101
|
Skarlis C, Anagnostouli M. The role of melatonin in Multiple Sclerosis. Neurol Sci 2019; 41:769-781. [PMID: 31845043 DOI: 10.1007/s10072-019-04137-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/31/2019] [Indexed: 12/20/2022]
Abstract
Melatonin is a neurohormone mainly produced by the pineal gland following a circadian rhythm. It is characterized as a pleiotropic factor because it not only regulates the wake-sleep rhythm but also exerts antinociceptive, antidepressant, anxiolytic, and immunomodulating properties. Recent studies suggest that dysregulation of melatonin secretion is associated with the pathogenesis of various autoimmune diseases, such as, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and multiple sclerosis (MS). MS is an autoimmune disorder characterized by an abnormal immune response directed against the myelin sheath in the central nervous system, demyelination, oligodendrocyte death, and axonal degeneration. Recent evidence reveals that melatonin secretion is dysregulated in MS patients, suggesting that melatonin could be a potential target for therapeutic intervention. Here, we summarize the available literature regarding the role of melatonin in immune processes relevant for experimental autoimmune encephalomyelitis (EAE), MS, and the current clinical trials of melatonin supplementation in MS patients.
Collapse
Affiliation(s)
- Charalampos Skarlis
- Immunogenetics Laboratory, 1st Department of Neurology, Medical School of National and Kapodistrian University of Athens, Aeginition Hospital, Vas. Sophias, 74, 115 28, Athens, Greece.
| | - Maria Anagnostouli
- Immunogenetics Laboratory, 1st Department of Neurology, Medical School of National and Kapodistrian University of Athens, Aeginition Hospital, Vas. Sophias, 74, 115 28, Athens, Greece. .,Demyelinating Diseases Clinic, 1st Department of Neurology, Medical School of National and Kapodistrian University of Athens, Aeginition Hospital, Athens, Greece.
| |
Collapse
|
102
|
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: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [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
|
103
|
Sylvestre DA, Slupsky CM, Aviv RI, Swardfager W, Taha AY. Untargeted metabolomic analysis of plasma from relapsing-remitting multiple sclerosis patients reveals changes in metabolites associated with structural changes in brain. Brain Res 2019; 1732:146589. [PMID: 31816317 DOI: 10.1016/j.brainres.2019.146589] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 11/18/2019] [Accepted: 12/03/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Changes in peripheral blood amino acids have been noted in Relapse Remitting Multiple Sclerosis (RRMS), suggesting their potential diagnostic value in anticipating disease progression. OBJECTIVE The present study sought to comprehensively assess the plasma metabolome, including amino acids, of RRMS patient and unaffected controls, to identify potential biomarkers of RRMS disease pathogenesis. METHODS Untargeted NMR metabolomics was performed on plasma from 28 RRMS patients and 18 unaffected controls to test the hypothesis that metabolomic markers are altered in RRMS patients in association with lesion load, brain atrophy and cognitive performance. RESULTS There were no significant differences between RRMS and controls in age, sex and total brain volume. Brain fractional volumes of gray matter, white matter, thalamus and parenchyma as well as multiple neurocognitive scores were significantly lower in RRMS patients compared to unaffected controls. Concentrations of nine plasma metabolites (arginine, isoleucine, citrate, serine, phenylalanine, methionine, asparagine, histidine, myo-inositol) were significantly lower in RRMS patients compared to controls. Plasma arginine concentrations were positively correlated with T1 holes and white matter lesions, and plasma methionine concentrations were positively correlated with T1 holes, but not white matter lesions. Serine was negatively correlated with performance on the Brief Visuospatial Memory Test in controls but not RRMS patients. CONCLUSIONS The identified disturbances in metabolite concentrations might be developed as new markers of neuroanatomical vulnerability in RRMS, should the findings be reproduced in larger cohort studies.
Collapse
Affiliation(s)
- Duncan A Sylvestre
- Department of Food Science and Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, USA
| | - Carolyn M Slupsky
- Department of Food Science and Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, USA
| | - Richard I Aviv
- Department of Radiology, Ottawa University, Division of Neuroradiology, The Ottawa Hospital, Ottawa, ON, K1H8L6, Canada
| | - Walter Swardfager
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada; Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada; Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada; LC Campbell Cognitive Neurology Unit, Sunnybrook Research Institute, Toronto, Canada; University Health Network Toronto Rehabilitation Institute, Toronto, Canada
| | - Ameer Y Taha
- Department of Food Science and Technology, University of California, Davis, USA.
| |
Collapse
|
104
|
The Na +/Ca 2+ exchangers in demyelinating diseases. Cell Calcium 2019; 85:102130. [PMID: 31812115 DOI: 10.1016/j.ceca.2019.102130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022]
Abstract
Intracellular [Na+]i and [Ca2+]i imbalance significantly contribute to neuro-axonal dysfunctions and maladaptive myelin repair or remyelination failure in chronic inflammatory demyelinating diseases such as multiple sclerosis. Progress in recent years has led to significant advances in understanding how [Ca2+]i signaling network drive degeneration or remyelination of demyelinated axons. The Na+/Ca2+ exchangers (NCXs), a transmembrane protein family including three members encoded by ncx1, ncx2, and ncx3 genes, are emerging important regulators of [Na+]i and [Ca2+]i both in neurons and glial cells. Here we review recent advance highlighting the role of NCX exchangers in axons and myelin-forming cells, i.e. oligodendrocytes, which represent the major targets of the aberrant inflammatory attack in multiple sclerosis. The contribution of NCX subtypes to axonal pathology and myelin synthesis will be discussed. Although a definitive understanding of mechanisms regulating axonal pathology and remyelination failure in chronic demyelinating diseases is still lacking and requires further investigation, current knowledge suggest that NCX activity plays a crucial role in these processes. Defining the relative contributions of each NCX transporter in axon pathology and myelinating glia will constitute not only a major advance in understanding in detail the intricate mechanism of neurodegeneration and remyelination failure in demyelinating diseases but also will help to identify neuroprotective or remyelinating strategies targeting selective NCX exchangers as a means of treating MS.
Collapse
|
105
|
Anselmo F, Tatomir A, Boodhoo D, Mekala AP, Nguyen V, Rus V, Rus H. JNK and phosphorylated Bcl-2 predict multiple sclerosis clinical activity and glatiramer acetate therapeutic response. Clin Immunol 2019; 210:108297. [PMID: 31698073 DOI: 10.1016/j.clim.2019.108297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/15/2019] [Accepted: 11/03/2019] [Indexed: 01/04/2023]
Abstract
In this study, we investigated the role of JNK and phospho-Bcl-2 as possible biomarkers of multiple sclerosis (MS) relapse and of glatiramer acetate (GA) therapeutic response in relapsing-remitting MS patients. We enrolled a cohort of 15 GA-treated patients and measured the expression of JNK1, JNK2, phospho-JNK and phospho-Bcl-2 through Western blotting of lysates from peripheral blood mononuclear cells collected at 0, 3, 6, and 12 months after initiating GA therapy. We found significantly higher levels of JNK1 p54 and JNK2 p54 and significantly lower levels of p-Bcl-2 in relapse patients and in GA non-responders. By using receiver operating characteristic analysis, we found that the probability of accurately detecting relapse and response to GA was: 92% and 75.5%, respectively, for JNK1 p54 and 86% and 94.6%, respectively, for p-Bcl-2. Our data suggest that JNK1 and p-Bcl-2 could serve as potential biomarkers for MS relapse and the therapeutic response to GA.
Collapse
Affiliation(s)
- Freidrich Anselmo
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alexandru Tatomir
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Neurosciences, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Armugam P Mekala
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Horea Rus
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA.
| |
Collapse
|
106
|
Bianchi VE, Herrera PF, Laura R. Effect of nutrition on neurodegenerative diseases. A systematic review. Nutr Neurosci 2019; 24:810-834. [PMID: 31684843 DOI: 10.1080/1028415x.2019.1681088] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neurodegenerative diseases are characterized by the progressive functional loss of neurons in the brain, causing cognitive impairment and motoneuron disability. Although multifactorial interactions are evident, nutrition plays an essential role in the pathogenesis and evolution of these diseases. A systematic literature search was performed, and the prevalence of studies evaluated the effect of the Mediterranean diet (MeDiet), nutritional support, EPA and DHA, and vitamins on memory and cognition impairment. The data showed that malnutrition and low body mass index (BMI) is correlated with the higher development of dementia and mortality. MeDiet, nutritional support, and calorie-controlled diets play a protective effect against cognitive decline, Alzheimer's disease (AD), Parkinson disease (PD) while malnutrition and insulin resistance represent significant risk factors. Malnutrition activates also the gut-microbiota-brain axis dysfunction that exacerbate neurogenerative process. Omega-3 and -6, and the vitamins supplementation seem to be less effective in protecting neuron degeneration. Insulin activity is a prevalent factor contributing to brain health while malnutrition correlated with the higher development of dementia and mortality.
Collapse
Affiliation(s)
| | - Pomares Fredy Herrera
- Director del Centro de Telemedicina, Grupo de investigación en Atención Primaria en salud/Telesalud, Doctorado en Medicina /Neurociencias, University of Cartagena, Colombia
| | - Rizzi Laura
- Molecular Biology, School of Medicine and Surgery, University of Milano-Bicocca, Monza Brianza, Italy
| |
Collapse
|
107
|
Wendrich K, van Oirschot P, Martens MB, Heerings M, Jongen PJ, Krabbenborg L. Toward Digital Self-monitoring of Multiple Sclerosis: Investigating First Experiences, Needs, and Wishes of People with MS. Int J MS Care 2019; 21:282-291. [PMID: 31889935 DOI: 10.7224/1537-2073.2018-083] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Digital self-monitoring, such as through smartphone applications (apps) or activity trackers, could be applied to monitor the health of people with multiple sclerosis (MS). This self-monitoring could facilitate personalized therapies and self-management of MS. The acceptance of digital self-monitoring tools by patients depends on them being able and willing to use these tools in their daily lives. Methods In-depth interviews were conducted with seven adults with MS before and after participation in a study in which they used an activity tracker and an MS-specific smartphone app for 4 weeks. We inquired about experiences with the tools in daily life and needs and wishes regarding further development and implementation of digital self-monitoring for people with MS. Results The smartphone app and the activity tracker increased respondents' awareness of their physical status and stimulated them to act on the data. Challenges, such as confrontation with their MS and difficulties with data interpretation, were discussed. The respondents desired 1) adaptation of digital self-monitoring tools to a patient's personal situation, 2) guidance to increase the value of the data, and 3) integration of digital self-monitoring into treatment plans. Conclusions These findings show that patients can provide detailed descriptions of their daily life experiences with new technologies. Mapping these experiences could help in better aligning the development and implementation of digital self-monitoring tools, in this case smartphones and activity trackers, with the needs and wishes of people with MS.
