1
|
Protopapa M, Schmaul S, Schraad M, Pape K, Zipp F, Bittner S, Uphaus T. Lower leukocytes pretreatment as a possible risk factor for therapy-induced leukopenia in interferon-beta-treated patients with multiple sclerosis. Ther Adv Neurol Disord 2024; 17:17562864241286497. [PMID: 39479177 PMCID: PMC11523160 DOI: 10.1177/17562864241286497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 09/06/2024] [Indexed: 11/02/2024] Open
Abstract
Background Interferon-beta (IFN-β) still plays a fundamental role in immunomodulation of people with multiple sclerosis (MS) with low disease activity and in clinically isolated syndrome (CIS). In 2014, pegylated (PEG) interferon was licensed by the European Medicines Agency (EMA) for relapsing-remitting MS (RRMS), enabling a lower dosing frequency. Objectives Our retrospective study compares laboratory findings and adverse events between subcutaneous (sc.) PEG-IFN-β-1a and IFN-β-1a in RRMS and CIS patients. Design Patients with CIS or RRMS fulfilling the revised McDonald criteria from 2017 visiting the neurology department of the University Medical Center of the Johannes Gutenberg University Mainz from 2010 to 2019 and treated with sc. PEG-IFN-β-1a or sc. IFN-β-1a (n = 202) were screened for eligibility. Patients who underwent regular laboratory controls in-house were included in our analysis (n = 128). Methods We evaluate disease progression through clinical examination, relapse history, and magnetic resonance imaging (MRI) disease activity (gadolinium-enhancing or new T2 lesions). Relevant laboratory findings such as leukopenia (leukocyte count < 3.5/nl) and neutropenia (neutrophil count <43% of lymphocytes or <1500/µl) were assessed. Telephone interviews evaluated the side effects of the respective medication. A subgroup of patients was analyzed regarding neutrophil quantities and qualities. Results Patients treated with sc. PEG-IFN-β-1a had significantly lower leukocyte counts (p = 0.046) and higher incidences of leukopenia (p = 0.006) and neutropenia (p = 0.03) compared to sc. IFN-β-1a. Clinical and MRI disease activity showed no significant differences, but people treated with sc. PEG-IFN-β-1a reported more common adverse events such as joint/muscle pain, injection-site reaction, and infections. No serious adverse events were reported. Conclusion Treatment with sc. PEG-IFN-β-1a compared to unpegylated sc. IFN-β resulted in a significantly greater reduction in leukocyte and neutrophil levels with a higher incidence of side effects. We suggest mandatory monitoring of differential blood counts before and during treatment.
Collapse
Affiliation(s)
- Maria Protopapa
- Department of Neurology, Focus Program Translational Neuroscience, (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Samantha Schmaul
- Department of Neurology, Focus Program Translational Neuroscience, (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Muriel Schraad
- Department of Neurology, Focus Program Translational Neuroscience, (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katrin Pape
- Department of Neurology, Focus Program Translational Neuroscience, (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience, (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience, Rhine-Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Timo Uphaus
- Department of Neurology, Focus Program Translational Neuroscience, Rhine-Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, Mainz 55131, Germany
| |
Collapse
|
2
|
KAYA S, UZDİL Z, SHIVAPPA N, HEBERT JR, SÖKÜLMEZ KAYA P, TERZİ M. Dietary Inflammatory Index score and its association with body mass index, body fat percentage, body fat mass, and lipid profile in patients with multiple sclerosis. Turk J Med Sci 2023; 53:1155-1165. [PMID: 38813018 PMCID: PMC10763805 DOI: 10.55730/1300-0144.5681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/26/2023] [Accepted: 06/21/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim Multiple sclerosis (MS) may cause modifications in body composition, particularly for body fat associated with obesity and some biochemical parameters such as lipid profiles. We investigated whether there is a link between the inflammatory contents of diets and body composition and lipid profiles in patients with MS. Materials and methods This was a cross-sectional study that included 85 MS patients. The study data of the patients were collected in the Neurology Clinic of Ondokuz Mayıs University's Health Practice and Research Center. The data included demographic characteristics; anthropometric measurements such as body weight, height, body mass index, waist circumference, hip circumference, body fat mass, body fat-free mass, and waist-hip ratio; and biochemical parameters such as high-density lipoprotein cholesterol (HDL-c), low-density lipoprotein cholesterol, triglyceride, and total cholesterol. Results The body fat percentages of the patients were higher among those with proinflammatory diets (p < 0.05). Body fat percentage had a positive and very weak correlation with the Dietary Inflammatory Index (DII) score (rho = 0.206 and rho = 0.217, respectively; p < 0.05). HDL-c levels were higher in the group with high DII scores and there was a positive and weak correlation between HDL-c and DII scores (rho = 0.307, p < 0.05). Crude and adjusted linear regression models showed that the effect of HDL-c on DII scores was significant (p < 0.05). Conclusion We showed that DII scores, associated with the inflammatory potential of the diet and proinflammatory diets, may be associated with adiposity in MS patients and can be used from a clinical point of view for assessment.
