51
|
Yan J, McCombe PA, Pender MP, Greer JM. Reduced IκB-α Protein Levels in Peripheral Blood Cells of Patients with Multiple Sclerosis-A Possible Cause of Constitutive NF-κB Activation. J Clin Med 2020; 9:jcm9082534. [PMID: 32781504 PMCID: PMC7465818 DOI: 10.3390/jcm9082534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 11/16/2022] Open
Abstract
NF-κB signaling pathways are dysregulated in both the central nervous system (CNS) and peripheral blood cells in multiple sclerosis (MS), but the cause of this is unknown. We have recently reported that peripheral blood mononuclear cells (PBMC) of patients with MS have increased constitutive activation and translocation of the transcription factor NF-κB to the nucleus compared to healthy subjects. NF-κB can be activated through either canonical or non-canonical pathways. In the canonical pathway, activation of NF-κB is normally negatively regulated by the inhibitor IκB. We therefore hypothesized that the increased activation of NF-κB could be caused by reduced IκB-α in the cells of patients with MS, possibly due to increased activity of the IκB kinase (IKK) complex, which regulates IκB-α. Alternatively, changes to the activity of key molecules in the non-canonical pathway, such as IKKα, could also lead to increased NF-κB activation. We therefore used Western blotting to detect IκB-α levels and ELISA to investigate NF-κB DNA binding activity and phosphorylation of IKKα and IKKβ in samples from PBMC of MS patients and controls. The level of full-length IκB-α protein in the cytosolic fraction of PBMC of MS patients was significantly reduced compared to healthy subjects, with significantly more evidence of multiple low molecular weight putative degradation products of IκB-α present in MS patients compared to healthy subjects. Conversely, the level of NF-κB DNA binding activity was increased in whole cell lysates from MS patients. Both IKKα and IKKβ showed increased overall activity in MS compared to healthy subjects, although not all of the MS patients showed increased activity compared to the healthy subjects, suggesting that there may be several different mechanisms underlying the constitutive activation of NF-κB in MS. Taken together, these findings suggest that there may be multiple points at which the NF-κB pathway is dysregulated in MS and that decreased levels of the full-length IκB-α protein are a major component in this.
Collapse
Affiliation(s)
- Jun Yan
- UQ Centre for Clinical Research, The University of Queensland Centre for Clinical Research, Brisbane, QLD 4029, Australia; (J.Y.); (P.A.M.)
| | - Pamela A. McCombe
- UQ Centre for Clinical Research, The University of Queensland Centre for Clinical Research, Brisbane, QLD 4029, Australia; (J.Y.); (P.A.M.)
- Wesley Medical Research, The Wesley Hospital, Auchenflower, QLD 4066, Australia
| | - Michael P. Pender
- Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia;
- Department of Neurology, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
| | - Judith M. Greer
- UQ Centre for Clinical Research, The University of Queensland Centre for Clinical Research, Brisbane, QLD 4029, Australia; (J.Y.); (P.A.M.)
- Wesley Medical Research, The Wesley Hospital, Auchenflower, QLD 4066, Australia
- Correspondence: ; Tel.: +(61)-07-3346-6018
| |
Collapse
|
52
|
Serum levels of inflammasome pathway factors in clinically isolated syndrome and multiple sclerosis patients: a pilot study. Cent Eur J Immunol 2020; 45:237-240. [PMID: 33456338 PMCID: PMC7792443 DOI: 10.5114/ceji.2020.96877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 04/18/2018] [Indexed: 12/30/2022] Open
Abstract
Pathogenic roles of nuclear factor κB (NF-κB) pathway and NLRP3 inflammasome complex factors are involved in multiple sclerosis (MS) development. Activation of the NF-κB, NLRP3, and caspase-1 cascade results in production of proinflammatory cytokines that lead to stimulation of macrophages, lymphocytes, and glial cells. Although increased levels of inflammasome complex factors are observed in MS, contribution of inflammasome pathway to conversion from clinically isolated syndrome (CIS) to relapsing remitting MS (RRMS) has been scarcely investigated. To examine predictive value of inflammasome factors in CIS-MS conversion, levels of NLRP3, caspase-1, and NFκB are measured by ELISA in sera of age-gender matched CIS (n = 18; 8 converting, 10 non-converting) and RRMS (n = 23) patients. CIS and RRMS patients have comparable serum levels of NLRP3, caspase-1, and NFκB. Similarly, no statistically significant difference can be found among converting and non-converting CIS patients by means of inflammasome complex factor levels. Inflammasome factors are presumably overexpressed at early stages of MS. Therefore, they are unlikely to be used as biomarkers to predict CIS-MS conversion.
Collapse
|
53
|
Shabani Sadr NK, Shafiei M, Galehdari H, Khirolah A. The Effect of Sialic Acid on the Expression of miR-218, NF-kB, MMP-9, and TIMP-1. Biochem Genet 2020; 58:883-900. [PMID: 32607676 DOI: 10.1007/s10528-020-09981-y] [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: 08/27/2019] [Accepted: 06/17/2020] [Indexed: 01/29/2023]
Abstract
Sialic acid (N-acetylneuraminic acid, NANA) is found at all cell surfaces of vertebrates. Although it is widely accepted that sialic acid is an essential substrate for brain development via a significant role in nerve transfers, structure of glycosides, and synaptogenesis phenomena, there are some reports on the elevated levels of sialic acid and prevalence of neurodegeneration. Matrix metalloproteases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) are involved in neuroinflammation disorders and produced by many cell types, including activated T cells, macrophages, neurons, astrocytes, and microglial cells. It can be hypothesized that sialic acid may have a potentially critical role in regulation of a wide range of uncovered neurodegeneration factors as its downstream targets. In this study, for the first time, we aimed to analyze the possible effect of the sialic acid solution exposure in the human C118 cell line, which was derived from a human brain astrocytoma (glial cells), on the expression patterns of miR-218, NF-kB, MMP-9, and TIMP-1. For MMP-9, protein levels were studied too. Half maximal inhibitory concentration (IC50) value of NANA was obtained by MTT assay. Glial cell line was treated with sialic acid (300, 500, and 1000 µg/ml) for 24 h to investigate the effects of this ligand on the expression of miR-218, NF-kB, MMP-9, and TIMP-1 genes. Protein levels were checked by Western blotting, and by using zymography, the gelatinolytic activity of MMP-9 secreted into conditioned media was assayed. At 300 µM, 500 µM, and 1000 µM sialic acid treatments, the expression of miR-218 was downregulated; subsequently, the NF-kB, MMP-9, and TIMP-1 genes as well as their protein expressions were upregulated. More interestingly, the enzyme activity of secreted MMP-9 was upregulated too (p-values ≤ 0.05). This study could demonstrate the significant effect of sialic acid on miR-218, NF-kB, MMP-9 , and TIMP-1 expressions in gene and protein levels and also the levels of enzyme activity of secreted MMP-9. Therefore, provided information indicates the novel idea of a possible linkage between sialic acid species and regulation of these neuroinflammation genes in Glial cell line.