Collapse
|
108
|
Ata E, Özsoy-Ünübol T, Demir S. Evaluation of Neuropathic Extremity Pain in Patients with Multiple Sclerosis. ACTA ACUST UNITED AC 2019; 58:213-216. [PMID: 34526844 PMCID: PMC8419729 DOI: 10.29399/npa.24802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/21/2019] [Indexed: 12/02/2022]
Abstract
Introduction: Neuropathic extremity pain is one of the most common symptoms of multiple sclerosis (MS). This study was aimed to evaluate and compare the frequency and severity of neuropathic extremity pain in subtypes of MS. Methods: Patients with the diagnosis of MS were included in the study, consecutively. Patients’ demographic and clinical data were recorded. Patients’ pain severity was assessed with visual analog scale (VAS). For the evaluation of neuropathic pain Leeds assessment of neuropathic symptoms and signs (LANSS) and douleur neuropathique 4 questions (DN4) were used. Disability was assessed with the Extended Disability Status Scale (EDSS). Results: One hundred and three patients were included and 82.5% of them had relapsing-remitting (RR) MS. According to LANSS and DN4, 47.6% of patients had neuropathic pain. Female patients had significantly higher pain scores. There was no significant difference between the subtypes of MS based on having neuropathic pain. Patients with seconder progressive (SP) MS had significantly higher EDSS. A significant positive correlation was detected between EDSS score and duration of disease. Conclusion: Regardless of the MS subtype patients had neuropathic pain. Patients with SP had significantly higher disease duration and EDSS which were correlated with each other. All patients with the diagnosis of MS should be evaluated for neuropathic pain.
Collapse
Affiliation(s)
- Emre Ata
- Department of Physical Medicine and Rehabilitation, Sultan Abdülhamid Han Training and Research Hospital, İstanbul, Turkey
| | - Tuğba Özsoy-Ünübol
- Department of Physical Medicine and Rehabilitation, Sultan Abdülhamid Han Training and Research Hospital, İstanbul, Turkey
| | - Serkan Demir
- Department of Neurology, Sultan Abdülhamid Han Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
109
|
Bisphenol A triggers axonal injury and myelin degeneration with concomitant neurobehavioral toxicity in C57BL/6J male mice. Toxicology 2019; 428:152299. [PMID: 31574244 DOI: 10.1016/j.tox.2019.152299] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/10/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022]
Abstract
Bisphenol A (BPA) is a ubiquitously distributed endocrine disrupting chemical (EDC). BPA exposure in humans has been a matter of concern due to its increased application in the products of day to day use. BPA has been reported to cause toxicity in almost all the vital organ systems even at a very low dose levels. It crosses the blood brain barrier and causes neurotoxicity. We studied the effect of BPA on the cerebral cortex of C57BL/6J mice and examined whether BPA exposure alters the expression of axonal and myelin structural proteins. Male mice were dosed orally to 40 μg and 400 μg BPA/kg body weight for 60 days. BPA exposure resulted in memory loss, muscle coordination deficits and allodynia. BPA exposure also caused degeneration of immature and mature oligodendrocytes as evaluated by decreased mRNA levels of 2',3'-cyclic nucleotide 3' phosphodiesterase (CNPase), nestin, myelin basic protein (MBP) and myelin-associated glycoprotein-1 (MAG-1) genes revealing myelin related pathology. It was observed that subchronic BPA exposure caused neuroinflammation through deregulation of inflammatory cytokines mRNA and protein expression which further resulted into neurotoxicity through axonal as well as myelin degeneration in the brain. BPA also caused increased oxidative stress in the brain. Our study indicates long-term subchronic low dose exposure to BPA has the potential to cause axonal degeneration and demyelination in the oligodendrocytes and neurons which may have implications in neurological and neuropsychological disorders including multiple sclerosis (MS), neuromyelitis optica and others.
Collapse
|
110
|
Salehi B, Berkay Yılmaz Y, Antika G, Boyunegmez Tumer T, Fawzi Mahomoodally M, Lobine D, Akram M, Riaz M, Capanoglu E, Sharopov F, Martins N, Cho WC, Sharifi-Rad J. Insights on the Use of α-Lipoic Acid for Therapeutic Purposes. Biomolecules 2019; 9:biom9080356. [PMID: 31405030 PMCID: PMC6723188 DOI: 10.3390/biom9080356] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022] Open
Abstract
α-lipoic acid (ALA, thioctic acid) is an organosulfur component produced from plants, animals, and humans. It has various properties, among them great antioxidant potential and is widely used as a racemic drug for diabetic polyneuropathy-associated pain and paresthesia. Naturally, ALA is located in mitochondria, where it is used as a cofactor for pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase complexes. Despite its various potentials, ALA therapeutic efficacy is relatively low due to its pharmacokinetic profile. Data suggests that ALA has a short half-life and bioavailability (about 30%) triggered by its hepatic degradation, reduced solubility as well as instability in the stomach. However, the use of various innovative formulations has greatly improved ALA bioavailability. The R enantiomer of ALA shows better pharmacokinetic parameters, including increased bioavailability as compared to its S enantiomer. Indeed, the use of amphiphilic matrices has capability to improve ALA bioavailability and intestinal absorption. Also, ALA's liquid formulations are associated with greater plasma concentration and bioavailability as compared to its solidified dosage form. Thus, improved formulations can increase both ALA absorption and bioavailability, leading to a raise in therapeutic efficacy. Interestingly, ALA bioavailability will be dependent on age, while no difference has been found for gender. The present review aims to provide an updated on studies from preclinical to clinical trials assessing ALA's usages in diabetic patients with neuropathy, obesity, central nervous system-related diseases and abnormalities in pregnancy.
Collapse
Affiliation(s)
- Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam 44340847, Iran
| | - Yakup Berkay Yılmaz
- Graduate Program of Biomolecular Sciences, Institute of Natural and Applied Sciences, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Gizem Antika
- Graduate Program of Biomolecular Sciences, Institute of Natural and Applied Sciences, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | | | - Devina Lobine
- Department of Health Sciences; Faculty of Science, University of Mauritius, Réduit 80837, Mauritius
| | - Muhammad Akram
- Department of Eastern Medicine, Government College University Faisalabad; Faisalabad 38000, Pakistan
| | - Muhammad Riaz
- Department of Allied Health Sciences, Sargodha Medical College, University of Sargodha, Sargodha 40100, Pakistan
| | - Esra Capanoglu
- Faculty of Chemical & Metallurgical Engineering, Food Engineering Department, Istanbul Technical University, Maslak 34469, Turkey
| | - Farukh Sharopov
- Department of Pharmaceutical Technology, Avicenna Tajik State Medical University, Rudaki 139, Dushanbe 734003, Tajikistan.
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong.
| | - Javad Sharifi-Rad
- Zabol Medicinal Plants Research Center, Zabol University of Medical Sciences, Zabol 61615-585, Iran.
| |
Collapse
|
111
|
Inojosa H, Proschmann U, Akgün K, Ziemssen T. A focus on secondary progressive multiple sclerosis (SPMS): challenges in diagnosis and definition. J Neurol 2019. [PMID: 31363847 DOI: 10.1007/s00415-019-09489-5.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
Secondary progressive multiple sclerosis (SPMS) is the second most common form of multiple sclerosis (MS). One in two relapse remitting multiple sclerosis (RRMS) patients will develop SPMS within 15 years and up to two-thirds after 30 years, leading to a progressive decrease of neurological function and limitation of daily activities. Nevertheless, the SPMS diagnosis is often established retrospectively and delayed up to 3 years due to several patient- and clinician-related factors. Definitive clinical diagnostic criteria are lacking and research is currently ongoing to identify imaging and biochemical biomarkers. As new therapies are introduced, early SPMS diagnosis may represent a window of opportunity for intervention. New approaches, endpoints or technologies could help physicians establishing a diagnosis. Here, we review SPMS in relation to its diagnostic and definition challenges and current screening techniques and tools.
Collapse
Affiliation(s)
- Hernan Inojosa
- Department of Neurology, Center of Clinical Neuroscience, Carl Gustav Carus University Clinic, University Hospital of Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Undine Proschmann
- Department of Neurology, Center of Clinical Neuroscience, Carl Gustav Carus University Clinic, University Hospital of Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Katja Akgün
- Department of Neurology, Center of Clinical Neuroscience, Carl Gustav Carus University Clinic, University Hospital of Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Tjalf Ziemssen
- Department of Neurology, Center of Clinical Neuroscience, Carl Gustav Carus University Clinic, University Hospital of Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
112
|
Malekzadeh A, Leurs C, van Wieringen W, Steenwijk MD, Schoonheim MM, Amann M, Naegelin Y, Kuhle J, Killestein J, Teunissen CE. Plasma proteome in multiple sclerosis disease progression. Ann Clin Transl Neurol 2019; 6:1582-1594. [PMID: 31364818 PMCID: PMC7651845 DOI: 10.1002/acn3.771] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 01/01/2023] Open
Abstract
Background The pathophysiology of multiple sclerosis disease progression remains undetermined. The aim of this study was to identify differences in plasma proteome during different stages of MS disease progression. Methods We used a multiplex aptamer proteomics platform (Somalogic) for sensitive detection of 1129 proteins in plasma. MS patients were selected and categorized based on baseline and a 4‐year follow‐up EDSS (delta EDSS) scores; relapse‐onset (RO) slow progression (n = 31), RO with rapid progression (n = 29), primary progressive (n = 30), and healthy controls (n = 20). The relation of baseline plasma protein levels with delta EDSS and different MRI progression parameters were assessed using linear regression models. Results Regression analyses of plasma proteins with delta EDSS showed six significant associations. Strong associations were found for the proteins LGLAS8 (P = 7.64 × 10−5, q = 0.06), CCL3 (P = 0.0001, q = 0.06), and RGMA (P = 0.0005, q = 0.09). In addition, associations of plasma proteins were found with percentage brain volume for C3 (P = 2,08 × 10−9, q = 1,70 × 10−6), FGF9 (P = 3,42 × 10−9, q = 1,70 × 10−6), and EHMT2 (P = 0.0007, q = 0.01). Most of the significant markers were associated with cell‐cell and cell‐extracellular matrix adhesion, immune system communication, immune system activation, and complement pathways. Conclusions Our results revealed eight novel biomarkers related to clinical and radiological progression in MS. These results indicate that changes in immune system, complement pathway and ECM remodeling proteins contribute to MS progression and may therefore be further explored for use in prognosis of MS.
Collapse
Affiliation(s)
- Arjan Malekzadeh
- Department of Clinical Chemistry, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Cyra Leurs
- Department of Neurology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Wessel van Wieringen
- Department of Mathematics, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Martijn D Steenwijk
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Michael Amann
- Division of Diagnostic and Interventional Neuroradiology, Department of Radiology and Nuclear Medicine, University Hospital Basel, Basel, Switzerland.,Medical Image Analysis Center (MIAC AG), Basel, Switzerland
| | - Yvonne Naegelin
- Department of Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Jens Kuhle
- Department of Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Joep Killestein
- Department of Neurology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
113
|
A focus on secondary progressive multiple sclerosis (SPMS): challenges in diagnosis and definition. J Neurol 2019; 268:1210-1221. [PMID: 31363847 DOI: 10.1007/s00415-019-09489-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/23/2022]
Abstract
Secondary progressive multiple sclerosis (SPMS) is the second most common form of multiple sclerosis (MS). One in two relapse remitting multiple sclerosis (RRMS) patients will develop SPMS within 15 years and up to two-thirds after 30 years, leading to a progressive decrease of neurological function and limitation of daily activities. Nevertheless, the SPMS diagnosis is often established retrospectively and delayed up to 3 years due to several patient- and clinician-related factors. Definitive clinical diagnostic criteria are lacking and research is currently ongoing to identify imaging and biochemical biomarkers. As new therapies are introduced, early SPMS diagnosis may represent a window of opportunity for intervention. New approaches, endpoints or technologies could help physicians establishing a diagnosis. Here, we review SPMS in relation to its diagnostic and definition challenges and current screening techniques and tools.
