1
|
Carver JJ, Lau KM, Puckett AE, Didonna A. Autoimmune demyelination alters hypothalamic transcriptome and endocrine function. J Neuroinflammation 2024; 21:12. [PMID: 38178091 PMCID: PMC10768476 DOI: 10.1186/s12974-023-03006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024] Open
Abstract
The hypothalamus is a brain structure that is deputed to maintain organism homeostasis by regulating autonomic function and hormonal production as part of the neuroendocrine system. Dysfunction in hypothalamic activity results in behavioral alterations, depression, metabolic syndromes, fatigue, and infertility. Remarkably, many of these symptoms are associated with multiple sclerosis (MS), a chronic autoimmune disorder of the central nervous system (CNS) characterized by focal demyelination, immune cell infiltration into the brain parenchyma, and neurodegeneration. Furthermore, altered hormonal levels have been documented in MS patients, suggesting the putative involvement of hypothalamic deficits in MS clinical manifestations. Yet, a systematic analysis of hypothalamic function in response to neuroinflammatory stress is still lacking. To fill this gap, here we performed a longitudinal profiling of the hypothalamic transcriptome upon experimental autoimmune encephalomyelitis (EAE)-a murine disease model recapitulating key MS phenotypes at both histopathological and molecular levels. We show that changes in gene expression connected with an anti-inflammatory response start already at pre-onset and persist along EAE progression. Altered levels of hypothalamic neuropeptides were also detected, which possibly underlie homeostatic responses to stress and aberrant feeding behaviors. Last, a thorough investigation of the principal endocrine glands highlighted defects in the main steroidogenic pathways upon disease. Collectively, our findings corroborate the central role of hypothalamic dysfunction in CNS autoimmunity.
Collapse
Affiliation(s)
- Jonathan J Carver
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA
| | - Kristy M Lau
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA
| | - Alexandra E Puckett
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA
| | - Alessandro Didonna
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, 600 Moye Blvd., Greenville, NC, USA.
| |
Collapse
|
2
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2023:1-23. [PMID: 38100543 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
3
|
Wang H, Mou H, Xu X, Liu C, Zhou G, Gao B. LncRNA KCNQ1OT1 (potassium voltage-gated channel subfamily Q member 1 opposite strand/antisense transcript 1) aggravates acute kidney injury by activating p38/NF-κB pathway via miR-212-3p/MAPK1 (mitogen-activated protein kinase 1) axis in sepsis. Bioengineered 2021; 12:11353-11368. [PMID: 34783627 PMCID: PMC8810185 DOI: 10.1080/21655979.2021.2005987] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/24/2022] Open
Abstract
Acute kidney injury (AKI), a common complication of sepsis, is characterized by a rapid loss of renal excretory function. A variety of etiologies and pathophysiological processes may contribute to AKI. Previously, mitogen-activated protein kinase 1 (MAPK1) was reported to regulate cellular processes in various sepsis-associated diseases. The current study aimed to further explore the biological function and regulatory mechanism of MAPK1 in sepsis-induced AKI. In our study, MAPK1 exhibited high expression in the serum of AKI patients. Functionally, knockdown of MAPK1 suppressed inflammatory response, cell apoptosis in response of lipopolysaccharide (LPS) induction in HK-2 cells. Moreover, MAPK1 deficiency alleviated renal inflammation, renal dysfunction, and renal injury in vivo. Mechanistically, MAPK1 could activate the downstream p38/NF-κB pathway. Moreover, long noncoding RNA potassium voltage-gated channel subfamily Q member 1 opposite strand/antisense transcript 1 (KCNQ1OT1) was identified to serve as a competing endogenous RNA for miR-212-3p to regulate MAPK1. Finally, rescue assays indicated that the inhibitory effect of KCNQ1OT1 knockdown on inflammatory response, cell apoptosis, and p38/NF-κB pathway was reversed by MAPK1 overexpression in HK-2 cells. In conclusion, KCNQ1OT1 aggravates acute kidney injury by activating p38/NF-κB pathway via miR-212-3p/MAPK1 axis in sepsis. Therefore, KCNQ1OT may serve as a potential biomarker for the prognosis and diagnosis of AKI patients.
Collapse
Affiliation(s)
- Haixia Wang
- Department of Critical Care Medicine, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Hongbin Mou
- Department of Nephrology, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Xiaolan Xu
- Department of Critical Care Medicine, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Changhua Liu
- Department of Nephrology, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Gang Zhou
- Department of Nephrology, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| | - Bo Gao
- Department of Nephrology, Subei People’s Hospital of Jiangsu Province, Yangzhou, China
| |
Collapse
|
4
|
Devi-Marulkar P, Moraes-Cabe C, Campagne P, Corre B, Meghraoui-Kheddar A, Bondet V, Llibre A, Duffy D, Maillart E, Papeix C, Pellegrini S, Michel F. Altered Immune Phenotypes and HLA-DQB1 Gene Variation in Multiple Sclerosis Patients Failing Interferon β Treatment. Front Immunol 2021; 12:628375. [PMID: 34113337 PMCID: PMC8185344 DOI: 10.3389/fimmu.2021.628375] [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: 11/11/2020] [Accepted: 04/23/2021] [Indexed: 11/25/2022] Open
Abstract
Background Interferon beta (IFNβ) has been prescribed as a first-line disease-modifying therapy for relapsing-remitting multiple sclerosis (RRMS) for nearly three decades. However, there is still a lack of treatment response markers that correlate with the clinical outcome of patients. Aim To determine a combination of cellular and molecular blood signatures associated with the efficacy of IFNβ treatment using an integrated approach. Methods The immune status of 40 RRMS patients, 15 of whom were untreated and 25 that received IFNβ1a treatment (15 responders, 10 non-responders), was investigated by phenotyping regulatory CD4+ T cells and naïve/memory T cell subsets, by measurement of circulating IFNα/β proteins with digital ELISA (Simoa) and analysis of ~600 immune related genes including 159 interferon-stimulated genes (ISGs) with the Nanostring technology. The potential impact of HLA class II gene variation in treatment responsiveness was investigated by genotyping HLA-DRB1, -DRB3,4,5, -DQA1, and -DQB1, using as a control population the Milieu Interieur cohort of 1,000 French healthy donors. Results Clinical responders and non-responders displayed similar plasma levels of IFNβ and similar ISG profiles. However, non-responders mainly differed from other subject groups with reduced circulating naïve regulatory T cells, enhanced terminally differentiated effector memory CD4+ TEMRA cells, and altered expression of at least six genes with immunoregulatory function. Moreover, non-responders were enriched for HLA-DQB1 genotypes encoding DQ8 and DQ2 serotypes. Interestingly, these two serotypes are associated with type 1 diabetes and celiac disease. Overall, the immune signatures of non-responders suggest an active disease that is resistant to therapeutic IFNβ, and in which CD4+ T cells, likely restricted by DQ8 and/or DQ2, exert enhanced autoreactive and bystander inflammatory activities.