Collapse
Affiliation(s)
- Narjes Khatoun Shabani Sadr
- Department of Genetics, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Biotechnology and Bioscience Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohammad Shafiei
- Department of Genetics, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran. .,Biotechnology and Bioscience Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Hamid Galehdari
- Department of Genetics, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Biotechnology and Bioscience Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Alireza Khirolah
- Cellular and Molecular Research Center, Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| |
Collapse
|
54
|
Krysko KM, Graves JS, Dobson R, Altintas A, Amato MP, Bernard J, Bonavita S, Bove R, Cavalla P, Clerico M, Corona T, Doshi A, Fragoso Y, Jacobs D, Jokubaitis V, Landi D, Llamosa G, Longbrake EE, Maillart E, Marta M, Midaglia L, Shah S, Tintore M, van der Walt A, Voskuhl R, Wang Y, Zabad RK, Zeydan B, Houtchens M, Hellwig K. Sex effects across the lifespan in women with multiple sclerosis. Ther Adv Neurol Disord 2020; 13:1756286420936166. [PMID: 32655689 PMCID: PMC7331774 DOI: 10.1177/1756286420936166] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating central nervous system disorder that is more common in women, with onset often during reproductive years. The female:male sex ratio of MS rose in several regions over the last century, suggesting a possible sex by environmental interaction increasing MS risk in women. Since many with MS are in their childbearing years, family planning, including contraceptive and disease-modifying therapy (DMT) counselling, are important aspects of MS care in women. While some DMTs are likely harmful to the developing fetus, others can be used shortly before or until pregnancy is confirmed. Overall, pregnancy decreases risk of MS relapses, whereas relapse risk may increase postpartum, although pregnancy does not appear to be harmful for long-term prognosis of MS. However, ovarian aging may contribute to disability progression in women with MS. Here, we review sex effects across the lifespan in women with MS, including the effect of sex on MS susceptibility, effects of pregnancy on MS disease activity, and management strategies around pregnancy, including risks associated with DMT use before and during pregnancy, and while breastfeeding. We also review reproductive aging and sexual dysfunction in women with MS.
Collapse
Affiliation(s)
- Kristen M Krysko
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco, 675 Nelson Rising Lane, Suite 221, San Francisco, CA 94158, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, UCSD ACTRI, La Jolla, CA, USA
| | - Ruth Dobson
- Preventive Neurology Unit, Wolfson Institute of Preventive Neurology, Queen Mary University of London, London, UK
| | - Ayse Altintas
- Department of Neurology, School of Medicine, Koc University, Istanbul, Turkey
| | - Maria Pia Amato
- Department NEUROFARBA, Section of Neurosciences, University of Florence, Florence, Italy
| | - Jacqueline Bernard
- Department of Neurology, Oregon Health Science University, Portland, OR, USA
| | - Simona Bonavita
- Department of Advanced Medical and Surgical Sciences, University of Campania, "Luigi Vanvitelli", Naples, Italy
| | - Riley Bove
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco CA, USA
| | - Paola Cavalla
- Department of Neuroscience and Mental Health, City of Health and Science University Hospital of Torino, Turin, Italy
| | - Marinella Clerico
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Teresa Corona
- Clinical Laboratory of Neurodegenerative Disease, National Institute of Neurology and Neurosurgery of Mexico, Mexico City, Mexico
| | - Anisha Doshi
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK
| | - Yara Fragoso
- Multiple Sclerosis & Headache Research Institute, Santos, SP, Brazil
| | - Dina Jacobs
- Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Vilija Jokubaitis
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Doriana Landi
- Department of Systems Medicine, Multiple Sclerosis Center and Research Unit, Tor Vergata University and Hospital, Rome, Italy
| | | | | | | | - Monica Marta
- Neurosciences and Trauma Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Luciana Midaglia
- Department of Neurology-Neuroimmunology, Multiple Sclerosis Centre of Catalonia, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Suma Shah
- Department of Neurology, Duke University, Durham, NC, USA
| | - Mar Tintore
- Department of Neurology-Neuroimmunology, Multiple Sclerosis Centre of Catalonia, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Rhonda Voskuhl
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Yujie Wang
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Rana K Zabad
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Burcu Zeydan
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Maria Houtchens
- Department of Neurology, Partners MS Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerstin Hellwig
- Department of Neurology, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
55
|
Safa A, Arsang-Jang S, Taheri M, Omrani MD, Ghafouri-Fard S. Dysregulation of NF-κB-Associated lncRNAs in Multiple Sclerosis Patients. J Mol Neurosci 2020; 71:80-88. [DOI: 10.1007/s12031-020-01628-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/08/2020] [Indexed: 12/22/2022]
|
56
|
Fazia T, Nova A, Gentilini D, Beecham A, Piras M, Saddi V, Ticca A, Bitti P, McCauley JL, Berzuini C, Bernardinelli L. Investigating the Causal Effect of Brain Expression of CCL2, NFKB1, MAPK14, TNFRSF1A, CXCL10 Genes on Multiple Sclerosis: A Two-Sample Mendelian Randomization Approach. Front Bioeng Biotechnol 2020; 8:397. [PMID: 32432099 PMCID: PMC7216783 DOI: 10.3389/fbioe.2020.00397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Multiple Sclerosis (MS) exhibits considerable heterogeneity in phenotypic expression, course, prognosis and response to therapy. This suggests this disease involves multiple, as yet poorly understood, causal mechanisms. In this work we assessed the possible causal link between gene expression level of five selected genes related to the pro-inflammatory NF-κB signaling pathway (i.e., CCL2, NFKB1, MAPK14, TNFRSF1A, CXCL10) in ten different brain tissues (i.e., cerebellum, frontal cortex, hippocampus, medulla, occipital cortex, putamen, substantia nigra, thalamus, temporal cortex and intralobular white matter) and MS. We adopted a two-stage Mendelian Randomization (MR) approach for the estimation of the causal effects of interest, based on summary-level data from 20 multiplex Sardinian families and data provided by the United Kingdom Brain Expression Consortium (UKBEC). Through Radial-MR and Cochrane's Q statistics we identified and removed genetic variants which are most likely to be invalid instruments. To estimate the total causal effect, univariable MR was carried out separately for each gene and brain region. We used Inverse-Variance Weighted estimator (IVW) as main analysis and MR-Egger Regression estimator (MR-ER) and Weighted Median Estimator (WME) as sensitivity analysis. As these genes belong to the same pathway and thus they can be closely related, we also estimated their direct causal effects by applying IVW and MR-ER within a multivariable MR (MVMR) approach using set of genetic instruments specific and common (composite) to each multiple exposures represented by the expression of the candidate genes. Univariate MR analysis showed a significant positive total causal effect for CCL2 and NFKB1 respectively in medulla and cerebellum. MVMR showed a direct positive causal effect for NFKB1 and TNFRSF1A, and a direct negative causal effect for CCL2 in cerebellum; while in medulla we observed a direct positive causal effect for CCL2. Since in general we observed a different magnitude for the gene specific causal effect we hypothesize that in cerebellum and medulla the effect of each gene expression is direct but also mediated by the others. These results confirm the importance of the involvement of NF-κB signaling pathway in brain tissue for the development of the disease and improve our understanding in the pathogenesis of MS.
Collapse
Affiliation(s)
- Teresa Fazia
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Andrea Nova
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Davide Gentilini
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Milan, Italy
- Molecular Biology Laboratory, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Ashley Beecham
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL, United States
| | - Marialuisa Piras
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Valeria Saddi
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Anna Ticca
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Pierpaolo Bitti
- Centro di Tipizzazione Tissutale, S.I.T., Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Jacob L. McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL, United States
| | - Carlo Berzuini
- Centre for Biostatistics, University of Manchester, Manchester, United Kingdom
| | - Luisa Bernardinelli
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
57
|
Astrocyte and Oligodendrocyte Cross-Talk in the Central Nervous System. Cells 2020; 9:cells9030600. [PMID: 32138223 PMCID: PMC7140446 DOI: 10.3390/cells9030600] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/23/2022] Open
Abstract
Over the last decade knowledge of the role of astrocytes in central nervous system (CNS) neuroinflammatory diseases has changed dramatically. Rather than playing a merely passive role in response to damage it is clear that astrocytes actively maintain CNS homeostasis by influencing pH, ion and water balance, the plasticity of neurotransmitters and synapses, cerebral blood flow, and are important immune cells. During disease astrocytes become reactive and hypertrophic, a response that was long considered to be pathogenic. However, recent studies reveal that astrocytes also have a strong tissue regenerative role. Whilst most astrocyte research focuses on modulating neuronal function and synaptic transmission little is known about the cross-talk between astrocytes and oligodendrocytes, the myelinating cells of the CNS. This communication occurs via direct cell-cell contact as well as via secreted cytokines, chemokines, exosomes, and signalling molecules. Additionally, this cross-talk is important for glial development, triggering disease onset and progression, as well as stimulating regeneration and repair. Its critical role in homeostasis is most evident when this communication fails. Here, we review emerging evidence of astrocyte-oligodendrocyte communication in health and disease. Understanding the pathways involved in this cross-talk will reveal important insights into the pathogenesis and treatment of CNS diseases.