Collapse
|
114
|
Baydyuk M, Cha DS, Hu J, Yamazaki R, Miller EM, Smith VN, Kelly KA, Huang JK. Tracking the evolution of CNS remyelinating lesion in mice with neutral red dye. Proc Natl Acad Sci U S A 2019; 116:14290-14299. [PMID: 31235582 PMCID: PMC6628798 DOI: 10.1073/pnas.1819343116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Animal models of central nervous system (CNS) demyelination, including toxin-induced focal demyelination and immune-mediated demyelination through experimental autoimmune encephalomyelitis (EAE), have provided valuable insights into the mechanisms of neuroinflammation and CNS remyelination. However, the ability to track changes in transcripts, proteins, and metabolites, as well as cellular populations during the evolution of a focal lesion, has remained challenging. Here, we developed a method to label CNS demyelinating lesions by the intraperitoneal injection of a vital dye, neutral red (NR), into mice before killing. We demonstrate that NR-labeled lesions can be easily identified on the intact spinal cord in both lysolecithin- and EAE-mediated demyelination models. Using fluorescence microscopy, we detected NR in activated macrophages/microglia and astrocytes, but not in oligodendrocytes present in lesions. Importantly, we successfully performed RT-qPCR, Western blot, flow cytometry, and mass spectrometry analysis of precisely dissected NR-labeled lesions at 5, 10, and 20 d postlesion (dpl) and found differential changes in transcripts, proteins, cell populations, and metabolites in lesions over the course of remyelination. Therefore, NR administration is a simple and powerful method to track and analyze the detailed molecular, cellular, and metabolic changes that occur within the lesion microenvironment over time following CNS injury. Furthermore, this method can be used to identify molecular and metabolic pathways that regulate neuroinflammation and remyelination and facilitate the development of therapies to promote repair in demyelinating disorders such as multiple sclerosis.
Collapse
Affiliation(s)
- Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC 20057
| | - David S Cha
- Department of Biology, Georgetown University, Washington, DC 20057
| | - Jingwen Hu
- Department of Biology, Georgetown University, Washington, DC 20057
| | - Reiji Yamazaki
- Department of Biology, Georgetown University, Washington, DC 20057
| | - Evan M Miller
- Department of Biology, Georgetown University, Washington, DC 20057
| | - Victoria N Smith
- Department of Biology, Georgetown University, Washington, DC 20057
| | | | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC 20057;
- Center for Cell Reprogramming, Georgetown University, Washington, DC 20057
| |
Collapse
|
115
|
Miller ED, Dziedzic A, Saluk-Bijak J, Bijak M. A Review of Various Antioxidant Compounds and their Potential Utility as Complementary Therapy in Multiple Sclerosis. Nutrients 2019; 11:nu11071528. [PMID: 31284389 PMCID: PMC6682972 DOI: 10.3390/nu11071528] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a complex disease of the central nervous system (CNS). The etiology of this multifactorial disease has not been clearly defined. Conventional medical treatment of MS has progressed, but is still based on symptomatic treatment. One of the key factors in the pathogenesis of MS is oxidative stress, enhancing inflammation and neurodegeneration. In MS, both reactive oxygen and nitrogen species are formed in the CNS mainly by activated macrophages and microglia structures, which can lead to demyelination and axon disruption. The course of MS is associated with the secretion of many inflammatory and oxidative stress mediators, including cytokines (IL-1b, IL-6, IL-17, TNF-α, INF-γ) and chemokines (MIP-1a, MCP-1, IP10). The early stage of MS (RRMS) lasts about 10 years, and is dominated by inflammatory processes, whereas the chronic stage is associated with neurodegenerative axon and neuron loss. Since oxidative damage has been known to be involved in inflammatory and autoimmune-mediated processes, antioxidant therapy could contribute to the reduction or even prevention of the progression of MS. Further research is needed in order to establish new aims for novel treatment and provide possible benefits to MS patients. The present review examines the roles of oxidative stress and non-pharmacological anti-oxidative therapies in MS.
Collapse
Affiliation(s)
- Elzbieta Dorota Miller
- Department of Physical Medicine, Medical University of Lodz, Pl. Hallera 1, 90-647 Lodz, Poland
- Neurorehabilitation Ward, General Hospital no III, Milionowa 14, 90-001 Lodz, Poland
| | - Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Michal Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
116
|
|
117
|
DiSano KD, Linzey MR, Royce DB, Pachner AR, Gilli F. Differential neuro-immune patterns in two clinically relevant murine models of multiple sclerosis. J Neuroinflammation 2019; 16:109. [PMID: 31118079 PMCID: PMC6532235 DOI: 10.1186/s12974-019-1501-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/06/2019] [Indexed: 01/12/2023] Open
Abstract
Background The mechanisms driving multiple sclerosis (MS), the most common cause of non-traumatic disability in young adults, remain unknown despite extensive research. Especially puzzling are the underlying molecular processes behind the two major disease patterns of MS: relapsing-remitting and progressive. The relapsing-remitting course is exemplified by acute inflammatory attacks, whereas progressive MS is characterized by neurodegeneration on a background of mild-moderate inflammation. The molecular and cellular features differentiating the two patterns are still unclear, and the role of inflammation during progressive disease is a subject of active debate. Methods We performed a comprehensive analysis of the intrathecal inflammation in two clinically distinct mouse models of MS: the PLP139-151-induced relapsing experimental autoimmune encephalomyelitis (R-EAE) and the chronic progressive, Theiler’s murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). Microarray technology was first used to examine global gene expression changes in the spinal cord. Inflammation in the spinal cord was further assessed by immunohistochemical image analysis and flow cytometry. Levels of serum and cerebrospinal fluid (CSF) immunoglobulin (Ig) isotypes and chemokines were quantitated using Luminex Multiplex technology, whereas a capture ELISA was used to measure serum and CSF albumin levels. Finally, an intrathecal Ig synthesis index was established with the ratio of CSF and serum test results corrected as a ratio of their albumin concentrations. Results Microarray analysis identified an enrichment of B cell- and Ig-related genes upregulated in TMEV-IDD mice. We also demonstrated an increased level of intrathecal Ig synthesis as well as a marked infiltration of late differentiated B cells, including antibody secreting cells (ASC), in the spinal cord of TMEV-IDD, but not R-EAE mice. An intact blood-brain barrier in TMEV-IDD mice along with higher CSF levels of CXCL13, CXCL12, and CCL19 provides evidence for an intrathecal synthesis of chemokines mediating B cell localization to the central nervous system (CNS). Conclusions Overall, these findings, showing increased concentrations of intrathecally produced Igs, substantial infiltration of ASC, and the presence of B cell supporting chemokines in the CNS of TMEV-IDD mice, but not R-EAE mice, suggest a potentially important role for Igs and ASC in the chronic progressive phase of demyelinating diseases. Electronic supplementary material The online version of this article (10.1186/s12974-019-1501-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Krista D DiSano
- Department of Neurology, Dartmouth Hitchcock Medical Center and Geisel School of Medicine, One Medical Center Drive, Lebanon, NH, 03756, USA
| | - Michael R Linzey
- Department of Neurology, Dartmouth Hitchcock Medical Center and Geisel School of Medicine, One Medical Center Drive, Lebanon, NH, 03756, USA.,Program in Experimental and Molecular Medicine, Dartmouth College, Hanover, NH, USA
| | - Darlene B Royce
- Department of Neurology, Dartmouth Hitchcock Medical Center and Geisel School of Medicine, One Medical Center Drive, Lebanon, NH, 03756, USA
| | - Andrew R Pachner
- Department of Neurology, Dartmouth Hitchcock Medical Center and Geisel School of Medicine, One Medical Center Drive, Lebanon, NH, 03756, USA
| | - Francesca Gilli
- Department of Neurology, Dartmouth Hitchcock Medical Center and Geisel School of Medicine, One Medical Center Drive, Lebanon, NH, 03756, USA.
| |
Collapse
|
118
|
Herbert LB, Zerkowski K, O'Brien S, Leonard KV, Bhowmick A. Impact on interpersonal relationships among patients with multiple sclerosis and their partners. Neurodegener Dis Manag 2019; 9:173-187. [PMID: 31116077 PMCID: PMC6609896 DOI: 10.2217/nmt-2018-0045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Aim: To determine how the diagnosis and symptoms of multiple sclerosis (MS) impact interpersonal relationships. Participants & methods: Data were obtained from a convenience sample of 1010 individuals with MS responding to an online survey; responses were compared with results of a fibromyalgia survey. Results: MS participants had few negative perceptions of relationships with romantic partners/children/close friends but described the impact of fatigue and limited mobility as feelings of being not well understood by others, burdensome and isolated. Conclusion: Despite increasing disability/pain severity, most patients with MS reported high satisfaction levels and few negative perceptions of relationships with romantic partners/children/close friends. Future research should focus on identifying effective coping strategies used by satisfied couples and improving access to high-quality relationship-enrichment programs.
Collapse
Affiliation(s)
- Leslie Beth Herbert
- Research & Analytics, Health Union LLC, 1 International Plaza Dr #550, Philadelphia, PA, USA
| | - Kristine Zerkowski
- Community Development, Health Union LLC, 1 International Plaza Dr #550, Philadelphia, PA, USA
| | | | | | - Amrita Bhowmick
- Community Development, Health Union LLC, 1 International Plaza Dr #550, Philadelphia, PA, USA.,Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
119
|
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: 1.0] [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
|
120
|
Prolonged cortical silent period is related to poor fitness and fatigue, but not tumor necrosis factor, in Multiple Sclerosis. Clin Neurophysiol 2019; 130:474-483. [PMID: 30771724 DOI: 10.1016/j.clinph.2018.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/08/2018] [Accepted: 12/21/2018] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Poor fitness among people with Multiple Sclerosis (MS) aggravates disease symptoms. Whether low fitness levels accompany brain functioning changes is unknown. METHODS MS patients (n = 82) completed a graded maximal exercise test, blood was drawn, and transcranial magnetic stimulation determined resting and active motor thresholds, motor evoked potential latency, and cortical silent period (CSP). RESULTS Sixty-two percent of participants had fitness levels ranked below 10th percentile. Fitness was not associated with disability measured using the Expanded Disability Status Scale (EDSS). Regression analyses revealed that, cardiorespiratory fitness, when controlling for disease demographics, contributed 23.7% (p < 0.001) to the model explaining variance in CSP. Regression analysis using cardiorespiratory fitness and CSP as predictors showed that CSP alone explained 19.9% of variance in subjective fatigue (p = 0.002). Tumor necrosis factor was not associated with any variable. CONCLUSION Low fitness was associated with longer CSP in MS. Longer CSP was, in turn, related to greater MS fatigue. SIGNIFICANCE MS patients had extremely low levels of cardiorespiratory fitness. Poor fitness predicted longer CSP, a marker of greater intracortical inhibition, which was linked to MS fatigue. Future research should examine whether aerobic training could shorten CSP and potentially lessen inhibition of cortical networks.