Collapse
Affiliation(s)
- Priyanka Devi-Marulkar
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Carolina Moraes-Cabe
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Pascal Campagne
- Center of Bioinformatics, Biostatistics and Integrative Biology, Institut Pasteur, Paris, France
| | - Béatrice Corre
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Aida Meghraoui-Kheddar
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincent Bondet
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | - Alba Llibre
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | - Darragh Duffy
- Translational Immunology Laboratory, Department of Immunology, Institut Pasteur, Paris, France
| | | | - Caroline Papeix
- Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France
| | - Sandra Pellegrini
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Frédérique Michel
- Cytokine Signaling Unit, Department of Immunology, Institut Pasteur, Paris, France.,INSERM U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
5
|
Mazewski C, Perez RE, Fish EN, Platanias LC. Type I Interferon (IFN)-Regulated Activation of Canonical and Non-Canonical Signaling Pathways. Front Immunol 2020; 11:606456. [PMID: 33329603 PMCID: PMC7719805 DOI: 10.3389/fimmu.2020.606456] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
For several decades there has been accumulating evidence implicating type I interferons (IFNs) as key elements of the immune response. Therapeutic approaches incorporating different recombinant type I IFN proteins have been successfully employed to treat a diverse group of diseases with significant and positive outcomes. The biological activities of type I IFNs are consequences of signaling events occurring in the cytoplasm and nucleus of cells. Biochemical events involving JAK/STAT proteins that control transcriptional activation of IFN-stimulated genes (ISGs) were the first to be identified and are referred to as "canonical" signaling. Subsequent identification of JAK/STAT-independent signaling pathways, critical for ISG transcription and/or mRNA translation, are denoted as "non-canonical" or "non-classical" pathways. In this review, we summarize these signaling cascades and discuss recent developments in the field, specifically as they relate to the biological and clinical implications of engagement of both canonical and non-canonical pathways.
Collapse
Affiliation(s)
- Candice Mazewski
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
- Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ricardo E. Perez
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
- Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Eleanor N. Fish
- Toronto General Hospital Research Institute, University Health Network and Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
- Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| |
Collapse
|
6
|
Torre-Fuentes L, Matías-Guiu JA, Pytel V, Montero-Escribano P, Maietta P, Álvarez S, Gómez-Pinedo U, Matías-Guiu J. Variants of genes encoding TNF receptors and ligands and proteins regulating TNF activation in familial multiple sclerosis. CNS Neurosci Ther 2020; 26:1178-1184. [PMID: 32951330 PMCID: PMC7564193 DOI: 10.1111/cns.13456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Numerous genetic variants have been associated with susceptibility to multiple sclerosis (MS). Variants located in genes involved in specific pathways, such as those affecting TNF-α, can contribute to the risk of MS. The purpose of this study was to determine whether variants of these genes are associated with greater risk of MS. METHODS We used whole-exome sequencing to study genes coding for TNF-α receptors and ligands, and proteins promoting TNF-α expression in 116 individuals from 19 families including at least two MS patients. We compared patients with MS, patients with other autoimmune diseases, and healthy individuals. RESULTS Greater polymorphism was observed in several genes in families with familial MS compared to the general population; this may reflect greater susceptibility to autoimmune diseases. Pedigree analysis also revealed that LT-α variants rs1041981 and rs2229094 and LT-β variant rs4647197 were associated with MS and that LT-β variant rs4647183 was associated with other autoimmune diseases. The association between autoimmune disease and TNFAIP2 variant rs1132339 is particularly noteworthy, as is the fact that TNFAIP6 variant rs1046668 appears to follow a recessive inheritance pattern. CONCLUSIONS Our findings support the idea that the risk of familial MS is associated with variants of signaling pathways, including those involving TNF-α.
Collapse
Affiliation(s)
- Laura Torre-Fuentes
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Jordi A Matías-Guiu
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Vanesa Pytel
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain.,Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Paloma Montero-Escribano
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | - Ulises Gómez-Pinedo
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Jorge Matías-Guiu
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain.,Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
7
|
Molecular Effects of FDA-Approved Multiple Sclerosis Drugs on Glial Cells and Neurons of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21124229. [PMID: 32545828 PMCID: PMC7352301 DOI: 10.3390/ijms21124229] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is characterized by peripheral and central inflammatory features, as well as demyelination and neurodegeneration. The available Food and Drug Administration (FDA)-approved drugs for MS have been designed to suppress the peripheral immune system. In addition, however, the effects of these drugs may be partially attributed to their influence on glial cells and neurons of the central nervous system (CNS). We here describe the molecular effects of the traditional and more recent FDA-approved MS drugs Fingolimod, Dimethyl Fumarate, Glatiramer Acetate, Interferon-β, Teriflunomide, Laquinimod, Natalizumab, Alemtuzumab and Ocrelizumab on microglia, astrocytes, neurons and oligodendrocytes. Furthermore, we point to a possible common molecular effect of these drugs, namely a key role for NFκB signaling, causing a switch from pro-inflammatory microglia and astrocytes to anti-inflammatory phenotypes of these CNS cell types that recently emerged as central players in MS pathogenesis. This notion argues for the need to further explore the molecular mechanisms underlying MS drug action.