Collapse
|
58
|
Neuroinflammation in CNS diseases: Molecular mechanisms and the therapeutic potential of plant derived bioactive molecules. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100176] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
59
|
Vaknine S, Soreq H. Central and peripheral anti-inflammatory effects of acetylcholinesterase inhibitors. Neuropharmacology 2020; 168:108020. [PMID: 32143069 DOI: 10.1016/j.neuropharm.2020.108020] [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: 11/10/2019] [Revised: 02/09/2020] [Accepted: 02/25/2020] [Indexed: 12/31/2022]
Abstract
Acetylcholinesterase (AChE) inhibitors modulate acetylcholine hydrolysis and hence play a key role in determining the cholinergic tone and in implementing its impact on the cholinergic blockade of inflammatory processes. Such inhibitors may include rapidly acting small molecule AChE-blocking drugs and poisonous anti-AChE insecticides or war agent inhibitors which penetrate both body and brain. Notably, traumatized patients may be hyper-sensitized to anti-AChEs due to their impaired cholinergic tone, higher levels of circulation pro-inflammatory cytokines and exacerbated peripheral inflammatory responses. Those largely depend on the innate-immune system yet reach the brain via vagus pathways and/or disrupted blood-brain-barrier. Other regulators of the neuro-inflammatory cascade are AChE-targeted microRNAs (miRs) and synthetic chemically protected oligonucleotide blockers thereof, whose size prevents direct brain penetrance. Nevertheless, these larger molecules may exert parallel albeit slower inflammatory regulating effects on brain and body tissues. Additionally, oligonucleotide aptamers interacting with innate immune Toll-Like Receptors (TLRs) may control inflammation through diverse routes and in different rates. Such aptamers may compete with the action of both small molecule inhibitors and AChE-inhibiting miRs in peripheral tissues including muscle and intestine. However, rapid adaptation processes, visualized in neuromuscular junctions enable murine survival under otherwise lethal anti-cholinesterase exposure; and both miR inhibitors and TLR-modulating aptamers may exert body-brain signals protecting experimental mice from acute inflammation. The complex variety of AChE inhibiting molecules identifies diverse body-brain communication pathways which may rapidly induce long-lasting central reactions to peripheral stressful and inflammatory insults in both mice and men. This article is part of the special issue entitled 'Acetylcholinesterase Inhibitors: From Bench to Bedside to Battlefield'.
Collapse
Affiliation(s)
- Shani Vaknine
- The Edmond and Lily Safra Center of Brain Science, The Life Sciences Institute, The Hebrew University of Jerusalem, 9190401, Israel
| | - Hermona Soreq
- The Edmond and Lily Safra Center of Brain Science, The Life Sciences Institute, The Hebrew University of Jerusalem, 9190401, Israel.
| |
Collapse
|
60
|
Paricalcitol improves experimental autoimmune encephalomyelitis (EAE) by suppressing inflammation via NF-κB signaling. Biomed Pharmacother 2020; 125:109528. [PMID: 32106388 DOI: 10.1016/j.biopha.2019.109528] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/25/2022] Open
Abstract
Multiple sclerosis (MS) is known as an autoimmune disease in the central nervous system (CNS) characterized by motor deficits, pain, fatigue, cognitive impairment, and sensory and visual dysfunction. MS is considered to be resulted from significant inflammatory response. Paricalcitol (Pari) is a vitamin D2 analogue, which has been indicated to show anti-inflammatory activities in kidney and heart diseases. In the present study, if Pari could ameliorate the experimental autoimmune encephalomyelitis (EAE) was investigated. Here, the C57BL/6 mice were immunized using myelin oligodendrocyte glycoprotein 35-55 (MOG35-55). Subsequently, Pari was intraperitoneally injected into the mice. As for in vitro analysis, RAW264.7 and Jurkat cells were incubated with Pari together with corresponding stimulus. The results indicated that Pari administration reduced the paralytic severity, neuropathology and apoptosis in MOG-treated mice compared to the MOG single group. Pari also exhibited a significantly inhibitory effect on immune cell infiltration, glial cell activation, expression of pro-inflammatory factors and the activation of nuclear factor κB (NF-κB). The expression of pro-inflammatory regulators and the translocation of NF-κB from cytoplasm into nuclear in RAW264.7 and Jurkat cells under specific stimulation was clearly down-regulated by Pari incubation. Furthermore, we found that suppressing NF-κB with its inhibitor combined with Pari could further reduce the expression of pro-inflammatory factors and associated proteins. These data illustrated that Pari could diminish MOG-triggered EAE, as well as macrophages and T cells activation through blocking NF-κB activation. Collectively, Pari might have therapeutic effects in mouse models with MS.
Collapse
|
61
|
Balzano T, Arenas YM, Dadsetan S, Forteza J, Gil-Perotin S, Cubas-Nuñez L, Casanova B, Gracià F, Varela-Andrés N, Montoliu C, Llansola M, Felipo V. Sustained hyperammonemia induces TNF-a IN Purkinje neurons by activating the TNFR1-NF-κB pathway. J Neuroinflammation 2020; 17:70. [PMID: 32087723 PMCID: PMC7035786 DOI: 10.1186/s12974-020-01746-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with liver cirrhosis may develop hepatic encephalopathy. Rats with chronic hyperammonemia exhibit neurological alterations mediated by peripheral inflammation and neuroinflammation. Motor incoordination is due to increased TNF-a levels and activation of its receptor TNFR1 in the cerebellum. The aims were to assess (a) whether peripheral inflammation is responsible for TNF-a induction in hyperammonemic rats, (b) the cell type(s) in which TNF-a is increased, (c) whether this increase is associated with increased nuclear NF-κB and TNFR1 activation, (d) the time course of TNF-a induction, and (e) if TNF-a is induced in the Purkinje neurons of patients who die with liver cirrhosis. METHODS We analyzed the level of TNF-a mRNA and NF-κB in microglia, astrocytes, and Purkinje neurons in the cerebellum after 1, 2, and 4 weeks of hyperammonemia. We assessed whether preventing peripheral inflammation by administering an anti-TNF-a antibody prevents TNF-a induction. We tested whether TNF-a induction is reversed by R7050, which inhibits the TNFR1-NF-κB pathway, in ex vivo cerebellar slices. RESULTS Hyperammonemia induced microglial and astrocyte activation at 1 week. This was followed by TNF-a induction in both glial cell types at 2 weeks and in Purkinje neurons at 4 weeks. The level of TNF-a mRNA increased in parallel with the TNF-a protein level, indicating that TNF-a was synthesized in Purkinje cells. This increase was associated with increased NF-κB nuclear translocation. The nuclear translocation of NF-κB and the increase in TNF-a were reversed by R7050, indicating that they were mediated by the activation of TNFR1. Preventing peripheral inflammation with an anti-TNF-a antibody prevents TNF-a induction. CONCLUSION Sustained (4 weeks) but not short-term hyperammonemia induces TNF-a in Purkinje neurons in rats. This is mediated by peripheral inflammation. TNF-a is also increased in the Purkinje neurons of patients who die with liver cirrhosis. The results suggest that hyperammonemia induces TNF-a in glial cells and that TNF-a released by glial cells activates TNFR1 in Purkinje neurons, leading to NF-κB nuclear translocation and the induction of TNF-a expression, which may contribute to the neurological alterations observed in hyperammonemia and hepatic encephalopathy.