Collapse
|
121
|
de Sousa CNS, da Silva Leite CMG, da Silva Medeiros I, Vasconcelos LC, Cabral LM, Patrocínio CFV, Patrocínio MLV, Mouaffak F, Kebir O, Macedo D, Patrocínio MCA, Vasconcelos SMM. Alpha-lipoic acid in the treatment of psychiatric and neurological disorders: a systematic review. Metab Brain Dis 2019; 34:39-52. [PMID: 30467770 DOI: 10.1007/s11011-018-0344-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/12/2018] [Indexed: 01/07/2023]
Abstract
Despite the existence of many preclinical studies, scientific evidence is lacking on the clinical use of alpha-lipoic acid (ALA) for central nervous system disorders. Therefore, we aimed at revising the literature concerning the use of ALA for the treatment of psychiatric and neurological conditions and to point out what is missing for the introduction of this antioxidant to this purpose. For this systematic review we performed a search using PubMed and SCOPUS databases with the following keywords: "alpha-Lipoic Acid AND central nervous system OR psychiatric disorders OR neurological disorders OR mood disorders OR anxiety OR psychosis OR Alzheimer OR Parkinson OR stroke". The total number of references found after automatically and manually excluding duplicates was 1061. After primary and secondary screening 32 articles were selected. Regarding psychiatric disorders, the studies of ALA in schizophrenia are advanced being ALA administration related to the improvement of schizophrenia symptoms and side effects of antipsychotic medication. In neurological disorders, ALA as a supplement was effective in the prevention of Alzheimer disease progression. For stroke, the use of the supplement ALAnerv® (containing 300 mg ALA) presented important results, since it was observed a reversal of clinical parameters and oxidative imbalance in these patients. For other neurological conditions, such as encephalopathy, multiple sclerosis, traumatic brain injury, mitochondrial disorders and migraine, the results are still preliminary. Overall, there is a need of well-designed clinical trials to enhance the clinical evidences of ALA effects for the treatment of neurological and psychiatric conditions.
Collapse
Affiliation(s)
- Caren Nádia Soares de Sousa
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1127, Fortaleza, CE, 60431-270, Brazil
| | - Cláudio Manuel Gonçalves da Silva Leite
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1127, Fortaleza, CE, 60431-270, Brazil
| | - Ingridy da Silva Medeiros
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1127, Fortaleza, CE, 60431-270, Brazil
| | - Luna Costa Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1127, Fortaleza, CE, 60431-270, Brazil
| | - Lucas Moraes Cabral
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1127, Fortaleza, CE, 60431-270, Brazil
| | | | | | - Fayçal Mouaffak
- Department of Seine Saint Denis, Ville Evrard Psychiatric Hospital, Paris, France
| | - Oussama Kebir
- Laboratory of Pathophysiology of Psychiatric Diseases, Center for Psychiatry and Neurosciences, INSERM U894, University Paris Descartes, Paris, France
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1127, Fortaleza, CE, 60431-270, Brazil
| | - Manoel Cláudio Azevedo Patrocínio
- School of Medicine, University Center Christus-Unichristus, Fortaleza, Ceará, Brazil
- Department of Anesthesiology, Dr. Jose Frota Institute Hospital/IJF, Fortaleza, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1127, Fortaleza, CE, 60431-270, Brazil.
| |
Collapse
|
122
|
Chaves AR, Wallack EM, Kelly LP, Pretty RW, Wiseman HD, Chen A, Moore CS, Stefanelli M, Ploughman M. Asymmetry of Brain Excitability: A New Biomarker that Predicts Objective and Subjective Symptoms in Multiple Sclerosis. Behav Brain Res 2019; 359:281-291. [DOI: 10.1016/j.bbr.2018.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/24/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022]
|
123
|
Health Related Quality of Life Among Patients With Multiple Sclerosis: The Role of Psychosocial Adjustment to Illness. Arch Psychiatr Nurs 2019; 33:11-16. [PMID: 30663613 DOI: 10.1016/j.apnu.2018.08.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/21/2018] [Accepted: 08/04/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Multiple Sclerosis (MS) is associated with poor quality of life (QOL). Individuals suffering from MS must make multiple adjustments as their condition changes. To date, little is known about the role of psychosocial adjustment in improving QOL of patients with MS. PURPOSE The purpose of this study is to identify the relationship between psychosocial adjustment and HRQOL controlling for demographic variables among patients with MS. METHODS This study used a descriptive-correlational design. A sample of 160 patients from two hospitals participated in the study. Self-reported data were collected using the demographic survey, Multiple Sclerosis Quality of Life (MSQoL-54) tool and Psychosocial Adjustment to Illness Scale-Self Report (PAIS-SR). RESULTS Participants reported poor QOL and difficulty with psychosocial adjustment. The QOL and psychosocial adjustment were correlated with various demographic variables. After controlling for demographic variables, psychosocial adjustment explained a large variance in the mental health composite of QOL (r square change = 44%) and the physical health composite of QOL = (r square change = 38%). CONCLUSION Psychosocial care could play a vital role in improving quality of life among MS patients.
Collapse
|
124
|
Arashloo FT, Hanzaei FF, Sedighi B, Amjad G, Younesi L. Efficacy of diffusion-weighted imaging in symptomatic and asymptomatic multiple sclerotic plaques. J Family Med Prim Care 2019; 8:2409-2413. [PMID: 31463267 PMCID: PMC6691452 DOI: 10.4103/jfmpc.jfmpc_420_19] [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] [Indexed: 11/09/2022] Open
Abstract
Introduction: Magnetic resonance imaging (MRI) currently accompanies clinical findings in disease diagnosis, patients’ follow-up, assessment of drugs complications, and evaluation of treatment response. Although contrast-enhanced MRI (CE-MRI) is considered as the imaging modality of choice for multiple sclerosis (MS), due to disease chronicity, applying multiple doses of gadolinium-based contrast agents (GBCAs) increases the risk of nephrogenic syndrome in patients with acute (ARF) and chronic renal syndromes (CRF). Moreover, the effect of gadolinium on the fetus is not well-known in pregnant patients. Therefore, this study evaluates the possibility of replacing postcontrast images with physiologically based MRI sequences such as diffusion weighted imaging (DWI) and apparent diffusion coefficient (ADC). Method: We prospectively evaluated 26 patients with known multiple sclerosis. The patients with MS attacks and the asymptomatic patients who were referred for follow-up were enrolled. Conventional MRI including postcontrast T1W, DWI, and ADC were performed for all patients. The signal intensity (SI) of all enhancing and nonenhancing plaques of more than 10 × 10 mm size were investigated in all sequences and analyzed. Results: A total of 83 plaques were detected in T2-FLAIR sequences of which 51 plaques were enhanced (68%) after gadolinium administration. While 42 MS plaques had hypersignal intensity in DWI (56%), 32 plaques had iso- or hyposignal intensities in DWI (44%). No statistically significant values were obtained. Conclusion: Although DWI could not replace CE-MRI, using these two modalities together could increase detection of active MS plaques and alter patients’ therapy and prognosis.
Collapse
Affiliation(s)
| | - Farnaz Fahimi Hanzaei
- Clinical Research Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Behnaz Sedighi
- Neurology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ghazaleh Amjad
- Shahid Akbar Abadi Clinical Research Development Unit, Iran University of Medical Sciences, Tehran, Iran
| | - Ladan Younesi
- Shahid Akbar Abadi Clinical Research Development Unit, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
125
|
Shimizu F, Nishihara H, Kanda T. Blood-brain barrier dysfunction in immuno-mediated neurological diseases. Immunol Med 2018; 41:120-128. [PMID: 30938273 DOI: 10.1080/25785826.2018.1531190] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The blood-brain barrier (BBB) is the brain-specific endothelial cell barrier that is important for maintaining brain homeostasis and preventing the entry of toxic substances. Pathological BBB dysfunction is a critical step of the disease process in several immuno-mediated neurological diseases, including multiple sclerosis (MS), neuromyelitis optica (NMO), neuropsychiatric systemic lupus erythematosus (NPSLE) and neuro-Behçet diseases. The pathological findings from patients with secondary progressive (SP) MS, NMO and NPSLE showed leaky BBB in the active lesions. NMO is a disease with strong evidence of disease-specific and pathogenic autoantibodies (aquaporin 4 [AQP4] autoantibodies). In the development of NMO, circulating AQP4 autoantibodies need to pass through the BBB in order to reach AQP4 on the astrocyte endfeet. Strong evidence suggests that NPSLE is associated with the disruption of the BBB and NPSLE patients frequently have antibodies bound to endothelial cells in their sera. We recently identified two BBB-reactive autoantibodies in immuno-mediated neurological diseases: galectin-3 autoantibodies in SPMS and GRP78 autoantibodies in NMO. In the present review article, we describe the basic structure and cellular biology of the BBB, discuss recent insights regarding the pathophysiology of the BBB breakdown in the setting of immuno-mediated neurological diseases, and describe our recent findings of autoantibody-mediated BBB breakdown.
Collapse
Affiliation(s)
- Fumitaka Shimizu
- a Department of Neurology and Clinical Neuroscience , Yamaguchi University Graduate School of Medicine , Ube , Japan
| | - Hideaki Nishihara
- a Department of Neurology and Clinical Neuroscience , Yamaguchi University Graduate School of Medicine , Ube , Japan
| | - Takashi Kanda
- a Department of Neurology and Clinical Neuroscience , Yamaguchi University Graduate School of Medicine , Ube , Japan
| |
Collapse
|
126
|
LoPresti P. Silent Free Fall at Disease Onset: A Perspective on Therapeutics for Progressive Multiple Sclerosis. Front Neurol 2018; 9:973. [PMID: 30542317 PMCID: PMC6277889 DOI: 10.3389/fneur.2018.00973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Central nervous system (CNS) degeneration occurs during multiple sclerosis (MS) following several years of reversible autoimmune demyelination. Progressive CNS degeneration appears later during the course of relapsing-remitting MS (RRMS), although it starts insidiously at disease onset. We propose that there is an early subclinical phase also for primary-progressive (PP) MS. Consensus exists that many different cell types are involved during disease onset. Furthermore, the response to the initial damage, which is specific for each individual, would result in distinct pathological pathways that add complexity to the disease and the mechanisms underlying progressive CNS degeneration. Progressive MS is classified as either active or not active, as well as with or without progression. Different forms of progressive MS might reflect distinct or overlapping pathogenetic pathways. Disease mechanisms should be determined for each patient at diagnosis and the time of treatment. Until individualized and time-sensitive treatments that specifically target the molecular mechanisms of the progressive aspect of the disease are identified, combined therapies directed at anti-inflammation, regeneration, and neuroprotection are the most effective for preventing MS progression. This review presents selected therapeutics in support of the overall idea of a multidimensional therapy applied early in the disease. This approach could limit damage and increase CNS repair. By targeting several cellular populations (i.e., microglia, astrocytes, neurons, oligodendrocytes, and lymphocytes) and multiple pathological processes (e.g., inflammation, demyelination, synaptopathy, and excitatory/inhibitory imbalance) progressive MS could be attenuated. Early timing for such multidimensional therapy is proposed as the prerequisite for effectively halting progressive MS.