Collapse
Affiliation(s)
- Seda KAYA
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara University, Ankara,
Turkiye
| | - Zeynep UZDİL
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ondokuz Mayıs University, Samsun,
Turkiye
| | - Nitin SHIVAPPA
- Department of Epidemiology and Biostatistics and the Cancer Prevention and Control Program, University of South Carolina, Columbia, South Carolina,
USA
- Connecting Health Innovations, LLC, Columbia, South Carolina,
USA
| | - James R. HEBERT
- Department of Epidemiology and Biostatistics and the Cancer Prevention and Control Program, University of South Carolina, Columbia, South Carolina,
USA
- Connecting Health Innovations, LLC, Columbia, South Carolina,
USA
| | - Pınar SÖKÜLMEZ KAYA
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ondokuz Mayıs University, Samsun,
Turkiye
| | - Murat TERZİ
- Department of Neurology, Faculty of Medicine, Ondokuz Mayıs University, Samsun,
Turkiye
| |
Collapse
|
3
|
Therapeutic potential of the target on NLRP3 inflammasome in multiple sclerosis. Pharmacol Ther 2021; 227:107880. [PMID: 33901504 DOI: 10.1016/j.pharmthera.2021.107880] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Inflammasomes are multi-protein macromolecular complexes that typically comprise of three units, a sensor, an adaptor and procaspase-1. The assembly of each inflammasome is dictated by a unique pattern recognition receptors (PRRs) in response to pathogen-associated molecular patterns (PAMPs) or other endogenous danger-associated molecular patterns (DAMPs) in the cytosol of the host cells, and promote the maturation and secretion of IL-1β and IL-18 during the inflammatory process. Specific inflammasomes are involved in the host defense response against different pathogens, and the latter have evolved multiple corresponding mechanisms to inhibit inflammasome activation. The nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome is the best understood in terms of molecular mechanisms, and is a promising therapeutic target in immune-related disorders. Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination of white matter in the central nervous system, increased levels of IL-1β in the cerebrospinal fluid (CSF) of relapsed patients, and deposition of caspase-1 in the spinal cord. The direct involvement of the NLRP3 inflammasome in the occurrence and development of MS was ascertained in the experimental autoimmune encephalomyelitis (EAE) animal model. In this review, we have focused on the mechanisms underlying activation of the NLRP3 inflammasome in MS or EAE, as well as inhibitors that specifically target the complex and alleviate disease progression, in order to unearth new therapeutic strategies against MS.