Collapse
Affiliation(s)
- Tiziano Balzano
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Yaiza M Arenas
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Sherry Dadsetan
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Jerónimo Forteza
- Instituto Valenciano de Patología, Unidad Mixta de Patología Molecular, Centro Investigación Príncipe Felipe/Universidad Católica de Valencia, Valencia, Spain
| | - Sara Gil-Perotin
- Multiple Sclerosis and Neuroimmunology Research Group, Fundación para la Investigación La Fe, Valencia, Spain
| | - Laura Cubas-Nuñez
- Multiple Sclerosis and Neuroimmunology Research Group, Fundación para la Investigación La Fe, Valencia, Spain
| | - Bonaventura Casanova
- Multiple Sclerosis and Neuroimmunology Research Group, Fundación para la Investigación La Fe, Valencia, Spain
| | - Francisco Gracià
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Natalia Varela-Andrés
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Carmina Montoliu
- Instituto de Investigacion Sanitaria INCLIVA, Hospital Clinico de Valencia, Valencia, Spain
| | - Marta Llansola
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain.
| |
Collapse
|
62
|
Poganik JR, Aye Y. Electrophile Signaling and Emerging Immuno- and Neuro-modulatory Electrophilic Pharmaceuticals. Front Aging Neurosci 2020; 12:1. [PMID: 32116644 PMCID: PMC7019031 DOI: 10.3389/fnagi.2020.00001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
With a lipid-rich environment and elevated oxygen consumption, the central nervous system (CNS) is subject to intricate regulation by lipid-derived electrophiles (LDEs). Investigations into oxidative damage and chronic LDE generation in neural disorders have spurred the development of tools that can detect and catalog the gamut of LDE-adducted proteins. Despite these advances, deconstructing the precise consequences of individual protein-specific LDE modifications remained largely impossible until recently. In this perspective, we first overview emerging toolsets that can decode electrophile-signaling events in a protein/context-specific manner, and how the accumulating mechanistic insights brought about by these tools have begun to offer new means to modulate pathways relevant to multiple sclerosis (MS). By surveying the latest data surrounding the blockbuster MS drug dimethyl fumarate that functions through LDE-signaling-like mechanisms, we further provide a vision for how chemical biology tools probing electrophile signaling may be leveraged toward novel interventions in CNS disease.
Collapse
Affiliation(s)
- Jesse R Poganik
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
63
|
Cao Q, Zheng C, Xie Z, Liu L, Zhu J, Jin T. The change of PD1, PDL1 in experimental autoimmune encephalomyelitis treated by 1,25(OH)2D3. J Neuroimmunol 2020; 338:577079. [DOI: 10.1016/j.jneuroim.2019.577079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/28/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022]
|
64
|
Kolahdouzan M, Futhey NC, Kieran NW, Healy LM. Novel Molecular Leads for the Prevention of Damage and the Promotion of Repair in Neuroimmunological Disease. Front Immunol 2019; 10:1657. [PMID: 31379852 PMCID: PMC6658885 DOI: 10.3389/fimmu.2019.01657] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/03/2019] [Indexed: 11/20/2022] Open
Abstract
Neuroinflammation is a prominent pathological feature of all neuroimmunological diseases, including, but not limited to, multiple sclerosis (MS), myasthenia gravis, neuromyelitis optica, and Guillain–Barré syndrome. All currently-approved therapies for the treatment of these diseases focus on controlling or modulating the immune (innate and adaptive) responses to limit demyelination and neuronal damage. The primary purpose of this review is to detail the pre-clinical data and proposed mechanism of action of novel drugs currently in clinical trial, with a focus on novel compounds that promote repair and regeneration in the central nervous system (CNS). As the most recent advances have been made in the field of MS research, this review will focus primarily on this disease and its animal models. However, these compounds are likely to be effective for a range of indications with a neuroinflammatory component. Traditionally, MS was thought to proceed through two distinct phases. The first, predominantly inflammatory stage, is characterized by acute episodes of clinical relapse, followed by periods of partial or total recovery with an apparent absence of overall disease progression. In the vast majority of patients, this relapsing-remitting disease subsequently progresses into a second more chronic, neurodegenerative phase, which is characterized by oligodendrocyte damage and axonal destruction leading to brain atrophy and an accumulation of disability. Recent work has shown that rather than occurring independently, both the inflammatory and degenerative phases may run concurrently. This, combined with evidence that early therapeutic intervention slows accumulation of disability and delays progression, highlights the need for novel therapeutic approaches that promote repair and regeneration early in the disease trajectory. Such compounds may be used as monotherapies or in conjunction with classical anti-inflammatory therapies. This review will highlight novel therapies currently in clinical trial, and likely to appear in clinical practice in the near future, focusing on compounds that target the immune system and/or enhance endogenous repair mechanisms in the CNS.
Collapse
Affiliation(s)
- Mahshad Kolahdouzan
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Naomi C Futhey
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Nicholas W Kieran
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Luke M Healy
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
65
|
El-Deeb OS, Ghanem HB, El-Esawy RO, Sadek MT. The modulatory effects of luteolin on cyclic AMP/Ciliary neurotrophic factor signaling pathway in experimentally induced autoimmune encephalomyelitis. IUBMB Life 2019; 71:1401-1408. [PMID: 31185137 DOI: 10.1002/iub.2099] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 02/03/2023]
Abstract
Multiple sclerosis (MS) is considered to be an autoimmune disorder of the central nervous system (CNS) manifested by chronic inflammation. Although its etiology is not completely understood, inflammation and apoptosis are known to be major players involved in its pathogenesis. Luteolin, the naturally occurring flavonoid, is known by strong antioxidant and anti-inflammatory properties, yet research studies about its therapeutic role in MS are still lacking. The study aimed to provide insight into effects of luteolin in experimental autoimmune encephalomyelitis (EAE) by monitoring inflammatory, apoptotic, and antioxidant biochemical parameters in addition to histological examination findings. The study included 45 adult female Wistar rats allocated to three equal groups: (a) group I: control group, (b) group II: EAE group, EAE was induced by single intradermal injection of 0.2 mL inoculum comprising 20-μg recombinant rat myelin oligodendrocyte glycoprotein (MOG), and (c) group III: luteolin-treated EAE group, luteolin was given in a dose of 10 mg/kg/day, i.p. All groups were subjected to assessment of brain ciliary neurotropic factor (CNTF) mRNA gene expression and measurement of cleaved caspase 3, nuclear factor kappa B (NF-κB), cyclic AMP (cAMP), and macrophage inflammatory protein 1 alpha (MIP-1α) by the ELISA technique, total antioxidant capacity (TAC) level is assessed spectrophotometrically. Compared with the EAE group, luteolin-treated EAE group showed upregulation of CNTF expression and significant increase in cAMP and TAC levels, while it showed significant decrease in cleaved caspase 3, NF-κB, and MIP-1α levels. Based on our data herein, luteolin may provide a promising preclinical therapeutic line in MS being anti-inflammatory, antiapoptotic, and neurotrophic agent. © 2019 IUBMB Life, 71(9):1401-1408, 2019.