Collapse
Affiliation(s)
- Patrizia LoPresti
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
127
|
Abstract
Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating and neurodegenerative disease of the central nervous system in young adults. This disorder is a heterogeneous, multifactorial, immune-mediated disease that is influenced by both genetic and environmental factors. In most patients, reversible episodes of neurological dysfunction lasting several days or weeks characterize the initial stages of the disease (that is, clinically isolated syndrome and relapsing-remitting MS). Over time, irreversible clinical and cognitive deficits develop. A minority of patients have a progressive disease course from the onset. The pathological hallmark of MS is the formation of demyelinating lesions in the brain and spinal cord, which can be associated with neuro-axonal damage. Focal lesions are thought to be caused by the infiltration of immune cells, including T cells, B cells and myeloid cells, into the central nervous system parenchyma, with associated injury. MS is associated with a substantial burden on society owing to the high cost of the available treatments and poorer employment prospects and job retention for patients and their caregivers.
Collapse
Affiliation(s)
- Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy. .,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.
| | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.,Neuroimmunology Unit, Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Paolo Preziosa
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandra Solari
- Unit of Neuroepidemiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-inflammation, Fondation Eugène Devic EDMUS Contre la Sclérose en Plaques, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
| | - Maria A Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.,Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
128
|
Mattay RR, Davtyan K, Bilello M, Mamourian AC. Do All Patients with Multiple Sclerosis Benefit from the Use of Contrast on Serial Follow-Up MR Imaging? A Retrospective Analysis. AJNR Am J Neuroradiol 2018; 39:2001-2006. [PMID: 30287455 DOI: 10.3174/ajnr.a5828] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/26/2018] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Patients with multiple sclerosis routinely have MR imaging with contrast every 6-12 months to assess response to medication. Multiple recent studies provide evidence of tissue deposition of MR imaging contrast agents, questioning the long-term safety of these agents. The goal of this retrospective image-analysis study was to determine whether contrast could be reserved for only those patients who show new MS lesions on follow-up examinations. MATERIALS AND METHODS We retrospectively reviewed brain MRIs of 138 patients. To increase our sensitivity, we used a previously described computerized image-comparison software to evaluate the stability or progression of multiple sclerosis white matter lesions in noncontrast FLAIR sequences. We correlated these findings with evidence of contrast-enhancing lesions on the enhanced T1 sequence from the same scan. RESULTS Thirty-three scans showed an increase in white matter lesion burden. Among those 33 patients, 14 examinations also demonstrated enhancing new lesions. While we found a single example of enhancement of a pre-existing white matter lesion that appeared unchanged in size, that same examination showed an overall increase in lesion burden with enhancement of other, new lesions. Thus, we found that all patients with enhancing lesions had evidence of progression on their noncontrast imaging. CONCLUSIONS Because all enhancing lesions were associated with new lesions on unenhanced imaging and progression was only evident in 24% of patients, in patients with relapsing-remitting MS, it is reasonable to consider reserving contrast for only those patients with evidence of progression on noncontrast MR images.
Collapse
Affiliation(s)
- R R Mattay
- From the Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.
| | - K Davtyan
- From the Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - M Bilello
- From the Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - A C Mamourian
- From the Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
129
|
Fayyazi E, Shaygannejad V, Maljaie MB, Mirmosayyeb O, Badihian S, Moosavian SP. Association Between Sunlight Exposure and Vitamin D Intake and Multiple Sclerosis Disability and Progression. CASPIAN JOURNAL OF NEUROLOGICAL SCIENCES 2018. [DOI: 10.29252/cjns.4.14.114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
130
|
Stem Cells as Potential Targets of Polyphenols in Multiple Sclerosis and Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1483791. [PMID: 30112360 PMCID: PMC6077677 DOI: 10.1155/2018/1483791] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) and multiple sclerosis are major neurodegenerative diseases, which are characterized by the accumulation of abnormal pathogenic proteins due to oxidative stress, mitochondrial dysfunction, impaired autophagy, and pathogens, leading to neurodegeneration and behavioral deficits. Herein, we reviewed the utility of plant polyphenols in regulating proliferation and differentiation of stem cells for inducing brain self-repair in AD and multiple sclerosis. Firstly, we discussed the genetic, physiological, and environmental factors involved in the pathophysiology of both the disorders. Next, we reviewed various stem cell therapies available and how they have proved useful in animal models of AD and multiple sclerosis. Lastly, we discussed how polyphenols utilize the potential of stem cells, either complementing their therapeutic effects or stimulating endogenous and exogenous neurogenesis, against these diseases. We suggest that polyphenols could be a potential candidate for stem cell therapy against neurodegenerative disorders.
Collapse
|
131
|
Li R, Patterson KR, Bar-Or A. Reassessing B cell contributions in multiple sclerosis. Nat Immunol 2018; 19:696-707. [PMID: 29925992 DOI: 10.1038/s41590-018-0135-x] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
There is growing recognition that B cell contributions to normal immune responses extend well beyond their potential to become antibody-producing cells, including roles at the innate-adaptive interface and their potential to modulate the responses of other immune cells such as T cells and myeloid cells. These B cell functions can have both pathogenic and protective effects in the context of central nervous system (CNS) inflammation. Here, we review recent advances in the field of multiple sclerosis (MS), which has traditionally been viewed as primarily a T cell-mediated disease, and we consider antibody-dependent and, particularly, emerging antibody-independent functions of B cells that may be relevant in both the peripheral and CNS disease compartments.
Collapse
Affiliation(s)
- Rui Li
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina R Patterson
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics (CNET) and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
132
|
Tiftikcioglu BI, Ilgezdi I, Zorlu Y, Sener U, Tokucoglu F. Long-term disability and progression in spinal onset multiple sclerosis. Acta Neurol Belg 2018; 118:217-225. [PMID: 28812249 DOI: 10.1007/s13760-017-0828-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/28/2017] [Indexed: 11/30/2022]
Abstract
The aim of this study is to investigate the long-term effects of the initial spinal cord (SC) involvement in MS patients. In this retrospective, single-center study, 824 patients with definite MS were screened. A total of 348 patients were excluded for ambiguous documentation of the initial relapse, pediatric onset, diagnosis of primary progressive disease, irregular assessments or visits causing doubt on the onset of progression time, and clinical follow-up duration less than 12 months. Eventually, 476 MS patients were included. Data regarding the demographics, initial symptoms, the degree of recovery from the initial relapse, neuroimaging, cerebrospinal fluid analysis, long-term disability, and progression were collected from the medical registry. The mean duration of follow-up was 7.49 ± 5.30 years. The percentage of patients entering the progressive disease course was 23.3 in the whole group. A total of 157 patients (33.0%) had SC involvement during the first clinical relapse. These patients were significantly older at disease onset (31.69 ± 10.18 vs. 29.55 ± 9.49; p = 0.028), had higher rates of progression (32.5 vs. 18.8%; p = 0.001), and had higher disability scores in long-term follow-up (3.41 ± 2.19 vs. 2.62 ± 1.81; p < 0.001). Mean age at the transition of progressive phase was 41.4 ± 11.2 years. The degree of recovery from the initial relapse significantly affected the long-term disability. The poor recovery from the initial relapse was associated with older onset age and higher EDSS scores. Being older than 40 years during MS onset and poor recovery from the initial relapse exerted an increased risk for progression. The initial SC involvement was related to a more severe relapse with less chance of complete recovery and higher risk for progression. Confirmation of risk factors in different MS cohorts would increase our understanding of the complex disease mechanisms.
Collapse
Affiliation(s)
- Bedile Irem Tiftikcioglu
- Department of Neurology, TCSB Izmir Tepecik Education and Research Hospital, Gaziler caddesi No: 468, Yenisehir, Konak, 35170, Izmir, Turkey.
| | - Irem Ilgezdi
- Department of Neurology, TCSB Izmir Tepecik Education and Research Hospital, Gaziler caddesi No: 468, Yenisehir, Konak, 35170, Izmir, Turkey
| | - Yasar Zorlu
- Department of Neurology, TCSB Izmir Tepecik Education and Research Hospital, Gaziler caddesi No: 468, Yenisehir, Konak, 35170, Izmir, Turkey
| | - Ufuk Sener
- Department of Neurology, TCSB Izmir Tepecik Education and Research Hospital, Gaziler caddesi No: 468, Yenisehir, Konak, 35170, Izmir, Turkey
| | - Figen Tokucoglu
- Department of Neurology, TCSB Izmir Tepecik Education and Research Hospital, Gaziler caddesi No: 468, Yenisehir, Konak, 35170, Izmir, Turkey
| |
Collapse
|
133
|
Dopkins N, Nagarkatti PS, Nagarkatti M. The role of gut microbiome and associated metabolome in the regulation of neuroinflammation in multiple sclerosis and its implications in attenuating chronic inflammation in other inflammatory and autoimmune disorders. Immunology 2018; 154:178-185. [PMID: 29392733 PMCID: PMC5980216 DOI: 10.1111/imm.12903] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 02/06/2023] Open
Abstract
The importance of the gut microbiome in the regulation of non-infectious diseases has earned unprecedented interest from biomedical researchers. Widespread use of next-generation sequencing techniques has prepared a foundation for further research by correlating the presence of specific bacterial species with the onset or severity of a disease state, heralding paradigm-shifting results. This review covers the mechanisms through which a dysbiotic gut microbiota contributes to the pathological symptoms in an autoimmune neurodegenerative disorder, multiple sclerosis (MS). Although the central nervous system (CNS) is protected by the blood-brain barrier (BBB), it is unclear how gut dysbiosis can trigger potential immunological changes in the CNS in the presence of the BBB. This review focuses on the immunoregulatory functionality of microbial metabolites, which can cross the BBB and mediate their effects directly on immune cells within the CNS and/or indirectly through modulating the response of peripheral T cells to stimulate or inhibit pro-inflammatory chemokines and cytokines, which in turn regulate the autoimmune response in the CNS. Although more research is clearly needed to directly link the changes in gut microbiome with neuroinflammation, focusing research on microbiota that produce beneficial metabolites with the ability to attenuate chronic inflammation systemically as well as in the CNS, can offer novel preventive and therapeutic modalities against a wide array of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Nicholas Dopkins
- Department of Pathology, Microbiology and ImmunologyUniversity of South Carolina School of MedicineColumbiaSCUSA
| | - Prakash S. Nagarkatti
- Department of Pathology, Microbiology and ImmunologyUniversity of South Carolina School of MedicineColumbiaSCUSA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and ImmunologyUniversity of South Carolina School of MedicineColumbiaSCUSA
| |
Collapse
|
134
|
Huwiler A, Zangemeister-Wittke U. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: Recent findings and new perspectives. Pharmacol Ther 2018; 185:34-49. [DOI: 10.1016/j.pharmthera.2017.11.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
135
|
Sanchez MA, Sullivan GM, Armstrong RC. Genetic detection of Sonic hedgehog (Shh) expression and cellular response in the progression of acute through chronic demyelination and remyelination. Neurobiol Dis 2018; 115:145-156. [PMID: 29627579 DOI: 10.1016/j.nbd.2018.04.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/26/2018] [Accepted: 04/01/2018] [Indexed: 01/20/2023] Open
Abstract
Multiple sclerosis is a demyelinating disease in which neurological deficits result from damage to myelin, axons, and neuron cell bodies. Prolonged or repeated episodes of demyelination impair remyelination. We hypothesized that augmenting Sonic hedgehog (Shh) signaling in chronically demyelinated lesions could enhance oligodendrogenesis and remyelination. Shh regulates oligodendrocyte development during postnatal myelination, and maintains adult neural stem cells. We used genetic approaches to detect Shh expression and Shh responding cells in vivo. ShhCreERT2 or Gli1CreERT2 mice were crossed to reporter mice for genetic fate-labeling of cells actively transcribing Shh or Gli1, an effective readout of canonical Shh signaling. Tamoxifen induction enabled temporal control of recombination at distinct stages of acute and chronic cuprizone demyelination of the corpus callosum. Gli1 fate-labeled cells were rarely found in the corpus callosum with tamoxifen given during acute demyelination stages to examine activated microglia, reactive astrocytes, or remyelinating cells. Gli1 fate-labeled cells, mainly reactive astrocytes, were observed in the corpus callosum with tamoxifen given after chronic demyelination. However, Shh expressing cells were not detected in the corpus callosum during acute or chronic demyelination. Finally, SAG, an agonist of both canonical and type II non-canonical Hedgehog signaling pathways, was microinjected into the corpus callosum after chronic demyelination. Significantly, SAG delivery increased proliferation and enhanced remyelination. SAG did not increase Gli1 fate-labeled cells in the corpus callosum, which may indicate signaling through the non-canonical Hedgehog pathway. These studies demonstrate that Hedgehog pathway interventions may have therapeutic potential to modulate astrogliosis and to promote remyelination after chronic demyelination.