Collapse
|
4
|
Pozzato M, Micaglio E, Starvaggi Cucuzza C, Cagol A, Galimberti D, Calandrella D, Cinnante C, Pappone C, Zanussi M, Meola G, Scarpini E, Bresolin N, Martinelli Boneschi F. Case Report: Efficacy of Rituximab in a Patient With Familial Mediterranean Fever and Multiple Sclerosis. Front Neurol 2021; 11:591395. [PMID: 33584496 PMCID: PMC7874168 DOI: 10.3389/fneur.2020.591395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/04/2020] [Indexed: 01/16/2023] Open
Abstract
Familial Mediterranean Fever (FMF) is a genetic autoinflammatory disease characterized by recurrent episodes of fever and serositis caused by mutations in the MEFV gene, while Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the CNS with genetic and environmental etiology. The two diseases rarely occur in association with relevant implications for clinical management and drug choice. In this paper, we present the case of a 53-year-old male with an autosomal dominant FMF since childhood who presented acute paresthesia at the right part of the body. He performed a brain and spinal cord MRI, which showed multiple brain lesions and a gd-enhancing lesion in the cervical spinal cord, and then received a diagnosis of MS. He then started Interferonβ-1a which was effective but not tolerated and caused hepatotoxicity, and then shifted to Rituximab with 3-month clinical and neuroradiological efficacy.
Collapse
Affiliation(s)
- Mattia Pozzato
- Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit & MS Centre, Milan, Italy.,Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Milan, Italy
| | - Emanuele Micaglio
- Arrhythmology and Electrophysiology Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Milan, Italy
| | - Chiara Starvaggi Cucuzza
- Department of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, San Donato Milanese, Italy
| | - Alessandro Cagol
- Department of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, San Donato Milanese, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Centre, University of Milan, Milan, Italy
| | - Daniela Calandrella
- Department of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Humanitas Research Hospital and University, Milan, Italy
| | - Claudia Cinnante
- Neuroradiology Unit, Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Carlo Pappone
- Arrhythmology and Electrophysiology Department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Milan, Italy
| | - Monica Zanussi
- Clinical Genomics-Molecular Genetics Service, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, San Raffaele Hospital, Milan, Italy
| | - Giovanni Meola
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,Department of Neurorehabilitation Sciences, Casa di Cura Privata del Policlinico, Milan, Italy
| | - Elio Scarpini
- Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit & MS Centre, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Centre, University of Milan, Milan, Italy
| | - Nereo Bresolin
- Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit & MS Centre, Milan, Italy.,Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Milan, Italy
| | - Filippo Martinelli Boneschi
- Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit & MS Centre, Milan, Italy.,Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Milan, Italy
| |
Collapse
|
5
|
Prolonged residence of an albumin-IL-4 fusion protein in secondary lymphoid organs ameliorates experimental autoimmune encephalomyelitis. Nat Biomed Eng 2020; 5:387-398. [PMID: 33046864 DOI: 10.1038/s41551-020-00627-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
Interleukin-4 (IL-4) suppresses the development of multiple sclerosis in a murine model of experimental autoimmune encephalomyelitis (EAE). Here, we show that, in mice with EAE, the accumulation and persistence in the lymph nodes and spleen of a systemically administered serum albumin (SA)-IL-4 fusion protein leads to higher efficacy in preventing disease development than the administration of wild-type IL-4 or of the clinically approved drug fingolimod. We also show that the SA-IL-4 fusion protein prevents immune-cell infiltration in the spinal cord, decreases integrin expression in antigen-specific CD4+ T cells, increases the number of granulocyte-like myeloid-derived suppressor cells (and their expression of programmed-death-ligand-1) in spinal cord-draining lymph nodes and decreases the number of T helper 17 cells, a pathogenic cell population in EAE. In mice with chronic EAE, SA-IL-4 inhibits immune-cell infiltration into the spinal cord and completely abrogates immune responses to myelin antigen in the spleen. The SA-IL-4 fusion protein may be prophylactically and therapeutically advantageous in the treatment of multiple sclerosis.