Collapse
Affiliation(s)
- Omnia Safwat El-Deeb
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Heba Bassiony Ghanem
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Mona Tayssir Sadek
- Histology and Cell Biology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
66
|
Webb LM, Narvaez Miranda J, Amici SA, Sengupta S, Nagy G, Guerau-de-Arellano M. NF-κB/mTOR/MYC Axis Drives PRMT5 Protein Induction After T Cell Activation via Transcriptional and Non-transcriptional Mechanisms. Front Immunol 2019; 10:524. [PMID: 30941147 PMCID: PMC6433977 DOI: 10.3389/fimmu.2019.00524] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/26/2019] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) mediated by CD4+ T cells and modeled via experimental autoimmune encephalomyelitis (EAE). Inhibition of PRMT5, the major Type II arginine methyltransferase, suppresses pathogenic T cell responses and EAE. PRMT5 is transiently induced in proliferating memory inflammatory Th1 cells and during EAE. However, the mechanisms driving PRMT5 protein induction and repression as T cells expand and return to resting is currently unknown. Here, we used naive mouse and memory mouse and human Th1/Th2 cells as models to identify mechanisms controlling PRMT5 protein expression in initial and recall T cell activation. Initial activation of naive mouse T cells resulted in NF-κB-dependent transient Prmt5 transcription and NF-κB, mTOR and MYC-dependent PRMT5 protein induction. In murine memory Th cells, transcription and miRNA loss supported PRMT5 induction to a lesser extent than in naive T cells. In contrast, NF-κB/MYC/mTOR-dependent non-transcriptional PRMT5 induction played a major role. These results highlight the importance of the NF-κB/mTOR/MYC axis in PRMT5-driven pathogenic T cell expansion and may guide targeted therapeutic strategies for MS.
Collapse
Affiliation(s)
- Lindsay M Webb
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Janiret Narvaez Miranda
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Stephanie A Amici
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Shouvonik Sengupta
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Gregory Nagy
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Mireia Guerau-de-Arellano
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, College of Medicine, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
67
|
Fingolimod Suppresses the Proinflammatory Status of Interferon-γ-Activated Cultured Rat Astrocytes. Mol Neurobiol 2019; 56:5971-5986. [PMID: 30701416 DOI: 10.1007/s12035-019-1481-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022]
Abstract
Astroglia, the primary homeostatic cells of the central nervous system, play an important role in neuroinflammation. They act as facultative immunocompetent antigen-presenting cells (APCs), expressing major histocompatibility complex (MHC) class II antigens upon activation with interferon (IFN)-γ and possibly other proinflammatory cytokines that are upregulated in disease states, including multiple sclerosis (MS). We characterized the anti-inflammatory effects of fingolimod (FTY720), an established drug for MS, and its phosphorylated metabolite (FTY720-P) in IFN-γ-activated cultured rat astrocytes. The expression of MHC class II compartments, β2 adrenergic receptor (ADR-β2), and nuclear factor kappa-light-chain enhancer of activated B cells subunit p65 (NF-κB p65) was quantified in immunofluorescence images acquired by laser scanning confocal microscopy. In addition, MHC class II-enriched endocytotic vesicles were labeled by fluorescent dextran and their mobility analyzed in astrocytes subjected to different treatments. FTY720 and FTY720-P treatment significantly reduced the number of IFN-γ-induced MHC class II compartments and substantially increased ADR-β2 expression, which is otherwise small or absent in astrocytes in MS. These effects could be partially attributed to the observed decrease in NF-κB p65 expression, because the NF-κB signaling cascade is activated in inflammatory processes. We also found attenuated trafficking and secretion from dextran-labeled endo-/lysosomes that may hinder efficient delivery of MHC class II molecules to the plasma membrane. Our data suggest that FTY720 and FTY720-P at submicromolar concentrations mediate anti-inflammatory effects on astrocytes by suppressing their action as APCs, which may further downregulate the inflammatory process in the brain, constituting the therapeutic effect of fingolimod in MS.
Collapse
|
68
|
Kronimus Y, Dodel R, Galuska SP, Neumann S. IgG Fc N-glycosylation: Alterations in neurologic diseases and potential therapeutic target? J Autoimmun 2019; 96:14-23. [DOI: 10.1016/j.jaut.2018.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/09/2018] [Accepted: 10/11/2018] [Indexed: 12/30/2022]
|
69
|
Jafarzadeh A, Nemati M. Therapeutic potentials of ginger for treatment of Multiple sclerosis: A review with emphasis on its immunomodulatory, anti-inflammatory and anti-oxidative properties. J Neuroimmunol 2018; 324:54-75. [PMID: 30243185 DOI: 10.1016/j.jneuroim.2018.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/24/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is characterized by chronic inflammatory response-induced demyelination of the neurons and degeneration of the axons within the central nervous system (CNS). A complex network of immunopathological-, inflammatory- and oxidative parameters involve in the development and advancement of MS. The anti-inflammatory, immunomodulatory and anti-oxidative characteristics of the ginger and several of its components have been indicated in some of experimental and clinical investigations. The possible therapeutic potentials of ginger and its ingredients in the treatment of MS may exert mainly through the regulation of the Th1-, Th2-, Th9-, Th17-, Th22- and Treg cell-related immune responses, down-regulation of the B cell-related immune responses, modulation of the macrophages-related responses, modulation of the production of pro- and anti-inflammatory cytokines, down-regulation of the arachidonic acid-derived mediators, interfering with the toll like receptor-related signaling pathways, suppression of the inflammasomes, down-regulation of the oxidative stress, reduction of the adhesion molecules expression, and down-regulation of the expression of the chemokines and chemokine receptors. This review aimed to provide a comprehensive knowledge regarding the immunomodulatory-, anti-inflammatory and anti-oxidative properties of ginger and its components, and highlight novel insights into the possible therapeutic potentials of this plant for treatment of MS. The review encourages more investigations to consider the therapeutic potentials of ginger and its effective components for managing of MS.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Maryam Nemati
- Department of Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
70
|
Hadjigeorgiou GM, Kountra PM, Koutsis G, Tsimourtou V, Siokas V, Dardioti M, Rikos D, Marogianni C, Aloizou AM, Karadima G, Ralli S, Grigoriadis N, Bogdanos D, Panas M, Dardiotis E. Replication study of GWAS risk loci in Greek multiple sclerosis patients. Neurol Sci 2018; 40:253-260. [PMID: 30361804 DOI: 10.1007/s10072-018-3617-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 10/20/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVES To validate in an ethnically homogeneous Greek multiple sclerosis (MS) cohort, genetic risk factors for the disease, identified through a number of previous multi-ethnic genome-wide association studies (GWAS). METHODS A total of 1228 MS cases and 1014 controls were recruited in the study, from 3 MS centers in Greece. We genotyped 35 susceptibility SNPs that emerged from previous GWAS or meta-analyses of GWAS. Allele and genotype single locus regression analysis, adjusted for gender and site, was performed. Permutation testing was applied to all analyses. RESULTS Six polymorphisms reached statistical significance (permutation p value < 0.05). In particular, rs2760524 of LOC105371664, near RGS1 (permutation p value 0.001), rs3129889 of HLA-DRA, near HLA-DRB1 (permutation p value < 1.00e-04), rs1738074 of TAGAP (permutation p value 0.007), rs703842 of METTL1/CYP27B1 (permutation p value 0.008), rs9596270 of DLEU1 (permutation p value < 1.00e-04), and rs17445836 of LincRNA, near IRF8 (permutation p value 0.001) were identified as susceptibility risk factors in our group. CONCLUSION The current study replicated a number of GWAS susceptibility SNPs, which implies that some similarities between the examined Greek population and the MS genetic architecture of the GWAS populations do exist.