Collapse
Affiliation(s)
- Maria A Sanchez
- Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Genevieve M Sullivan
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Regina C Armstrong
- Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| |
Collapse
|
136
|
Metz LM, Liu WQ. Effective treatment of progressive MS remains elusive. Lancet 2018; 391:1239-1240. [PMID: 29576503 DOI: 10.1016/s0140-6736(18)30426-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 02/14/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Luanne M Metz
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 2T9, Canada.
| | - Wei-Qiao Liu
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 2T9, Canada
| |
Collapse
|
137
|
Mische LJ, Mowry EM. The Evidence for Dietary Interventions and Nutritional Supplements as Treatment Options in Multiple Sclerosis: a Review. Curr Treat Options Neurol 2018; 20:8. [PMID: 29549521 DOI: 10.1007/s11940-018-0494-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW This review aims to critically evaluate published studies examining diets and nutritional supplements (excepting vitamin D) for the impact on prevention and prognosis of multiple sclerosis (MS). RECENT FINDINGS There is a negative relationship between the Mediterranean diet and vascular disease, and vascular co-morbidities are associated with a worse MS prognosis. Low-fat, fish-based diets, sodium-restricted diets, calorie restriction, the paleo diet, and gluten-free diets have been examined, mostly in observational studies; results are inconclusive. With regard to nutritional supplements, pilot data show a possible benefit of biotin with respect to disability worsening in people with progressive MS (PMS). The best designed randomized controlled trials (RCTs) for PUFA supplementation have not shown significant impact, but several weaker RCTs have. Many other nutritional supplements have been tested, including several anti-oxidants. While some early studies show positive results, no result has been definitive. Unfortunately, there is no strong evidence for a direct benefit of any given dietary intervention on MS risk or prognosis. However, due to its relationship with vascular co-morbidities, the Mediterranean diet has the strongest rationale for employment in PwMS. Higher-quality clinical trials are needed to ascertain the possible benefits of nutritional supplements.
Collapse
Affiliation(s)
- Leah J Mische
- Johns Hopkins School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA.
| | - Ellen M Mowry
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21218, USA
| |
Collapse
|
138
|
Abstract
Multiple sclerosis (MS) is an inflammatory disorder targeting the central nervous system (CNS). The relapsing-remitting phase of MS is largely driven by peripheral activation of autoreactive T-helper (Th) 1 and Th17 lymphocytes. In contrast, compartmentalized inflammation within the CNS, including diffuse activation of innate myeloid cells, characterizes the progressive phase of MS, the most debilitating phase that currently lacks satisfactory treatments. Recently, bryostatin-1 (bryo-1), a naturally occurring, CNS-permeable compound with a favorable safety profile in humans, has been shown to act on antigen-presenting cells to promote differentiation of lymphocytes into Th2 cells, an action that might benefit Th1-driven inflammatory conditions such as MS. In the present study, we show that bryo-1 provides marked benefit in mice with experimental autoimmune encephalomyelitis (EAE), an experimental MS animal model. Preventive treatment with bryo-1 abolishes the onset of neurologic deficits in EAE. More strikingly, bryo-1 reverses neurologic deficits after EAE onset, even when treatment is initiated at a late stage of disease when peak adaptive immunity has subsided. Treatment with bryo-1 in vitro promotes an anti-inflammatory phenotype in antigen-presenting dendritic cells, macrophages, and to a lesser extent, lymphocytes. These findings suggest the potential for bryo-1 as a therapeutic agent in MS, particularly given its established clinical safety. Furthermore, the benefit of bryo-1, even in late treatment of EAE, combined with its targeting of innate myeloid cells suggests therapeutic potential in progressive forms of MS.
Collapse
|
139
|
Meijer KA, Eijlers AJC, Geurts JJG, Schoonheim MM. Staging of cortical and deep grey matter functional connectivity changes in multiple sclerosis. J Neurol Neurosurg Psychiatry 2018; 89:205-210. [PMID: 28986469 DOI: 10.1136/jnnp-2017-316329] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/31/2017] [Accepted: 09/13/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Functional connectivity is known to increase as well as decrease throughout the brain in multiple sclerosis (MS), which could represent different stages of the disease. In addition, functional connectivity changes could follow the atrophy pattern observed with disease progression, that is, moving from the deep grey matter towards the cortex. This study investigated when and where connectivity changes develop and explored their clinical and cognitive relevance across different MS stages. METHODS A cohort of 121 patients with early relapsing-remitting MS (RRMS), 122 with late RRMS and 53 with secondary progressive MS (SPMS) as well as 96 healthy controls underwent MRI and neuropsychological testing. Functional connectivity changes were investigated for (1) within deep grey matter connectivity, (2) connectivity between the deep grey matter and cortex and (3) within-cortex connectivity. A post hoc regional analysis was performed to identify which regions were driving the connectivity changes. RESULTS Patients with late RRMS and SPMS showed increased connectivity of the deep grey matter, especially of the putamen and palladium, with other deep grey matter structures and with the cortex. Within-cortex connectivity was decreased, especially for temporal, occipital and frontal regions, but only in SPMS relative to early RRMS. Deep grey matter connectivity alterations were related to cognition and disability, whereas within-cortex connectivity was only related to disability. CONCLUSION Increased connectivity of the deep grey matter became apparent in late RRMS and further increased in SPMS. The additive effect of cortical network degeneration, which was only seen in SPMS, may explain the sudden clinical deterioration characteristic to this phase of the disease.
Collapse
Affiliation(s)
- Kim A Meijer
- Department of Anatomy and Neurosciences, VUmc MS Center Amsterdam, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Anand J C Eijlers
- Department of Anatomy and Neurosciences, VUmc MS Center Amsterdam, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, VUmc MS Center Amsterdam, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- Department of Anatomy and Neurosciences, VUmc MS Center Amsterdam, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
140
|
Scott-Hewitt NJ, Folts CJ, Hogestyn JM, Piester G, Mayer-Pröschel M, Noble MD. Heterozygote galactocerebrosidase (GALC) mutants have reduced remyelination and impaired myelin debris clearance following demyelinating injury. Hum Mol Genet 2018; 26:2825-2837. [PMID: 28575206 DOI: 10.1093/hmg/ddx153] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/07/2017] [Indexed: 12/20/2022] Open
Abstract
Genome-wide association studies are identifying multiple genetic risk factors for several diseases, but the functional role of these changes remains mostly unknown. Variants in the galactocerebrosidase (GALC) gene, for example, were identified as a risk factor for Multiple Sclerosis (MS); however, the potential biological relevance of GALC variants to MS remains elusive. We found that heterozygote GALC mutant mice have reduced myelin debris clearance and diminished remyelination after a demyelinating insult. We found no histological or behavioral differences between adult wild-type and GALC +/- animals under normal conditions. Following exposure to the demyelinating agent cuprizone, however, GALC +/- animals had significantly reduced remyelination during recovery. In addition, the microglial phagocytic response and elevation of Trem2, both necessary for clearing damaged myelin, were markedly reduced in GALC +/- animals. These altered responses could be corrected in vitro by treatment with NKH-477, a compound discovered as protective in our previous studies on Krabbe disease, which is caused by mutations in both GALC alleles. Our data are the first to show remyelination defects in individuals with a single mutant GALC allele, suggesting such carriers may have increased vulnerability to myelin damage following injury or disease due to inefficient myelin debris clearance. We thus provide a potential functional link between GALC variants and increased MS susceptibility, particularly due to the failure of remyelination associated with progressive MS. Finally, this work demonstrates that genetic variants identified through genome-wide association studies may contribute significantly to complex diseases, not by driving initial symptoms, but by altering repair mechanisms.
Collapse
Affiliation(s)
- Nicole J Scott-Hewitt
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Christopher J Folts
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jessica M Hogestyn
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Gavin Piester
- Department of Biochemistry, University of Rochester, Rochester, NY 14642, USA
| | - Margot Mayer-Pröschel
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Mark D Noble
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
141
|
Scott-Hewitt NJ, Folts CJ, Noble MD. Heterozygous carriers of galactocerebrosidase mutations that cause Krabbe disease have impaired microglial function and defective repair of myelin damage. Neural Regen Res 2018; 13:393-401. [PMID: 29623914 PMCID: PMC5900492 DOI: 10.4103/1673-5374.228712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This review addresses two puzzling findings related to mutations in galactocerebrosidase (GALC) that cause Krabbe disease (KD), a severe lysosomal storage disorder characterized by extensive myelin damage in children with mutations in both GALC alleles. First, heterozygous carriers of KD-causing mutations, which include the biological parents of children with KD, exhibit increased risk for developing other diseases. Second, variants in the GALC locus increase the risk of developing multiple sclerosis (MS), another disease characterized by extensive myelin damage. What explains these correlations? In studies on cuprizone-induced myelin damage in heterozygous (GALC+/–) mice carrying one copy of a mutation that causes KD-like disease, the extent of damage was similar in GALC+/– and wild-type (WT) mice. In contrast, GALC+/- mice had striking defects in repair of cuprizone-induced damage. We further found unexpected microglial defects in myelin debris clearance and in the ability to up-regulate the Trem2 microglial protein critical for debris uptake. These defects were rescued by exposure to a lysosomal re-acidifying drug discovered in our studies on KD, and which provides multiple clinically relevant benefits in the twitcher (GALC+/–) mouse model of KD. Thus, heterozygous GALC mutations cause effects on biological function that may help to understand the increased disease risk in heterozygous carriers of such mutations and to understand why GALC variations increase the risk of MS. Our findings indicate that while some genetic risk factors may contribute to complex diseases by increasing the risk of tissue damage, others may do so by compromising tissue repair.