Collapse
|
8
|
Hurtado-Guerrero I, Hernáez B, Pinto-Medel MJ, Calonge E, Rodriguez-Bada JL, Urbaneja P, Alonso A, Mena-Vázquez N, Aliaga P, Issazadeh-Navikas S, Pavia J, Leyva L, Alcamí J, Alcamí A, Fernández Ó, Oliver-Martos B. Antiviral, Immunomodulatory and Antiproliferative Activities of Recombinant Soluble IFNAR2 without IFN-ß Mediation. J Clin Med 2020; 9:jcm9040959. [PMID: 32244308 PMCID: PMC7230527 DOI: 10.3390/jcm9040959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 12/21/2022] Open
Abstract
Soluble receptors of cytokines are able to modify cytokine activities and therefore the immune system, and some have intrinsic biological activities without mediation from their cytokines. The soluble interferon beta (IFN-ß) receptor is generated through alternative splicing of IFNAR2 and has both agonist and antagonist properties for IFN-ß, but its role is unknown. We previously demonstrated that a recombinant human soluble IFN-ß receptor showed intrinsic therapeutic efficacy in a mouse model of multiple sclerosis. Here we evaluate the potential biological activities of recombinant sIFNAR2 without the mediation of IFN-ß in human cells. Recombinant sIFNAR2 down-regulated the production of IL-17 and IFN-ɣ and reduced the cell proliferation rate. Moreover, it showed a strong antiviral activity, fully protecting the cell monolayer after being infected by the virus. Specific inhibitors completely abrogated the antiviral activity of IFN-ß, but not that of the recombinant sIFNAR2, and there was no activation of the JAK-STAT signaling pathway. Consequently, r-sIFNAR2 exerts immunomodulatory, antiproliferative and antiviral activities without IFN-ß mediation, and could be a promising treatment against viral infections and immune-mediated diseases.
Collapse
Affiliation(s)
- Isaac Hurtado-Guerrero
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Bruno Hernáez
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (B.H.); (A.A.)
| | - María J. Pinto-Medel
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Esther Calonge
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda 28220 Madrid, Spain; (E.C.); (J.A.)
| | - José L. Rodriguez-Bada
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Patricia Urbaneja
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Ana Alonso
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - Natalia Mena-Vázquez
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC de Reumatología, Hospital Regional Universitario de Málaga, 29009 Málaga, Spain
| | - Pablo Aliaga
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, Copenhagen Biocentre, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - José Pavia
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- Departamento de Farmacología y Pediatría, Facultad de Medicina. Universidad de Málaga, 29010 Málaga, Spain
| | - Laura Leyva
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
| | - José Alcamí
- AIDS Immunopathology Unit, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda 28220 Madrid, Spain; (E.C.); (J.A.)
- HIV Unit, Infectious Disease Service, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (B.H.); (A.A.)
| | - Óscar Fernández
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- Departamento de Farmacología y Pediatría, Facultad de Medicina. Universidad de Málaga, 29010 Málaga, Spain
| | - Begoña Oliver-Martos
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29009 Málaga, Spain; (I.H.-G.); (M.J.P.-M.); (J.L.R.-B.); (P.U.); (A.A.); (N.M.-V.); (P.A.); (J.P.); (L.L.); (Ó.F.)
- UGC Neurociencias. Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Red Temática de Investigación Cooperativa: Red Española de Esclerosis Múltiple REEM (RD16/0015/0010), 28049 Madrid, Spain
- Correspondence: ; Tel.: +34-951-290-223
| |
Collapse
|
9
|
Tsai MH, Pai LM, Lee CK. Fine-Tuning of Type I Interferon Response by STAT3. Front Immunol 2019; 10:1448. [PMID: 31293595 PMCID: PMC6606715 DOI: 10.3389/fimmu.2019.01448] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/10/2019] [Indexed: 12/20/2022] Open
Abstract
Type I interferon (IFN-I) is induced during innate immune response and is required for initiating antiviral activity, growth inhibition, and immunomodulation. STAT1, STAT2, and STAT3 are activated in response to IFN-I stimulation. STAT1, STAT2, and IRF9 form ISGF3 complex which transactivates downstream IFN-stimulated genes and mediates antiviral response. However, the role of STAT3 remains to be characterized. Here, we review the multiple actions of STAT3 on suppressing IFN-I responses, including blocking IFN-I signaling, downregulating the expression of ISGF3 components, and antagonizing the transcriptional activity of ISGF3. Finally, we discuss the evolution of the suppressive activity of STAT3 and the therapeutic potential of STAT3 inhibitors in host defense against viral infections and IFN-I-associated diseases.
Collapse
Affiliation(s)
- Ming-Hsun Tsai
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Li-Mei Pai
- Department of Biochemistry and Molecular Biology, Chang Gung University, Taoyuan, Taiwan.,Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.,Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chien-Kuo Lee
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
10
|
Mufarrege EF, Haile LA, Etcheverrigaray M, Verthelyi DI. Multiplexed Gene Expression as a Characterization of Bioactivity for Interferon Beta (IFN-β) Biosimilar Candidates: Impact of Innate Immune Response Modulating Impurities (IIRMIs). AAPS JOURNAL 2019; 21:26. [DOI: 10.1208/s12248-019-0300-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/12/2019] [Indexed: 01/10/2023]
|
11
|
Impairment of human neural crest cell migration by prolonged exposure to interferon-beta. Arch Toxicol 2017; 91:3385-3402. [PMID: 28365849 PMCID: PMC5608792 DOI: 10.1007/s00204-017-1966-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/21/2017] [Indexed: 12/31/2022]
Abstract
Human cell-based toxicological assays have been used successfully to detect known toxicants, and to distinguish them from negative controls. However, there is at present little experience on how to deal with hits from screens of compounds with yet unknown hazard. As a case study to this issue, we characterized human interferon-beta (IFNβ) as potential developmental toxicant affecting neural crest cells (NCC). The protein was identified as a hit during a screen of clinically used drugs in the ‘migration inhibition of neural crest’ (MINC) assay. Concentration–response studies in the MINC combined with immunocytochemistry and mRNA quantification of cellular markers showed that IFNβ inhibited NCC migration at concentrations as low as 20 pM. The effective concentrations found here correspond to levels found in human plasma, and they were neither cytostatic nor cytotoxic nor did they did they affect the differentiation state and overall phenotype of NCC. Data from two other migration assays confirmed that picomolar concentration of IFNβ reduced the motility of NCC, while other interferons were less potent. The activation of JAK kinase by IFNβ, as suggested by bioinformatics analysis of the transcriptome changes, was confirmed by biochemical methods. The degree and duration of pathway activation correlated with the extent of migration inhibition, and pharmacological block of this signaling pathway before, or up to 6 h after exposure to the cytokine prevented the effects of IFNβ on migration. Thus, the reduction of vital functions of human NCC is a hitherto unknown potential hazard of endogenous or pharmacologically applied interferons.