Collapse
Affiliation(s)
| | - Persia-Maria Kountra
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Georgios Koutsis
- Neurogenetics Unit, 1st Department of Neurology, Eginition Hospital, University of Athens, Medical School, Athens, Greece
| | - Vana Tsimourtou
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Maria Dardioti
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Dimitrios Rikos
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Chrysoula Marogianni
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Georgia Karadima
- Neurogenetics Unit, 1st Department of Neurology, Eginition Hospital, University of Athens, Medical School, Athens, Greece
| | - Styliani Ralli
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios Bogdanos
- Cellular Immunotherapy & Molecular Immunodiagnostics, Biomedical Section, Centre for Research and Technology-Hellas (CERTH), Institute for Research and Technology-Thessaly (IRETETH), Larissa, Greece
| | - Marios Panas
- Neurogenetics Unit, 1st Department of Neurology, Eginition Hospital, University of Athens, Medical School, Athens, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| |
Collapse
|
71
|
Torabi S, Tamaddon M, Asadolahi M, Shokri G, Tavakoli R, Tasharrofi N, Rezaei R, Tavakolpour V, Sazegar H, Kouhkan F. miR-455-5p downregulation promotes inflammation pathways in the relapse phase of relapsing-remitting multiple sclerosis disease. Immunogenetics 2018; 71:87-95. [DOI: 10.1007/s00251-018-1087-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/21/2018] [Indexed: 01/01/2023]
|
72
|
Abstract
Hypoxia has been associated with multiple sclerosis (MS) and is an important area of research. Hypoxia can exacerbate inflammation via the prolylhydroxylase pathway. Inflammation can also trigger hypoxia by damaging mitochondria and endothelial cells to impair blood flow regulation. We hypothesize that there is a “hypoxia–inflammation cycle” in MS which plays an important role in MS disease progression. Therapies that break this cycle may be an interesting area of exploration for treatment of MS.
Collapse
Affiliation(s)
- Runze Yang
- Department of Radiology, University of Calgary, Calgary, AB, Canada/Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada/Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff F Dunn
- Department of Radiology, University of Calgary, Calgary, AB, Canada/Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada/Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
73
|
Stein T, Wollschlegel A, Te H, Weiss J, Joshi K, Kinzel B, Billich A, Guntermann C, Lehmann JCU. Interferon regulatory factor 5 and nuclear factor kappa‐B exhibit cooperating but also divergent roles in the regulation of pro‐inflammatory cytokines important for the development of
TH
1 and
TH
17 responses. FEBS J 2018; 285:3097-3113. [DOI: 10.1111/febs.14600] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 05/16/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Thomas Stein
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Audrey Wollschlegel
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Helene Te
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Jessica Weiss
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Kushal Joshi
- Department of Chemical Biology & Therapeutics Novartis Institutes for Biomedical Research Novartis Pharma AG Cambridge MA USA
| | - Bernd Kinzel
- Department of Chemical Biology & Therapeutics Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Andreas Billich
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Christine Guntermann
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Joachim C. U. Lehmann
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| |
Collapse
|
74
|
Wang J, Wang X, Chen X, Lu S, Kuang Y, Fei J, Wang Z. Gpr97/Adgrg3 ameliorates experimental autoimmune encephalomyelitis by regulating cytokine expression. Acta Biochim Biophys Sin (Shanghai) 2018; 50:666-675. [PMID: 29860267 DOI: 10.1093/abbs/gmy060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis and its primary animal model, experimental autoimmune encephalomyelitis (EAE), are inflammatory diseases of the central nervous system (CNS) characterized by immune-mediated demyelination and neurodegeneration that may be mediated by inhibition of the nuclear factor-κB (NF-κB) signaling pathway. Gpr97, encoded by Adgrg3, has been reported to regulate the activity of NF-κB. In this study, using a previously established Adgrg3-knockout mouse model, we investigated the roles of Gpr97 in the development of autoimmune CNS disease in mice. We found a marked increase in the expression of Adgrg3 in spinal cords of mice with EAE. Adgrg3-deficient (Adgrg3-/-) mice with EAE exhibited increases in peak severity and the cumulative disease score compared with littermate controls, followed by a notable increase of leukocyte infiltration and more extensive demyelination. The percentages of Th1/Th17 cells in the CNS were significantly increased in Adgrg3-/- mice and accompanied by high levels of interleukin (IL)-6, interferon-γ, tumor necrosis factor-α, and IL-17. An in vitro culture assay verified that Gpr97 regulated proinflammatory cytokine production. Taken together, our results show that Gpr97 plays an important role in the development of EAE and may have a therapeutic potential for the treatment of CNS autoimmunity.
Collapse
Affiliation(s)
- Jinjin Wang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Xiyi Wang
- Department of Medical Genetics, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Xuejiao Chen
- Department of Medical Genetics, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Shunyuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai, China
| | - Ying Kuang
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Jian Fei
- Shanghai Research Center for Model Organisms, Shanghai, China
| | - Zhugang Wang
- Shanghai Research Center for Model Organisms, Shanghai, China
- Department of Medical Genetics, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine of Rui-Jin Hospital, Shanghai, China
| |
Collapse
|
75
|
Yan J, Winterford CM, Catts VS, Pat BK, Pender MP, McCombe PA, Greer JM. Increased constitutive activation of NF-κB p65 (RelA) in peripheral blood cells of patients with progressive multiple sclerosis. J Neuroimmunol 2018; 320:111-116. [DOI: 10.1016/j.jneuroim.2018.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/06/2018] [Accepted: 04/02/2018] [Indexed: 12/17/2022]
|
76
|
Zarbato GF, de Souza Goldim MP, Giustina AD, Danielski LG, Mathias K, Florentino D, de Oliveira Junior AN, da Rosa N, Laurentino AO, Trombetta T, Gomes ML, Steckert AV, Moreira AP, Schuck PF, Fortunato JJ, Barichello T, Petronilho F. Dimethyl Fumarate Limits Neuroinflammation and Oxidative Stress and Improves Cognitive Impairment After Polymicrobial Sepsis. Neurotox Res 2018; 34:418-430. [PMID: 29713994 DOI: 10.1007/s12640-018-9900-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/02/2018] [Accepted: 04/06/2018] [Indexed: 12/18/2022]
Abstract
Sepsis is caused by a dysregulated host response to infection, often associated with acute central nervous system (CNS) dysfunction, which results in long-term cognitive impairment. Dimethyl fumarate (DMF) is an important agent against inflammatory response and reactive species in CNS disorders. Evaluate the effect of DMF on acute and long-term brain dysfunction after experimental sepsis in rats. Male Wistar rats were submitted to the cecal ligation and puncture (CLP) model. The groups were divided into sham (control) + vehicle, sham + NAC, sham + DMF, CLP + vehicle, CLP + NAC, and CLP + DMF. The animals were treated with DMF (15 mg/kg at 0 and 12 h after CLP, per gavage) and the administration of n-acetylcysteine (NAC) (20 mg/kg; 3, 6, and 12 h after CLP, subcutaneously) was used as positive control. Twenty-four hours after CLP, cytokines, myeloperoxidase (MPO), nitrite/nitrate (N/N), oxidative damage to lipids and proteins, and antioxidant enzymes were evaluated in the hippocampus, total cortex, and prefrontal cortex. At 10 days after sepsis induction, behavioral tests were performed to assess cognitive damage. We observed an increase in cytokine levels, MPO activity, N/N concentration, and oxidative damage, a reduction in SOD and GPx activity in the brain structures, and cognitive damage in CLP rats. DMF treatment was effective in reversing these parameters. DMF reduces sepsis-induced neuroinflammation, oxidative stress, and cognitive impairment in rats subjected to the CLP model.
