, an objective that has yet to be undertaken.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, FLENI, Buenos Aires 1428, Argentina.
| | | | | |
Collapse
|
49
|
Iannetta M, Zingaropoli MA, Latronico T, Pati I, Pontecorvo S, Prezioso C, Pietropaolo V, Cortese A, Frontoni M, D'Agostino C, Francia A, Vullo V, Mastroianni CM, Liuzzi GM, Ciardi MR. Dynamic changes of MMP-9 plasma levels correlate with JCV reactivation and immune activation in natalizumab-treated multiple sclerosis patients. Sci Rep 2019; 9:311. [PMID: 30670793 PMCID: PMC6342994 DOI: 10.1038/s41598-018-36535-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
The aim of the study was to investigate the changes of matrix metalloproteinase (MMP)-2 and MMP-9 plasma levels during natalizumab treatment and their correlation with JC virus (JCV) reactivation and T-lymphocyte phenotypic modifications in peripheral blood samples from 34 relapsing-remitting multiple sclerosis (RRMS) patients. MMP-9 levels were assessed by zymography in plasma samples. JCV-DNA was detected through quantitative real time PCR in plasma samples. T-lymphocyte phenotype was assessed with flow cytometry. MMP-9 plasma levels resulted increased from 12 to 24 natalizumab infusions. Stratifying plasma samples according to JCV-DNA detection, MMP-9 plasma levels were significantly increased in JCV-DNA positive than JCV-DNA negative samples. MMP-9 plasma levels resulted positively correlated with JCV viral load. CD4 immune senescence, CD8 immune activation and CD8 effector percentages were positively correlated to MMP-9 plasma levels, whereas a negative correlation between CD8 naïve percentages and MMP-9 plasma levels was found. Our data indicate an increase of MMP-9 plasma levels between 12 and 24 natalizumab infusions and a correlation with JCV-DNA detection in plasma, T-lymphocyte immune activation and senescence. These findings could contribute to understand PML pathogenesis under natalizumab treatment, suggesting a potential role of MMP-9 as a predictive marker of PML in RRMS patients.
Collapse
Affiliation(s)
- Marco Iannetta
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | | | - Tiziana Latronico
- Department of Biosciences, Biotechnology and Biopharmaceutics, Aldo Moro University, Bari, Italy
| | - Ilaria Pati
- Department of Biosciences, Biotechnology and Biopharmaceutics, Aldo Moro University, Bari, Italy
| | - Simona Pontecorvo
- Department of Human Neuroscience, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Carla Prezioso
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Valeria Pietropaolo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Antonio Cortese
- Department of Human Neuroscience, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Marco Frontoni
- Department of Human Neuroscience, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Claudia D'Agostino
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Ada Francia
- Department of Human Neuroscience, Multiple Sclerosis Center, Sapienza University, Rome, Italy
| | - Vincenzo Vullo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | | | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnology and Biopharmaceutics, Aldo Moro University, Bari, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| |
Collapse
|
50
|
Koskimäki F, Bernard J, Yong J, Arndt N, Carroll T, Lee SK, Reder AT, Javed A. Gray matter atrophy in multiple sclerosis despite clinical and lesion stability during natalizumab treatment. PLoS One 2018; 13:e0209326. [PMID: 30576361 PMCID: PMC6303064 DOI: 10.1371/journal.pone.0209326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 12/04/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Brain volume loss is an important surrogate marker for assessing disability in MS; however, contribution of gray and white matter to the whole brain volume loss needs further examination in the context of specific MS treatment. OBJECTIVES To examine whole and segmented gray, white, thalamic, and corpus callosum volume loss in stable patients receiving natalizumab for 2-5 years. METHODS This was a retrospective study of 20 patients undergoing treatment with natalizumab for 24-68 months. Whole brain volume loss was determined with SIENA. Gray and white matter segmentation was done using FAST. Thalamic and corpus callosum volumes were determined using Freesurfer. T1 relaxation values of chronic hypointense lesions (black holes) were determined using a quantitative, in-house developed method to assess lesion evolution. RESULTS Over a mean of 36.6 months, median percent brain volume change (PBVC) was -2.0% (IQR 0.99-2.99). There was decline in gray (p = 0.001) but not white matter (p = 0.6), and thalamic (p = 0.01) but not corpus callosum volume (p = 0.09). Gray matter loss correlated with PBVC (Spearman's r = 0.64, p = 0.003) but not white matter (Spearman's r = 0.42, p = 0.07). Age significantly influenced whole brain volume loss (p = 0.010, multivariate regression), but disease duration and baseline T2 lesion volume did not. There was no change in T1 relaxation values of lesions or T2 lesion volume over time. All patients remained clinically stable. CONCLUSIONS These results demonstrate that brain volume loss in MS is primarily driven by gray matter changes and may be independent of clinically effective treatment.
Collapse
Affiliation(s)
- Fredrika Koskimäki
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Jacqueline Bernard
- Department of Neurology, Oregon Health Science University, Portland, Oregon, United States of America
| | - Jeong Yong
- Northwestern University, Biomedical Engineering, Chicago, Illinois, United States of America
| | - Nancy Arndt
- Department of Neurology, The University of Chicago, Chicago, Illinois, United States of America
| | - Timothy Carroll
- Department of Radiology, The University of Chicago, Chicago, Illinois, United States of America
| | - Seon-Kyu Lee
- Department of Radiology, The University of Chicago, Chicago, Illinois, United States of America
| | - Anthony T. Reder
- Department of Neurology, The University of Chicago, Chicago, Illinois, United States of America
| | - Adil Javed
- Department of Neurology, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|