Collapse
|
12
|
Clift MJD, Fytianos K, Vanhecke D, Hočevar S, Petri-Fink A, Rothen-Rutishauser B. A novel technique to determine the cell type specific response within an in vitro co-culture model via multi-colour flow cytometry. Sci Rep 2017; 7:434. [PMID: 28348366 PMCID: PMC5428288 DOI: 10.1038/s41598-017-00369-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/23/2017] [Indexed: 11/10/2022] Open
Abstract
Determination of the cell type specific response is essential towards understanding the cellular mechanisms associated with disease states as well as assessing cell-based targeting of effective therapeutic agents. Recently, there have been increased calls for advanced in vitro multi-cellular models that provide reliable and valuable tools correlative to in vivo. In this pursuit the ability to assess the cell type specific response is imperative. Herein, we report a novel approach towards resolving each specific cell type of a multi-cellular model representing the human lung epithelial tissue barrier via multi-colour flow cytometry (FACS). We proved via ≤ five-colour FACS that the manipulation of this in vitro model allowed each cell type to be resolved with no impact upon cell viability. Subsequently, four-colour FACS verified the ability to determine the biochemical effect (e.g. oxidative stress) of each specific cell type. This technique will be vital in gaining information upon cellular mechanics when using next-level, multi-cellular in vitro strategies.
Collapse
Affiliation(s)
- Martin J D Clift
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland. .,In Vitro Toxicology Group, Swansea University Medical School, Wales, UK.
| | - Kleanthis Fytianos
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Dimitri Vanhecke
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Sandra Hočevar
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Alke Petri-Fink
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland.,Department of Chemistry, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
13
|
Activation of the JAK-STAT Signaling Pathway after In Vitro Stimulation with IFNß in Multiple Sclerosis Patients According to the Therapeutic Response to IFNß. PLoS One 2017; 12:e0170031. [PMID: 28103257 PMCID: PMC5245989 DOI: 10.1371/journal.pone.0170031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 12/26/2016] [Indexed: 11/19/2022] Open
Abstract
Interferon beta (IFNß) is a common treatment used for multiple sclerosis (MS) which acts through the activation of the JAK-STAT pathway. However, this therapy is not always effective and currently there are no reliable biomarkers to predict therapeutic response. We postulate that the heterogeneity in the response to IFNß therapy could be related to differential activation patterns of the JAK-STAT signaling pathway. Our aim was to evaluate the basal levels and the short term activation of this pathway after IFNß stimulation in untreated and IFNß treated patients, as well as according to therapeutic response. Therefore, cell surface levels of IFNAR subunits (IFNAR1 and IFNAR2) and the activated forms of STAT1 and STAT2 were assessed in peripheral blood mononuclear cells from MS patients by flow cytometry. Basal levels of each of the markers strongly correlated with the expression of the others in untreated patients, but many of these correlations lost significance in treated patients and after short term activation with IFNß. Patients who had undergone IFNß treatment showed higher basal levels of IFNAR1 and pSTAT1, but a reduced response to in vitro exposure to IFNß. Conversely, untreated patients, with lower basal levels, showed a greater ability of short term activation of this pathway. Monocytes from responder patients had lower IFNAR1 levels (p = 0.039) and higher IFNAR2 levels (p = 0.035) than non-responders just after IFNß stimulation. A cluster analysis showed that levels of IFNAR1, IFNAR2 and pSTAT1-2 in monocytes grouped 13 out of 19 responder patients with a similar expression pattern, showing an association of this pattern with the phenotype of good response to IFNß (p = 0.013). Our findings suggest that an activation pattern of the IFNß signaling pathway in monocytes could be associated with a clinical phenotype of good response to IFNß treatment and that a differential modulation of the IFNAR subunits in monocytes could be related with treatment effectiveness.
Collapse
|
14
|
Dixon BJ, Chen D, Zhang Y, Flores J, Malaguit J, Nowrangi D, Zhang JH, Tang J. Intranasal Administration of Interferon Beta Attenuates Neuronal Apoptosis via the JAK1/STAT3/BCL-2 Pathway in a Rat Model of Neonatal Hypoxic-Ischemic Encephalopathy. ASN Neuro 2016; 8:1759091416670492. [PMID: 27683877 PMCID: PMC5043595 DOI: 10.1177/1759091416670492] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/25/2016] [Accepted: 08/22/2016] [Indexed: 12/17/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is an injury that often leads to detrimental neurological deficits. Currently, there are no established therapies for HIE and it is critical to develop treatments that provide protection after HIE. The objective of this study was to investigate the ability of interferon beta (IFNβ) to provide neuroprotection and reduce apoptosis after HIE. Postnatal Day 10 rat pups were subjected to unilateral carotid artery ligation followed by 2.5 hr of exposure to hypoxia (8% O2). Intranasal administration of human recombinant IFNβ occurred 2 hr after HIE and infarct volume, body weight, neurobehavioral tests, histology, immunohistochemistry, brain water content, blood-brain barrier permeability, enzyme-linked immunosorbent assay, and Western blot were all used to evaluate various parameters. The results showed that both IFNβ and the Type 1 interferon receptor expression decreases after HIE. Intranasal administration of human recombinant IFNβ was able to be detected in the central nervous system and was able to reduce brain infarction volumes and improve neurological behavior tests 24 hr after HIE. Western blot analysis also revealed that human recombinant IFNβ treatment stimulated Stat3 and Bcl-2 expression leading to a decrease in cleaved caspase-3 expression after HIE. Positive Fluoro-Jade C staining also demonstrated that IFNβ treatment was able to decrease neuronal apoptosis. Furthermore, the beneficial effects of IFNβ treatment were reversed when a Stat3 inhibitor was applied. Also an intraperitoneal administration of human recombinant IFNβ into the systemic compartment was unable to confer the same protective effects as intranasal IFNβ treatment.