Collapse
Affiliation(s)
- Graciela Freitas Zarbato
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Mariana Pereira de Souza Goldim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Amanda Della Giustina
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Lucinéia Gainski Danielski
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Khiany Mathias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Drielly Florentino
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Aloir Neri de Oliveira Junior
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Naiana da Rosa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Ana Olivia Laurentino
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Taina Trombetta
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Maria Luiza Gomes
- Laboratory Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Amanda Valnier Steckert
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Ana Paula Moreira
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Patricia Fernanda Schuck
- Laboratory Inborn Errors of Metabolism, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Jucelia Jeremias Fortunato
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil.,Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabricia Petronilho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil.
| |
Collapse
|
77
|
Enderami A, Fouladi R, Hosseini SH. First-episode psychosis as the initial presentation of multiple sclerosis: a case report. Int Med Case Rep J 2018; 11:73-76. [PMID: 29670407 PMCID: PMC5896665 DOI: 10.2147/imcrj.s157287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Multiple sclerosis (MS) is an inflammatory disease that affects the central nervous system (CNS). MS with episode of psychosis is a rare entity, and to the best of our knowledge, no case has been reported from Iran till date. Case presentation We report a case of MS with first-episode psychosis in a 27-year-old single man with no history of psychiatric disorder or drug abuse. The patient developed neurological symptoms after 3 months and was finally diagnosed as a case of MS. His symptoms started with behavioral dysfunctions and progressively resulted in depression. Subsequently, treatment was performed with citalopram 20 mg daily, risperidone 2 mg three times a day, and biperiden 2 mg three times a day; however, no improvements in the symptoms were observed. T2-weighted magnetic resonance imaging has demonstrated periventricular and white matter multiple sclerotic plugs with lesions. Eventually, MS was diagnosed after the appearance of paresthesia, upper and lower limb muscle weakness, ataxia, and urinary incontinency as typical signs. Then, the medications were changed to methylprednisolone and interferon therapy, which resulted in improvements in the clinical conditions of the patient. Conclusion Based on the fact that organic disorders such as MS may sometimes appear with initial pure psychiatric symptoms without any neurological signs and symptoms, examinations for symptoms linked to CNS dysfunction, cognitive changes, atypical symptoms, detailed neurological examination, and limited response to conventional antipsychotic drugs are highly recommended to be carried out for patients with first-episode psychosis and even in the followup period.
Collapse
Affiliation(s)
- Athena Enderami
- Department of Psychiatry, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Sari, Iran
| | - Rose Fouladi
- Sport Injuries and Corrective Exercise, University of Mazandaran, Mazandaran, Babolsar, Iran
| | - Seyed Hamzeh Hosseini
- Psychiatry and Behavioral Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
78
|
Zhang M, Jin F. 1α,25-Dihydroxyvitamin D3 Ameliorates Seawater Aspiration-Induced Lung Injury By Inhibiting The Translocation Of NF-κB and RhoA. Inflammation 2018; 40:832-839. [PMID: 28161733 PMCID: PMC5429345 DOI: 10.1007/s10753-017-0527-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our previous study have reported that 1α,25-Dihydroxyvitamin D3 (calcitriol) suppresses seawater aspiration-induced ALI in vitro and in vivo. We also have confirmed that treatment with calcitriol ameliorates seawater aspiration-induced inflammation and pulmonary edema via the inhibition of NF-κB and RhoA/Rho kinase pathway activation. In our further work, we investigated the effect of calcitriol on nuclear translocation of NF-κB and membrane translocation of RhoA in vitro. A549 cells and rat pulmonary microvascular endothelial cells (RPMVECs) were cultured with calcitriol or not for 48 h and then stimulated with 25% seawater for 40 min. After these treatments, cells were collected and performed with immunofluorescent staining to observe the translocation of NF-κB and RhoA and the cytoskeleton remodeling. In vitro, seawater stimulation activates nuclear translocation of NF-κB and membrane translocation of RhoA in A549 cells. In addition, seawater administration also induced cytoskeleton remodeling in A549 cells and RPMVECs. However, pretreatment with calcitriol significantly inhibited the activation of NF-κB and RhoA/Rho kinase pathways, as demonstrated by the reduced nuclear translocation of NF-κB and membrane translocation of RhoA in A549 cells. Meanwhile, treatment of calcitriol also regulated the cytoskeleton remodeling in both A549 cells and RPMVECs. These results demonstrated that treatment with calcitriol ameliorates seawater aspiration-induced ALI via inhibition of nuclear translocation of NF-κB and membrane translocation of RhoA and protection of alveolar epithelial and pulmonary microvascular endothelial barrier.
Collapse
Affiliation(s)
- Minlong Zhang
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Faguang Jin
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China.
| |
Collapse
|
79
|
Neuroprotective effect of S-allyl cysteine on an experimental model of multiple sclerosis: Antioxidant effects. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
80
|
Yazdanpanahi N, Etemadifar M, Kardi MT, Shams E, Shahbazi S. Investigation of ERG Gene Expression in Iranian Patients with Multiple Sclerosis. Immunol Invest 2018; 47:351-359. [DOI: 10.1080/08820139.2018.1433203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Nasrin Yazdanpanahi
- Department of Genetics, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Masoud Etemadifar
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Elahe Shams
- Young Researchers and Elite Club, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Shirin Shahbazi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
81
|
Perga S, Martire S, Montarolo F, Giordani I, Spadaro M, Bono G, Corvisieri S, Messuti I, Panzica G, Orlandi F, Bertolotto A. The Footprints of Poly-Autoimmunity: Evidence for Common Biological Factors Involved in Multiple Sclerosis and Hashimoto's Thyroiditis. Front Immunol 2018. [PMID: 29527211 PMCID: PMC5829620 DOI: 10.3389/fimmu.2018.00311] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Autoimmune diseases are a diverse group of chronic disorders and affect a multitude of organs and systems. However, the existence of common pathophysiological mechanisms is hypothesized and reports of shared risk are emerging as well. In this regard, patients with multiple sclerosis (MS) have been shown to have an increased susceptibility to develop chronic autoimmune thyroid diseases, in particular Hashimoto's thyroiditis (HT), suggesting an autoimmune predisposition. However, studies comparing such different pathologies of autoimmune origin are still missing till date. In the present study, we sought to investigate mechanisms which may lead to the frequent coexistence of MS and HT by analyzing several factors related to the pathogenesis of MS and HT in patients affected by one or both diseases, as well as in healthy donors. In particular, we analyzed peripheral blood mononuclear cell gene-expression levels of common candidate genes such as TNFAIP3, NR4A family, BACH2, FOXP3, and PDCD5, in addition to the regulatory T cell (Treg) percentage and the 25-hydroxy vitamin D serum levels. Our findings support the plausibility of the existence of common deregulated mechanisms shared by MS and HT, such as BACH2/PDCD5-FOXP3 pathways and Tregs. Although the biological implications of these data need to be further investigated, we have highlighted the relevance of studies comparing different autoimmune pathologies for the understanding of the core concepts of autoimmunity.