Collapse
Affiliation(s)
- Nicole J Scott-Hewitt
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christopher J Folts
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Mark D Noble
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
142
|
Araújo SES, Mendonça HR, Wheeler NA, Campello-Costa P, Jacobs KM, Gomes FCA, Fox MA, Fuss B. Inflammatory demyelination alters subcortical visual circuits. J Neuroinflammation 2017; 14:162. [PMID: 28821276 PMCID: PMC5562979 DOI: 10.1186/s12974-017-0936-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/07/2017] [Indexed: 11/13/2022] Open
Abstract
Background Multiple sclerosis (MS) is an inflammatory demyelinating disease classically associated with axonal damage and loss; more recently, however, synaptic changes have been recognized as additional contributing factors. An anatomical area commonly affected in MS is the visual pathway; yet, changes other than those associated with inflammatory demyelination of the optic nerve, i.e., optic neuritis, have not been described in detail. Methods Adult mice were subjected to a diet containing cuprizone to mimic certain aspects of inflammatory demyelination as seen in MS. Demyelination and inflammation were assessed by real-time polymerase chain reaction and immunohistochemistry. Synaptic changes associated with inflammatory demyelination in the dorsal lateral geniculate nucleus (dLGN) were determined by immunohistochemistry, Western blot analysis, and electrophysiological field potential recordings. Results In the cuprizone model, demyelination was observed in retinorecipient regions of the subcortical visual system, in particular the dLGN, where it was found accompanied by microglia activation and astrogliosis. In contrast, anterior parts of the pathway, i.e., the optic nerve and tract, appeared largely unaffected. Under the inflammatory demyelinating conditions, as seen in the dLGN of cuprizone-treated mice, there was an overall decrease in excitatory synaptic inputs from retinal ganglion cells. At the same time, the number of synaptic complexes arising from gamma-aminobutyric acid (GABA)-generating inhibitory neurons was found increased, as were the synapses that contain the N-methyl-d-aspartate receptor (NMDAR) subunit GluN2B and converge onto inhibitory neurons. These synaptic changes were functionally found associated with a shift toward an overall increase in network inhibition. Conclusions Using the cuprizone model of inflammatory demyelination, our data reveal a novel form of synaptic (mal)adaption in the CNS that is characterized by a shift of the excitation/inhibition balance toward inhibitory network activity associated with an increase in GABAergic inhibitory synapses and a possible increase in excitatory input onto inhibitory interneurons. In addition, our data recognize the cuprizone model as a suitable tool in which to assess the effects of inflammatory demyelination on subcortical retinorecipient regions of the visual system, such as the dLGN, in the absence of overt optic neuritis.
Collapse
Affiliation(s)
- Sheila Espírito Santo Araújo
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.,Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Biologia, Programa de Neurociências, Universidade Federal Fluminense, Niterói, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Henrique Rocha Mendonça
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.,Instituto de Biologia, Programa de Neurociências, Universidade Federal Fluminense, Niterói, Brazil
| | - Natalie A Wheeler
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Paula Campello-Costa
- Instituto de Biologia, Programa de Neurociências, Universidade Federal Fluminense, Niterói, Brazil
| | - Kimberle M Jacobs
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Flávia C A Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michael A Fox
- Developmental and Translational Neurobiology Center, Virginia Tech Carilion Research Institute, Roanoke, VA, USA
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
143
|
Guo A, Niyazov A, Macaulay D, Terasawa E, Schmerold L, Wu EQ, Krieger S. Inpatient Admissions and Costs Associated with Persistent Use of Dalfampridine Extended-Release in Multiple Sclerosis: A Claims Database Analysis. J Manag Care Spec Pharm 2017. [PMID: 28650249 PMCID: PMC10398050 DOI: 10.18553/jmcp.2017.23.7.771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND While the clinical benefits of dalfampridine extended-release (D-ER) have been established in patients with multiple sclerosis (MS) through multiple clinical trials, there is limited real-world data on D-ER use, in particular the persistent use of D-ER, and associated acute care resource utilization and costs. OBJECTIVE To examine the real-world association of D-ER use and inpatient admissions and costs among patients with MS. METHODS This study was a retrospective observational claims analysis of the MarketScan database (April 2009-March 2014). Eligible patients consisted of adult enrollees aged 18-64 years who had (a) 12 months of continuous private plan enrollment preceding (baseline) and following (follow-up) the first D-ER claim; (b) ≥ 2 MS diagnosis codes with ≥ 1 during the baseline period; (c) ≥ 2 consecutive D-ER claims; and (d) no alternate gait-impairing etiologies during the baseline and follow-up periods. Patients were separated into 2 D-ER cohorts in the main analysis: persistent (≥ 360 days of D-ER supply) and nonpersistent (< 360 days of supply) users. Sensitivity analyses were conducted, examining additional breakdowns of days of supply within the nonpersistent cohort. Inpatient admissions (all-cause and MS-related) and health care expenditures were calculated and compared between the cohorts during follow-up using Wilcoxon rank-sum and chi-square tests. Regression models were conducted, controlling for age, sex, MS relapses, comorbidities, disease-modifying therapy use, and other baseline factors, including inpatient admissions and costs. RESULTS Of 1,598 eligible patients, 719 (45.0%) were persistent D-ER users, and 879 (55.0%) were nonpersistent D-ER users. The 2 cohorts had similar demographic and clinical characteristics, with mean (SD) ages of 51.0 (8.4) and 50.6 (8.6) years and were 71.3% and 66.6% female, respectively. Compared with nonpersistent D-ER use, persistent D-ER use was associated with lower odds of all-cause inpatient admissions (OR = 0.58, P = 0.010) and MS-related inpatient admissions (OR = 0.50, P = 0.004). Persistent use was also associated with lower inpatient expenditures for all-cause admissions ($669 vs. $1,515, P = 0.002) and MS-related admissions ($388 vs. $891, P = 0.008). CONCLUSIONS Persistent D-ER use was associated with significantly lower rates of all-cause and MS-related inpatient admissions and costs. DISCLOSURES Funding for this research and medical writing assistance was provided by Acorda Therapeutics. The study sponsor was involved in all stages of the study research and manuscript preparation. Guo and Niyazov were employees of Acorda Therapeutics at the time of this study and may own stock/stock options. Wu, Macaulay, Terasawa, and Schmerold are employees of Analysis Group, which received consultancy fees from Acorda Therapeutics for this project. Krieger was a consultant for Acorda Therapeutics for this project and has the following additional financial interests to report: consulting/advisory board work with Bayer, Biogen, EMD Serono, Novartis, Genentech, Genzyme, and Teva. Study concept and design were contributed by Guo, Niyazov, Macaulay, and Wu. Macaulay, Terasawa, Schmerold, and Wu helped prepare the data, and data interpretation was performed by Krieger, Guo, Niyazov, and Macaulay, along with Terasawa and Wu. The manuscript was written by Terasawa and Schmerold, along with Macaulay, and revised by all the authors. A portion of the current research was presented in poster format at the 2106 American Academy of Neurology Annual Meeting, which took place in Vancouver, BC, Canada, on April 15-21, 2016.
Collapse
Affiliation(s)
- Amy Guo
- 1 Acorda Therapeutics, Ardsley, New York
| | | | | | | | | | - Eric Q Wu
- 3 Analysis Group, Boston, Massachusetts
| | - Stephen Krieger
- 4 Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
144
|
Correale J, Gaitán MI, Ysrraelit MC, Fiol MP. Progressive multiple sclerosis: from pathogenic mechanisms to treatment. Brain 2017; 140:527-546. [PMID: 27794524 DOI: 10.1093/brain/aww258] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/18/2016] [Indexed: 12/30/2022] Open
Abstract
During the past decades, better understanding of relapsing-remitting multiple sclerosis disease mechanisms have led to the development of several disease-modifying therapies, reducing relapse rates and severity, through immune system modulation or suppression. In contrast, current therapeutic options for progressive multiple sclerosis remain comparatively disappointing and challenging. One possible explanation is a lack of understanding of pathogenic mechanisms driving progressive multiple sclerosis. Furthermore, diagnosis is usually retrospective, based on history of gradual neurological worsening with or without occasional relapses, minor remissions or plateaus. In addition, imaging methods as well as biomarkers are not well established. Magnetic resonance imaging studies in progressive multiple sclerosis show decreased blood-brain barrier permeability, probably reflecting compartmentalization of inflammation behind a relatively intact blood-brain barrier. Interestingly, a spectrum of inflammatory cell types infiltrates the leptomeninges during subpial cortical demyelination. Indeed, recent magnetic resonance imaging studies show leptomeningeal contrast enhancement in subjects with progressive multiple sclerosis, possibly representing an in vivo marker of inflammation associated to subpial demyelination. Treatments for progressive disease depend on underlying mechanisms causing central nervous system damage. Immunity sheltered behind an intact blood-brain barrier, energy failure, and membrane channel dysfunction may be key processes in progressive disease. Interfering with these mechanisms may provide neuroprotection and prevent disability progression, while potentially restoring activity and conduction along damaged axons by repairing myelin. Although most previous clinical trials in progressive multiple sclerosis have yielded disappointing results, important lessons have been learnt, improving the design of novel ones. This review discusses mechanisms involved in progressive multiple sclerosis, correlations between histopathology and magnetic resonance imaging studies, along with possible new therapeutic approaches.
Collapse
|
145
|
Ahn YH, Jeon SB, Chang CY, Goh EA, Kim SS, Kim HJ, Song J, Park EJ. Glatiramer acetate attenuates the activation of CD4 + T cells by modulating STAT1 and -3 signaling in glia. Sci Rep 2017; 7:40484. [PMID: 28094337 PMCID: PMC5240344 DOI: 10.1038/srep40484] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022] Open
Abstract
Interactions between immune effector cells of the central nervous system appear to directly or indirectly influence the progress/regression of multiple sclerosis (MS). Here, we report that glial STAT1 and -3 are distinctively phosphorylated following the interaction of activated lymphocytes and glia, and this effect is significantly inhibited by glatiramer acetate (GA), a disease-modifying drug for MS. GA also reduces the activations of STAT1 and -3 by MS-associated stimuli such as IFNγ or LPS in primary glia, but not neurons. Experiments in IFNγ- and IFNγ receptor-deficient mice revealed that GA-induced inhibitions of STAT signaling are independent of IFNγ and its receptor. Interestingly, GA induces the expression levels of suppressor of cytokine signaling-1 and -3, representative negative regulators of STAT signaling in glia. We further found that GA attenuates the LPS-triggered enhancement of IL-2, a highly produced cytokine in patients with active MS, in CD4+ T cells co-cultured with glia, but not in CD4+ T cells alone. Collectively, these results provide that activation of glial STATs is an essential event in the interaction between glia and T cells, which is a possible underlying mechanism of GA action in MS. These findings provide an insight for the development of targeted therapies against MS.