Collapse
Affiliation(s)
- Brandon J Dixon
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA
| | - Di Chen
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA
| | - Yang Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA
| | - Jerry Flores
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA
| | - Jay Malaguit
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA
| | - Derek Nowrangi
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA Department of Neurosurgery, Loma Linda University School of Medicine, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, CA, USA
| |
Collapse
|
15
|
Tisato V, Gonelli A, Voltan R, Secchiero P, Zauli G. Clinical perspectives of TRAIL: insights into central nervous system disorders. Cell Mol Life Sci 2016; 73:2017-27. [PMID: 26910728 PMCID: PMC4834097 DOI: 10.1007/s00018-016-2164-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/01/2022]
Abstract
The TNF-related apoptosis inducing ligand TRAIL is a member of the TNF superfamily that has been firstly studied and evaluated for its anti-cancer activity, and the insights into its biology have already led to the identification of several TRAIL-based anticancer strategies with strong clinical therapeutic potentials. Nonetheless, the TRAIL system is far more complex and it can lead to a wider range of biological effects other than the ability of inducing apoptosis in cancer cells. By virtue of the different receptors and the different signalling pathways involved, TRAIL plays indeed a role in the regulation of different processes of the innate and adaptive immune system and this feature makes it an intriguing molecule under consideration in the development/progression/treatment of several immunological disorders. In this context, central nervous system represents a peculiar anatomic site where, despite its "status" of immune-privileged site, both innate and adaptive inflammatory responses occur and are involved in several pathological conditions. A number of studies have evaluated the role of TRAIL and of TRAIL-related pathways as pro-inflammatory or protective stimuli, depending on the specific pathological condition, confirming a twofold nature of this molecule. In this light, the aim of this review is to summarize the main preclinical evidences of the potential/involvement of TRAIL molecule and TRAIL pathways for the treatment of central nervous system disorders and the key suggestions coming from their assessment in preclinical models as proof of concept for future clinical studies.
Collapse
Affiliation(s)
- Veronica Tisato
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy.
| | - Arianna Gonelli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Rebecca Voltan
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| |
Collapse
|
16
|
López-Gómez C, Oliver-Martos B, Pinto-Medel MJ, Suardiaz M, Reyes-Garrido V, Urbaneja P, Fernández Ó, Leyva L. TRAIL and TRAIL receptors splice variants during long-term interferon β treatment of patients with multiple sclerosis: evaluation as biomarkers for therapeutic response. J Neurol Neurosurg Psychiatry 2016; 87:130-7. [PMID: 25736057 PMCID: PMC4752633 DOI: 10.1136/jnnp-2014-309932] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/06/2015] [Indexed: 01/16/2023]
Abstract
OBJECTIVE We aimed to assess the effects of interferon β (IFNβ) treatment on the expression of the splice variants of the Tumour necrosis factor-Related Apoptosis Inducing Ligand (TRAIL) and its receptors in different cell subpopulations (CD14+, CD4+ and CD8+) from patients with multiple sclerosis (MS), and to determine whether this expression discriminated responders from non-responders to IFNβ therapy. METHODS We examined mRNA expression of the TRAIL and TRAIL receptors variants in patients with MS, at baseline and after one year of IFNβ therapy, according to responsiveness to this drug. RESULTS Long-term therapy with IFNβ increased the expression of TRAIL-α in T cell subsets exclusively from responders and decreased the expression of the isoform 2 of TRAILR-2 in monocytes from responders as well as non-responders. Lower expression of TRAIL-α, and higher expression of TRAIL-β in monocytes and T cells, was found before the onset of IFNβ therapy in patients who will subsequently become responders. Baseline expression of TRAILR-1 was also significantly higher in monocytes and CD4+ T cells from responders. CONCLUSIONS The present study shows that long-term IFNβ treatment has a direct influence on TRAIL-α and TRAILR-2 isoform 2 expression. Besides, receiver operating characteristic analysis revealed that the baseline expression of TRAIL-α in monocytes and T cells, and that of TRAILR-1 in monocytes and CD4+ T cells, showed a predictive value of the clinical response to IFNβ therapy, pointing to a role of TRAIL system in the mechanism of action of IFNβ in MS that will need further investigation.
Collapse
Affiliation(s)
- Carlos López-Gómez
- Research Laboratory, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - Begoña Oliver-Martos
- Research Laboratory, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - María-Jesús Pinto-Medel
- Research Laboratory, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - Margarita Suardiaz
- Research Laboratory, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - Virginia Reyes-Garrido
- Department of Neurology, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - Patricia Urbaneja
- Department of Neurology, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - Óscar Fernández
- Department of Neurology, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| | - Laura Leyva
- Research Laboratory, UGCI Neurociencias Clínicas, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga, Spain
| |
Collapse
|
17
|
Targeted mutations in Val101 and Arg27 interferon beta protein increase its transcriptional and translational activities. Cytokine 2015; 78:1-6. [PMID: 26615566 DOI: 10.1016/j.cyto.2015.11.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/11/2015] [Accepted: 11/14/2015] [Indexed: 01/14/2023]
Abstract
Interferon β (IFNβ) is the most prescribed drug that has been used frequently for the treatment of multiple sclerosis (MS) patients. The aim of this study is to improve the production of IFNβ by induction of site directed mutagenesis. Accordingly, recombinant constructs were designed in order to enhance the expression of IFNβ mRNA and protein. The recombinant plasmids were transfected to the CHO cell line, following RNA extractions and cDNA synthesis. The effects of recombinant constructs were analyzed by real time PCR, ELISA and MTT assay. Transfected samples with either IFNβ101 or IFNβ101+27 have shown 11.55 and 2.26 fold elevation and over-expression compare to the wild type construct respectively. Our data also indicated that the IFNβ101 and IFNβ101+27 constructs increase IFNβ protein expression more than 2.2 and 4.5 fold, respectively compared to the control group. It could be concluded that the substitution of Phe in the codon 101 position, which may increase the binding activity of IFNβ with its receptors and introduction of an additional N glycosylation site (Asn-X-Thr) in the position 27 of IFNβ protein may cause such an effect. The proliferative activity of transfected cells by a recombinant IFNβ101 decreases in comparison to the wild type, although it was not statistically significant. Over-expression of IFNβ in such a level is promising not only for the patients but also for the pharmaceutical industries.