Collapse
Affiliation(s)
- Simona Perga
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy.,Regional Reference Centre for Multiple Sclerosis (CReSM), University Hospital S. Luigi Gonzaga, Orbassano, Turin, Italy.,Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Serena Martire
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy.,Regional Reference Centre for Multiple Sclerosis (CReSM), University Hospital S. Luigi Gonzaga, Orbassano, Turin, Italy
| | - Francesca Montarolo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy.,Regional Reference Centre for Multiple Sclerosis (CReSM), University Hospital S. Luigi Gonzaga, Orbassano, Turin, Italy.,Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Ilaria Giordani
- SCDU Endocrinology and Metabolism, Humanitas Gradenigo Hospital, Department of Oncology, University of Turin, Turin, Italy
| | - Michela Spadaro
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy.,Regional Reference Centre for Multiple Sclerosis (CReSM), University Hospital S. Luigi Gonzaga, Orbassano, Turin, Italy
| | - Gabriele Bono
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy.,Regional Reference Centre for Multiple Sclerosis (CReSM), University Hospital S. Luigi Gonzaga, Orbassano, Turin, Italy
| | - Stefania Corvisieri
- SCDU Endocrinology and Metabolism, Humanitas Gradenigo Hospital, Department of Oncology, University of Turin, Turin, Italy
| | - Ilaria Messuti
- SCDU Endocrinology and Metabolism, Humanitas Gradenigo Hospital, Department of Oncology, University of Turin, Turin, Italy
| | - Giancarlo Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy.,Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Fabio Orlandi
- SCDU Endocrinology and Metabolism, Humanitas Gradenigo Hospital, Department of Oncology, University of Turin, Turin, Italy
| | - Antonio Bertolotto
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Turin, Italy.,Regional Reference Centre for Multiple Sclerosis (CReSM), University Hospital S. Luigi Gonzaga, Orbassano, Turin, Italy
| |
Collapse
|
82
|
Jafarzadeh A, Azizi SV, Arabi Z, Ahangar-Parvin R, Mohammadi-Kordkhayli M, Larussa T, Khatami F, Nemati M. Vitamin D down-regulates the expression of some Th17 cell-related cytokines, key inflammatory chemokines, and chemokine receptors in experimental autoimmune encephalomyelitis. Nutr Neurosci 2018; 22:725-737. [DOI: 10.1080/1028415x.2018.1436237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Abdollah Jafarzadeh
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Sayyed Vahab Azizi
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Arabi
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Rayhaneh Ahangar-Parvin
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Tiziana Larussa
- Department of Health Science, University of Catanzaro ‘Magna Graecia’, Catanzaro, Italy
| | - Fariba Khatami
- Department of Pathology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Maryam Nemati
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
- Department of Laboratory Sciences, Para-Medicine School, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
83
|
Gharagozloo M, Gris KV, Mahvelati T, Amrani A, Lukens JR, Gris D. NLR-Dependent Regulation of Inflammation in Multiple Sclerosis. Front Immunol 2018; 8:2012. [PMID: 29403486 PMCID: PMC5778124 DOI: 10.3389/fimmu.2017.02012] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/28/2017] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) associated with inappropriate activation of lymphocytes, hyperinflammatory responses, demyelination, and neuronal damage. In the past decade, a number of biological immunomodulators have been developed that suppress the peripheral immune responses and slow down the progression of the disease. However, once the inflammation of the CNS has commenced, it can cause serious permanent neuronal damage. Therefore, there is a need for developing novel therapeutic approaches that control and regulate inflammatory responses within the CNS. Nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) are intracellular regulators of inflammation expressed by many cell types within the CNS. They redirect multiple signaling pathways initiated by pathogens and molecules released by injured tissues. NLR family members include positive regulators of inflammation, such as NLRP3 and NLRC4 and anti-inflammatory NLRs, such as NLRX1 and NLRP12. They exert immunomodulatory effect at the level of peripheral immune responses, including antigen recognition and lymphocyte activation and differentiation. Also, NLRs regulate tissue inflammatory responses. Understanding the molecular mechanisms that are placed at the crossroad of innate and adaptive immune responses, such as NLR-dependent pathways, could lead to the discovery of new therapeutic targets. In this review, we provide a summary of the role of NLRs in the pathogenesis of MS. We also summarize how anti-inflammatory NLRs regulate the immune response within the CNS. Finally, we speculate the therapeutic potential of targeting NLRs in MS.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Program of Immunology, Faculty of Medicine and Health Sciences, Department of Pediatrics, CR-CHUS, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Katsiaryna V. Gris
- Program of Immunology, Faculty of Medicine and Health Sciences, Department of Pediatrics, CR-CHUS, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Tara Mahvelati
- Program of Immunology, Faculty of Medicine and Health Sciences, Department of Pediatrics, CR-CHUS, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Abdelaziz Amrani
- Program of Immunology, Faculty of Medicine and Health Sciences, Department of Pediatrics, CR-CHUS, University of Sherbrooke, Sherbrooke, QC, Canada
| | - John R. Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Denis Gris
- Program of Immunology, Faculty of Medicine and Health Sciences, Department of Pediatrics, CR-CHUS, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
84
|
Yue Y, Stone S, Lin W. Role of nuclear factor κB in multiple sclerosis and experimental autoimmune encephalomyelitis. Neural Regen Res 2018; 13:1507-1515. [PMID: 30127103 PMCID: PMC6126134 DOI: 10.4103/1673-5374.237109] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The transcription factor nuclear factor κB (NF-κB) plays major roles in inflammatory diseases through regulation of inflammation and cell viability. Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS). It has been shown that NF-κB is activated in multiple cell types in the CNS of MS patients, including T cells, microglia/macrophages, astrocytes, oligodendrocytes, and neurons. Interestingly, data from animal model studies, particularly studies of experimental autoimmune encephalomyelitis, have suggested that NF-κB activation in these individual cell types has distinct effects on the development of MS. In this review, we will cover the current literature on NF-κB and the evidence for its role in the development of MS and its animal model experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Yuan Yue
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Sarrabeth Stone
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Wensheng Lin
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
85
|
Li D, Tomljenovic L, Li Y, Shaw CA. RETRACTED: Subcutaneous injections of aluminum at vaccine adjuvant levels activate innate immune genes in mouse brain that are homologous with biomarkers of autism. J Inorg Biochem 2017; 177:39-54. [PMID: 28923356 DOI: 10.1016/j.jinorgbio.2017.08.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Dan Li
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lucija Tomljenovic
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yongling Li
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher A Shaw
- Dept. of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Program in Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
86
|
Shao L, Yu S, Ji W, Li H, Gao Y. The Contribution of Necroptosis in Neurodegenerative Diseases. Neurochem Res 2017; 42:2117-2126. [PMID: 28382594 DOI: 10.1007/s11064-017-2249-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 03/06/2017] [Accepted: 03/25/2017] [Indexed: 12/29/2022]
Abstract
Over the past decades, cell apoptosis has been significantly reputed as an accidental, redundant and alternative manner of cell demise which partakes in homeostasis in the development of extensive diseases. Nevertheless, necroptosis, another novel manner of cell death through a caspase-independent way, especially in neurodegenerative diseases remains ambiguous. The cognition of this form of cell demise is helpful to understand other forms of morphological resemblance of necrosis. Additionally, the concrete signal mechanism in the regulation of necroptosis is beneficial to the diagnosis and treatment of neurodegenerative diseases. Recent studies have demonstrated that necroptotic inhibitor, 24(S)-Hydroxycholesterol and partial specific histone deacetylase inhibitors could alleviate pathogenetic conditions of neurodegenerative diseases via necroptosis pathway. In this review, we summarize recent researches about mechanisms and modulation of necroptotic signaling pathways and probe into the role of programmed necroptotic cell demise in neurodegenerative diseases such as Parkinson's disease, Multiple sclerosis, Amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Lifei Shao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.,Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Shuping Yu
- Department of Blood Transfusion, The Forth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu, China.,Center of Laboratory Medicine, Affiliate Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei Ji
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.,Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Haizhen Li
- Medical College, Nantong University, Nantong, 226001, Jiangsu, China.,Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Yilu Gao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|