Collapse
Affiliation(s)
- Ye-Hyeon Ahn
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
- Dept.of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Sae-Bom Jeon
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
| | - Chi Young Chang
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
| | - Eun-Ah Goh
- Dept. of System Cancer Science, Graduate School of Cancer Science and Policy, Goyang, South Korea
| | - Sang Soo Kim
- Radiation Medicine Branch, National Cancer Center, Goyang, South Korea
| | - Ho Jin Kim
- Dept. of System Cancer Science, Graduate School of Cancer Science and Policy, Goyang, South Korea
- Dept. of Neurology, National Cancer Center, Goyang, South Korea
| | - Jaewhan Song
- Dept.of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Eun Jung Park
- Cancer Immunology Branch, National Cancer Center, Goyang, South Korea
- Dept. of System Cancer Science, Graduate School of Cancer Science and Policy, Goyang, South Korea
| |
Collapse
|
146
|
Lee A, Pike J, Edwards MR, Petrillo J, Waller J, Jones E. Quantifying the Benefits of Dimethyl Fumarate Over β Interferon and Glatiramer Acetate Therapies on Work Productivity Outcomes in MS Patients. Neurol Ther 2017; 6:79-90. [PMID: 28093681 PMCID: PMC5447554 DOI: 10.1007/s40120-016-0061-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Indexed: 01/01/2023] Open
Abstract
Introduction Dimethyl fumarate (DMF) is a novel oral therapy used for the treatment of relapse-remitting multiple sclerosis (RRMS). In two 2-year pivotal Phase 3 trials in patients with RRMS, DMF significantly reduced disease activity based on both clinical and magnetic resonance imaging (MRI) findings and demonstrated an acceptable safety profile. However, there is currently a lack of comparative data which explore the relationship between work productivity and health-related quality of life (HRQoL) outcomes in RRMS and how these differ among RRMS therapies, including DMF. Methods We explored this relationship through patient-reported data from the EuroQol Five-Dimensions (EQ-5D) tool, Work Productivity and Activity Impairment Questionnaire (WPAI), and the Hamburg Quality of Life Questionnaire in Multiple Sclerosis (HAQUAMS) using the Adelphi MS DSP® dataset. Results Our data demonstrated that patients receiving DMF experienced better outcomes, relative to patients receiving beta (β)interferons or glatiramer acetate, in all WPAI subscales [overall; average treatment effect (ATE) −13.92, 95% confidence interval (CI) −18.87 to −7.08; p < 0.001], EQ-5D (ATE +0.075, 95% Cl 0.014–0.136; p = 0.016) and HAQUAMS [ATE −0.45, 95% Cl −0.61 to −0.29; p < 0.001]. The EQ-5D and HAQUAMS were used with WPAI to determine the relationship between HRQoL outcomes and work productivity. Multiple linear regression analyses were performed, adjusting for age, sex, body mass index, ethnicity and number of comorbid conditions. Conclusions These data demonstrate that therapy with DMF was associated with increased work productivity and HRQoL for patients with RRMS and that these outcomes were consistently improved compared to outcomes with interferon and glatiramer acetate therapies.
Collapse
Affiliation(s)
- Andrew Lee
- Biogen, 225 Binney Street, Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
147
|
Savarin C, Bergmann CC. Viral-induced suppression of self-reactive T cells: Lessons from neurotropic coronavirus-induced demyelination. J Neuroimmunol 2017; 308:12-16. [PMID: 28108025 PMCID: PMC5474352 DOI: 10.1016/j.jneuroim.2017.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/06/2017] [Accepted: 01/10/2017] [Indexed: 12/16/2022]
Abstract
Genetic and environmental factors, i.e. infections, have been proposed to contribute to disease induction and relapsing events in multiple sclerosis (MS), an autoimmune demyelinating disease of the central nervous system (CNS). While research has mainly focused on virus associated autoimmune activation, less is known about prevention of autoimmunity, especially following resolving infections associated with CNS tissue damage. This review discusses novel insights on control of self-reactive (SR) T cells activated during neurotropic coronavirus-induced demyelination. A new concept is introduced that SR T cells can be dampened by distinct regulatory mechanisms in the periphery and the CNS, thereby preventing autoimmune disease. Virus-induced demyelination activates myelin specific T cells. Virus-induced regulatory mechanisms limit pathogenic self-reactive R CD4 T cells. Self-reactive CD4 T cells are controlled by distinct mechanisms in the CLN and CNS.
Collapse
Affiliation(s)
- Carine Savarin
- Lerner Research Institute, Cleveland Clinic, Neuroscience Department NC-30, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| | - Cornelia C Bergmann
- Lerner Research Institute, Cleveland Clinic, Neuroscience Department NC-30, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
148
|
Garay L, Gonzalez Giqueaux P, Guennoun R, Schumacher M, Gonzalez Deniselle MC, De Nicola AF. Progesterone treatment modulates mRNA OF neurosteroidogenic enzymes in a murine model of multiple sclerosis. J Steroid Biochem Mol Biol 2017; 165:421-429. [PMID: 27597394 DOI: 10.1016/j.jsbmb.2016.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 11/20/2022]
Abstract
Previous studies of experimental autoimmune encephalomyelitis (EAE) have shown that progesterone decreases inflammatory cell infiltration and proinflammatory factors, increases myelination and attenuates clinical grade of EAE mice. To elucidate potential mediators of these effects, we analyzed the mRNA expression of neurosteroidogenic enzymes in the spinal cord, in view of the protective role of steroids in EAE. We also analyzed mitochondrial morphology and dynamics (fusion and fission proteins), considering the role of mitochondria in neurosteroidogenesis. EAE was induced in C57Bl6 mice using MOG40-54 and killed on day 16 after induction. Using qPCR, we found in steroid-untreated EAE mice decreased mRNAs for the steroidogenic acute regulatory protein (Star), voltage-dependent anion channel (VDAC), P450scc (cholesterol side-chain cleavage), 5α-reductase, 3α-hydroxysteroid dehydrogenase (3α-HSD) and aromatase, whereas levels of 3β-hydroxysteroid dehydrogenase (3β-HSD) showed a large intra-group variance. We also found increased mRNA expression of 18Kd translocator protein (TSPO), which likely resulted from the reactive microgliosis in this model. EAE mice also showed pathological mitochondrial morphology and reduced expression of fission and fusion protein mRNAs. Most importantly, pretreatment with progesterone a week before EAE induction increased Star,VDAC, P450scc, 5α-reductase type I, 3α-HSD and aromatase mRNAs and did not modify 3β-HSD. TSPO mRNA was decreased, consequent with the inhibition of microgliosis. Mitochondrial morphology was improved and fission/fusion protein mRNAs were enhanced by progesterone treatment. Furthermore, progesterone protective effects on mitochondrial and endoplasmic reticulum may allow the recovery of neurosteroidogenesis. In this way, endogenously synthesized neurosteroids may reinforce the beneficial effects of exogenous progesterone previously shown in MS mice.
Collapse
Affiliation(s)
- Laura Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry(,) Faculty of Medicine, University of Buenos Aires, Argentina
| | - Paula Gonzalez Giqueaux
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina
| | - Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - Maria Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Physiology, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental, Buenos Aires, Argentina; Dept. of Human Biochemistry(,) Faculty of Medicine, University of Buenos Aires, Argentina.
| |
Collapse
|
149
|
Psachoulia K, Chamberlain KA, Heo D, Davis SE, Paskus JD, Nanescu SE, Dupree JL, Wynn TA, Huang JK. IL4I1 augments CNS remyelination and axonal protection by modulating T cell driven inflammation. Brain 2016; 139:3121-3136. [PMID: 27797811 PMCID: PMC5382940 DOI: 10.1093/brain/aww254] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/01/2016] [Accepted: 08/22/2016] [Indexed: 01/01/2023] Open
Abstract
SEE PLUCHINO AND PERUZZOTTI-JAMETTI DOI101093/AWW266 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: Myelin regeneration (remyelination) is a spontaneous process that occurs following central nervous system demyelination. However, for reasons that remain poorly understood, remyelination fails in the progressive phase of multiple sclerosis. Emerging evidence indicates that alternatively activated macrophages in central nervous system lesions are required for oligodendrocyte progenitor differentiation into remyelinating oligodendrocytes. Here, we show that an alternatively activated macrophage secreted enzyme, interleukin-four induced one (IL4I1), is upregulated at the onset of inflammation resolution and remyelination in mouse central nervous system lesions after lysolecithin-induced focal demyelination. Focal demyelination in mice lacking IL4I1 or interleukin 4 receptor alpha (IL4Rα) results in increased proinflammatory macrophage density, remyelination impairment, and axonal injury in central nervous system lesions. Conversely, recombinant IL4I1 administration into central nervous system lesions reduces proinflammatory macrophage density, enhances remyelination, and rescues remyelination impairment in IL4Rα deficient mice. We find that IL4I1 does not directly affect oligodendrocyte differentiation, but modulates inflammation by reducing interferon gamma and IL17 expression in lesioned central nervous system tissues, and in activated T cells from splenocyte cultures. Remarkably, intravenous injection of IL4I1 into mice with experimental autoimmune encephalomyelitis at disease onset significantly reversed disease severity, resulting in recovery from hindlimb paralysis. Analysis of post-mortem tissues reveals reduced axonal dystrophy in spinal cord, and decreased CD4+ T cell populations in spinal cord and spleen tissues. These results indicate that IL4I1 modulates inflammation by regulating T cell expansion, thereby permitting the formation of a favourable environment in the central nervous system tissue for remyelination. Therefore, IL4I1 is a potentially novel therapeutic for promoting central nervous system repair in multiple sclerosis.
Collapse
Affiliation(s)
| | | | - Dongeun Heo
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Stephanie E Davis
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Jeremiah D Paskus
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Sonia E Nanescu
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Jeffrey L Dupree
- 2 Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Thomas A Wynn
- 3 Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey K Huang
- 1 Department of Biology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
150
|
Thalamus Degeneration and Inflammation in Two Distinct Multiple Sclerosis Animal Models. J Mol Neurosci 2016; 60:102-14. [PMID: 27491786 DOI: 10.1007/s12031-016-0790-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/21/2016] [Indexed: 12/31/2022]
Abstract
There is a broad consensus that multiple sclerosis (MS) represents more than an inflammatory disease: it harbors several characteristic aspects of a classical neurodegenerative disorder, i.e., damage to axons, synapses, and nerve cell bodies. While several accepted paraclinical methods exist to monitor the inflammatory-driven aspects of the disease, techniques to monitor progression of early and late neurodegeneration are still in their infancy and have not been convincingly validated. It was speculated that the thalamus with its multiple reciprocal connections is sensitive to inflammatory processes occurring in different brain regions, thus acting as a "barometer" for diffuse brain parenchymal damage in MS. To what extent the thalamus is affected in commonly applied MS animal models is, however, not known. In this article we describe direct and indirect damage to the thalamus in two distinct MS animal models. In the cuprizone model, we observed primary oligodendrocyte stress which is followed by demyelination, microglia/astrocyte activation, and acute axonal damage. These degenerative cuprizone-induced lesions were found to be more severe in the lateral compared to the medial part of the thalamus. In MOG35-55-induced EAE, in contrast, most parts of the forebrain, including the thalamus were not directly involved in the autoimmune attack. However, important thalamic afferent fiber tracts, such as the spinothalamic tract were inflamed and demyelinated on the spinal cord level. Quantitative immunohistochemistry revealed that this spinal cord inflammatory-demyelination is associated with neuronal loss within the target region of the spinothalamic tract, namely the sensory ventral posterolateral nucleus of the thalamus. This study highlights the possibility of trans-neuronal degeneration as one mechanism of secondary neuronal damage in MS. Further studies are now warranted to investigate involved cell types and cellular mechanisms.
Collapse
|