Collapse
|
18
|
D'Ambrosio A, Pontecorvo S, Colasanti T, Zamboni S, Francia A, Margutti P. Peripheral blood biomarkers in multiple sclerosis. Autoimmun Rev 2015; 14:1097-110. [PMID: 26226413 DOI: 10.1016/j.autrev.2015.07.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/23/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis is the most common autoimmune disorder affecting the central nervous system. The heterogeneity of pathophysiological processes in MS contributes to the highly variable course of the disease and unpredictable response to therapies. The major focus of the research on MS is the identification of biomarkers in biological fluids, such as cerebrospinal fluid or blood, to guide patient management reliably. Because of the difficulties in obtaining spinal fluid samples and the necessity for lumbar puncture to make a diagnosis has reduced, the research of blood-based biomarkers may provide increasingly important tools for clinical practice. However, currently there are no clearly established MS blood-based biomarkers. The availability of reliable biomarkers could radically alter the management of MS at critical phases of the disease spectrum, allowing for intervention strategies that may prevent evolution to long-term neurological disability. This article provides an overview of this research field and focuses on recent advances in blood-based biomarker research.
Collapse
Affiliation(s)
- Antonella D'Ambrosio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Pontecorvo
- Multiple Sclerosis Center of Department of Neurology and Psychiatry of "Sapienza" University of Rome, Italy
| | - Tania Colasanti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Zamboni
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Ada Francia
- Multiple Sclerosis Center of Department of Neurology and Psychiatry of "Sapienza" University of Rome, Italy
| | - Paola Margutti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
19
|
Huber AK, Duncker PC, Irani DN. The conundrum of interferon-β non-responsiveness in relapsing-remitting multiple sclerosis. Cytokine 2015; 74:228-36. [PMID: 25691330 DOI: 10.1016/j.cyto.2015.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 01/10/2015] [Indexed: 01/01/2023]
Abstract
A series of controlled clinical trials have shown that exogenous interferon-beta (IFN-β) benefits patients with relapsing-remitting multiple sclerosis (RRMS) by reducing relapse rate, disability progression, and the formation of new brain and spinal cord lesions on magnetic resonance imaging (MRI) scans. Unfortunately, however, the effectiveness of IFN-β is limited in this setting by the occurrence of treatment non-responsiveness in nearly 25% of patients. Furthermore, clinicians who care for RRMS patients remain unable to accurately identify IFN-β non-responders prior to the initiation of therapy, causing delays in the use of alternative treatments and sometimes requiring that patients turn to medications with more significant side effects to control their disease. Progress has been made toward understanding how both endogenous and exogenous IFN-β act to slow RRMS as well as the related mouse model, experimental autoimmune encephalomyelitis (EAE). Most studies point to its inhibitory actions on circulating immune cells as being important for suppressing both disorders, but multiple potential target cells and inflammatory pathways have been implicated and those essential to confer its benefits remain undefined. This review focuses on the role of both endogenous and exogenous IFN-β in RRMS, paying particular attention to the issue of why certain individuals appear refractory to its disease-modifying effects. A continued goal in this field remains the identification of a convenient biomarker that accurately predicts IFN-β treatment non-responsiveness in individual RRMS patients. Development of such an assay will allow clinicians to customize therapy for patients with this complex disorder.
Collapse
Affiliation(s)
- Amanda K Huber
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Patrick C Duncker
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David N Irani
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
20
|
Severa M, Rizzo F, Giacomini E, Salvetti M, Coccia EM. IFN-β and multiple sclerosis: cross-talking of immune cells and integration of immunoregulatory networks. Cytokine Growth Factor Rev 2014; 26:229-39. [PMID: 25498525 DOI: 10.1016/j.cytogfr.2014.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/05/2014] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is characterized by autoimmune inflammation affecting the central nervous system and subsequent neurodegeneration. Historically, damage was thought to be mediated exclusively by auto-antigen-activated pro-inflammatory T cells. However, more recently, we are gaining increasing knowledge on the pathogenic role played in MS by B cells, dendritic cells and monocytes. IFN-β therapy was one the first approved therapy for MS for its ability to reduce relapse rate and MRI lesion activity and to significantly decrease risk of disability progression. IFN-β-mediated mechanisms of action, even if not completely understood, mainly rely on its multifaceted pleiotropic effects resulting in sustained anti-inflammatory properties directed toward almost every immune cell type. Here, we will discuss in detail literature data characterizing the pathogenic activity of the different immune cell subsets involved in MS pathogenesis and how IFN-β therapy regulates their function by modulating bystander responses. We believe that the effectiveness of this drug in MS treatment, even if in use for a long time, can unveil new insights on this disease and still teach a lesson to researchers in the MS field.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Fabiana Rizzo
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Giacomini
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Salvetti
- Centre for Experimental Neurological Therapies (CENTERS) - Neurology and Department of Neurosciences, Mental Health and Sensory Organs; Sapienza, University of Rome, S. Andrea Hospital Site, Italy
| | - Eliana M Coccia
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
21
|
Interferon-β1a protects neurons against mitochondrial toxicity via modulation of STAT1 signaling: electrophysiological evidence. Neurobiol Dis 2013; 62:387-93. [PMID: 24135008 DOI: 10.1016/j.nbd.2013.09.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 09/09/2013] [Accepted: 09/24/2013] [Indexed: 11/22/2022] Open
Abstract
Multiple sclerosis, one of the main causes of non-traumatic neurological disability in young adults, is an inflammatory and neurodegenerative disorder of the central nervous system. Although the pathogenesis of neuroaxonal damage occurring during the course of the disease is still largely unknown, there is accumulating evidence highlighting the potential role of mitochondria in multiple sclerosis-associated neuronal degeneration. The aim of the present study was to investigate, by utilizing electrophysiological techniques in brain striatal slices, the potential protective effects of interferon-β1a, one of the most widely used medication for multiple sclerosis, against acute neuronal dysfunction induced by mitochondrial toxins. Interferon-β1a was found to exert a dose-dependent protective effect against the progressive loss of striatal field potential amplitude induced by the mitochondrial complex I inhibitor rotenone. Interferon-β1a also reduced the generation of the rotenone-induced inward current in striatal spiny neurons. Conversely, interferon-β1a did not influence the electrophysiological effects of the mitochondrial complex II inhibitor 3-nitropropionic acid. The protective effect of interferon-β1a against mitochondrial complex I inhibition was found to be dependent on the activation of STAT1 signaling. Conversely, endogenous dopamine depletion and the modulation of the p38 MAPK and mTOR pathways did not influence the effects of interferon-β1a. During experimental autoimmune encephalomyelitis (EAE) striatal rotenone toxicity was enhanced but the protective effect of interferon-β1a was still evident. These results support future studies investigating the role played by specific intracellular signaling pathways in mediating the potential link among inflammation, mitochondrial impairment and neuroaxonal degeneration in multiple sclerosis.
Collapse
|
22
|
Ahmed CM, Johnson HM. The role of a non-canonical JAK-STAT pathway in IFN therapy of poxvirus infection and multiple sclerosis: An example of Occam's Broom? JAKSTAT 2013; 2:e26227. [PMID: 24416655 PMCID: PMC3876437 DOI: 10.4161/jkst.26227] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/19/2013] [Accepted: 08/20/2013] [Indexed: 11/21/2022] Open
Abstract
Signaling by cytokines such as the interferons (IFNs) involves Janus kinases (JAKs) and signal transducer and activator of transcription (STAT) transcription factors. The beauty of the classical model of JAK-STAT signaling is its simplicity in that JAK-activated STATs in the nucleus are responsible for specific gene activation. The fact that many ligands, growth factors, and hormones use the same STAT transcription factors, but exert different functions at the level of the cell, tissue, and organ would suggest significant shortcomings in the classical model. Our studies have resulted in the development of a non-canonical, more complex model of IFN signaling that bears a striking resemblance to that of steroid hormone (SH)/steroid receptor (SR) signaling. Thus, both types I and II IFN signaling involves nuclear translocation of complexed ligand, receptor, activated JAKs, and activated STATs to the promoters of the genes that are specifically activated by the IFNs, where they are involved in specific gene activation and epigenetic remodeling. Receptor intracellular domains play an important role in binding the C-terminus of the IFNs, which is the basis for our development of IFN mimetics. The IFN mimetics are not recognized by poxvirus decoy receptors, since the decoy receptors compete for extracellular binding and not intracellular binding. Further, the type I IFN mimetics provide therapeutic protection against experimental allergic encephalomyelitis (EAE), a model of multiple sclerosis, without the side effects. Extracellular receptor binding by intact IFN is the primary reason for undesirable side effects of flu-like symptoms, bone-marrow suppression, and weight loss. The non-canonical model of IFN signaling thus provides insight into the specificity of such signaling and a mechanism for development of IFN mimetics. It is our contention that this model applies to other cytokines.
Collapse
Affiliation(s)
- Chulbul M Ahmed
- Department of Microbiology and Cell Science; University of Florida; Gainesville, FL USA
| | - Howard M Johnson
- Department of Microbiology and Cell Science; University of Florida; Gainesville, FL USA
| |
Collapse
|
23
|
Nuclear factor kappa B (NF-κB) in multiple sclerosis pathology. Trends Mol Med 2013; 19:604-13. [PMID: 24007818 DOI: 10.1016/j.molmed.2013.08.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/12/2013] [Accepted: 08/02/2013] [Indexed: 12/15/2022]
Abstract
The nuclear factor kappa B (NF-κB) signaling cascade plays a critical role in the regulation of immune and inflammatory responses and has been implicated in the pathogenesis of autoimmune demyelinating diseases such as multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), the main animal model of MS. NF-κB is essential for peripheral immune cell activation and the induction of pathology, but also plays crucial roles in resident cells of the central nervous system (CNS) during disease development. Here we review recent evidence clarifying the role of NF-κB in the different cell compartments contributing to MS pathology and its implications for the development of therapeutic strategies for the treatment of MS and other demyelinating pathologies of the CNS.
Collapse
|
24
|
Giacomini E, Severa M, Rizzo F, Mechelli R, Annibali V, Ristori G, Riccieri V, Salvetti M, Coccia EM. IFN-β therapy modulates B-cell and monocyte crosstalk via TLR7 in multiple sclerosis patients. Eur J Immunol 2013; 43:1963-72. [DOI: 10.1002/eji.201243212] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/19/2013] [Accepted: 04/25/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Elena Giacomini
- Department of Infectious, Parasitic and Immune-mediated Diseases; Istituto Superiore di Sanità; Rome; Italy
| | - Martina Severa
- Department of Infectious, Parasitic and Immune-mediated Diseases; Istituto Superiore di Sanità; Rome; Italy
| | - Fabiana Rizzo
- Department of Infectious, Parasitic and Immune-mediated Diseases; Istituto Superiore di Sanità; Rome; Italy
| | - Rosella Mechelli
- Centre for Experimental Neurological Therapies (CENTERS), S. Andrea Hospital-site; Sapienza University; Rome; Italy
| | - Viviana Annibali
- Centre for Experimental Neurological Therapies (CENTERS), S. Andrea Hospital-site; Sapienza University; Rome; Italy
| | - Giovanni Ristori
- Centre for Experimental Neurological Therapies (CENTERS), S. Andrea Hospital-site; Sapienza University; Rome; Italy
| | - Valeria Riccieri
- Internal Medicine and Medical Specialities Department; Sapienza University; Rome; Italy
| | - Marco Salvetti
- Centre for Experimental Neurological Therapies (CENTERS), S. Andrea Hospital-site; Sapienza University; Rome; Italy
| | - Eliana Marina Coccia
- Department of Infectious, Parasitic and Immune-mediated Diseases; Istituto Superiore di Sanità; Rome; Italy
| |
Collapse
|
25
|
López-Gómez C, Pino-Ángeles A, Órpez-Zafra T, Pinto-Medel MJ, Oliver-Martos B, Ortega-Pinazo J, Arnáiz C, Guijarro-Castro C, Varadé J, Álvarez-Lafuente R, Urcelay E, Sánchez-Jiménez F, Fernández Ó, Leyva L. Candidate gene study of TRAIL and TRAIL receptors: association with response to interferon beta therapy in multiple sclerosis patients. PLoS One 2013; 8:e62540. [PMID: 23658636 PMCID: PMC3639207 DOI: 10.1371/journal.pone.0062540] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 03/22/2013] [Indexed: 12/31/2022] Open
Abstract
TRAIL and TRAIL Receptor genes have been implicated in Multiple Sclerosis pathology as well as in the response to IFN beta therapy. The objective of our study was to evaluate the association of these genes in relation to the age at disease onset (AAO) and to the clinical response upon IFN beta treatment in Spanish MS patients. We carried out a candidate gene study of TRAIL, TRAILR-1, TRAILR-2, TRAILR-3 and TRAILR-4 genes. A total of 54 SNPs were analysed in 509 MS patients under IFN beta treatment, and an additional cohort of 226 MS patients was used to validate the results. Associations of rs1047275 in TRAILR-2 and rs7011559 in TRAILR-4 genes with AAO under an additive model did not withstand Bonferroni correction. In contrast, patients with the TRAILR-1 rs20576-CC genotype showed a better clinical response to IFN beta therapy compared with patients carrying the A-allele (recessive model: p = 8.88×10−4, pc = 0.048, OR = 0.30). This SNP resulted in a non synonymous substitution of Glutamic acid to Alanine in position 228 (E228A), a change previously associated with susceptibility to different cancer types and risk of metastases, suggesting a lack of functionality of TRAILR-1. In order to unravel how this amino acid change in TRAILR-1 would affect to death signal, we performed a molecular modelling with both alleles. Neither TRAIL binding sites in the receptor nor the expression levels of TRAILR-1 in peripheral blood mononuclear cell subsets (monocytes, CD4+ and CD8+ T cells) were modified, suggesting that this SNP may be altering the death signal by some other mechanism. These findings show a role for TRAILR-1 gene variations in the clinical outcome of IFN beta therapy that might have relevance as a biomarker to predict the response to IFN beta in MS.
Collapse
Affiliation(s)
- Carlos López-Gómez
- Research Laboratory. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - Almudena Pino-Ángeles
- Department of Molecular Biology and Biochemistry, University of Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Málaga, Spain
| | - Teresa Órpez-Zafra
- Research Laboratory. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - María Jesús Pinto-Medel
- Research Laboratory. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - Begoña Oliver-Martos
- Research Laboratory. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - Jesús Ortega-Pinazo
- Research Laboratory. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - Carlos Arnáiz
- Department of Neurology. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | - Cristina Guijarro-Castro
- Department of Neurology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - Jezabel Varadé
- Multiple Sclerosis Unit, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - Roberto Álvarez-Lafuente
- Multiple Sclerosis Unit, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - Elena Urcelay
- Multiple Sclerosis Unit, Hospital Clínico San Carlos, IdISSC, Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - Francisca Sánchez-Jiménez
- Department of Molecular Biology and Biochemistry, University of Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Málaga, Spain
| | - Óscar Fernández
- Department of Neurology. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
| | - Laura Leyva
- Research Laboratory. Clinical Neurosciences Institute, Hospital Regional Universitario Carlos Haya, Málaga, Spain
- Red Española de Esclerosis Múltiple (REEM RD 07/0060), Málaga, Spain
- * E-mail:
| |
Collapse
|
26
|
Bustamante MF, Nurtdinov RN, Río J, Montalban X, Comabella M. Baseline gene expression signatures in monocytes from multiple sclerosis patients treated with interferon-beta. PLoS One 2013; 8:e60994. [PMID: 23637780 PMCID: PMC3630153 DOI: 10.1371/journal.pone.0060994] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 03/05/2013] [Indexed: 11/19/2022] Open
Abstract
Background A relatively large proportion of relapsing-remitting multiple sclerosis (RRMS) patients do not respond to interferon-beta (IFNb) treatment. In previous studies with peripheral blood mononuclear cells (PBMC), we identified a subgroup of IFNb non-responders that was characterized by a baseline over-expression of type I IFN inducible genes. Additional mechanistic experiments carried out in IFNb non-responders suggested a selective alteration of the type I IFN signaling pathway in the population of blood monocytes. Here, we aimed (i) to investigate whether the type I IFN signaling pathway is up-regulated in isolated monocytes from IFNb non-responders at baseline; and (ii) to search for additional biological pathways in this cell population that may be implicated in the response to IFNb treatment. Methods Twenty RRMS patients classified according to their clinical response to IFNb treatment and 10 healthy controls were included in the study. Monocytes were purified from PBMC obtained before treatment by cell sorting and the gene expression profiling was determined with oligonucleotide microarrays. Results and discussion Purified monocytes from IFNb non-responders were characterized by an over-expression of type I IFN responsive genes, which confirms the type I IFN signature in monocytes suggested from previous studies. Other relevant signaling pathways that were up-regulated in IFNb non-responders were related with the mitochondrial function and processes such as protein synthesis and antigen presentation, and together with the type I IFN signaling pathway, may also be playing roles in the response to IFNb.
Collapse
Affiliation(s)
- Marta F. Bustamante
- Servei de Neurología/Neuroimmunología. Centre d’Esclerosi Múltiple de Catalunya, Cemcat. Hospital Universitari Vall dHebron (HUVH), Barcelona, Spain
| | - Ramil N. Nurtdinov
- Servei de Neurología/Neuroimmunología. Centre d’Esclerosi Múltiple de Catalunya, Cemcat. Hospital Universitari Vall dHebron (HUVH), Barcelona, Spain
| | - Jordi Río
- Servei de Neurología/Neuroimmunología. Centre d’Esclerosi Múltiple de Catalunya, Cemcat. Hospital Universitari Vall dHebron (HUVH), Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurología/Neuroimmunología. Centre d’Esclerosi Múltiple de Catalunya, Cemcat. Hospital Universitari Vall dHebron (HUVH), Barcelona, Spain
| | - Manuel Comabella
- Servei de Neurología/Neuroimmunología. Centre d’Esclerosi Múltiple de Catalunya, Cemcat. Hospital Universitari Vall dHebron (HUVH), Barcelona, Spain
- * E-mail:
| |
Collapse
|