Collapse
|
6
|
Deleu D, Canibaño B, Mesraoua B, Adeli G, Abdelmoneim MS, Ali Y, Elalamy O, Melikyan G, Boshra A. Management of relapsing-remitting multiple sclerosis in Qatar: an expert consensus. Curr Med Res Opin 2020; 36:251-260. [PMID: 31530036 DOI: 10.1080/03007995.2019.1669378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Healthcare systems vary greatly between countries. International, evidence-based guidelines for the management of multiple sclerosis (MS) may need to be adapted for use in particular countries. Two years ago, the authors published a comprehensive consensus guideline for the management of MS in Qatar. Since that time, the availability of disease-modifying treatments for relapsing-remitting MS (RRMS), and our understanding of how to apply those treatments, has increased. The authors present an update to our guidance, focussing on the management of relapsing-remitting RRMS. In particular, the authors consider the optimal use of different DMTs in patients presenting with mild, medium or high disease activity.
Collapse
Affiliation(s)
- Dirk Deleu
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | - Beatriz Canibaño
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | - Boulenouar Mesraoua
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | - Gholamreza Adeli
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | - Mohamed S Abdelmoneim
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | - Yasir Ali
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | - Osama Elalamy
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | - Gayane Melikyan
- Department of Neurology (Neuroscience Institute), Hamad Medical Corporation, Doha, Qatar
| | | |
Collapse
|
7
|
Chung JH, Hong SH, Seo N, Kim TS, An HJ, Lee P, Shin EC, Kim HM. Structure-based glycoengineering of interferon lambda 4 enhances its productivity and anti-viral potency. Cytokine 2019; 125:154833. [PMID: 31479875 PMCID: PMC7129780 DOI: 10.1016/j.cyto.2019.154833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 01/12/2023]
Abstract
Structure-based glycoengineering was applied to improve IFNλ4′s expression level. Model of IFNλ4 signaling complex was used to select de novo N-glycosylation sites. Glycoengineered IFNλ4 variants showed enhanced expression and anti-viral activity.
Interferon lambda 4 (IFNλ4) has been recently known and studied for its role in hepatitis C virus (HCV) infection, but its clinical potential is significantly hampered due to its poor expression in vitro. Our study reports the successful production of IFNλ4 from a mammalian cell line through a glycoengineering and structure-based approach. We introduced de novo N-glycosylation of IFNλ4, guided by structural analysis, and produced IFNλ4 variants in Expi293F that displayed improved expression and potency. To preserve the structure and functionality of IFNλ4, the model structure of the IFNλ4 signaling complex was analyzed and the N-glycosylation candidate sites were selected. The receptor binding activity of engineered IFNλ4 variants and their receptor-mediated signaling pathway were similar to the E. coli version of IFNλ4 (eIFNλ4), while the antiviral activity and induction levels of interferon-stimulated gene (ISG) were all more robust in our variants. Our engineered IFNλ4 variants may be further developed for clinical applications and utilized in basic research to decipher the immunological roles of IFNλ4.
Collapse
Affiliation(s)
- Jae-Hee Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Seon-Hui Hong
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Nari Seo
- Graduate School of Analytical Science & Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Tae-Shin Kim
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Hyun Joo An
- Graduate School of Analytical Science & Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Pedro Lee
- Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Eui-Cheol Shin
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Ho Min Kim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Graduate School of Medical Science & Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; Center for Biomolecular & Cellular Structure, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea.
| |
Collapse
|
8
|
pVAXhsp65 Vaccination Primes for High IL-10 Production and Decreases Experimental Encephalomyelitis Severity. J Immunol Res 2017; 2017:6257958. [PMID: 28321419 PMCID: PMC5339488 DOI: 10.1155/2017/6257958] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/03/2017] [Accepted: 01/23/2017] [Indexed: 11/18/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a demyelinating pathology of the central nervous system (CNS) used as a model to study multiple sclerosis immunopathology. EAE has also been extensively employed to evaluate potentially therapeutic schemes. Considering the presence of an immune response directed to heat shock proteins (hsps) in autoimmune diseases and the immunoregulatory potential of these molecules, we evaluated the effect of a previous immunization with a genetic vaccine containing the mycobacterial hsp65 gene on EAE development. C57BL/6 mice were immunized with 4 pVAXhsp65 doses and 14 days later were submitted to EAE induction by immunization with myelin oligodendrocyte glycoprotein (MOG35–55) emulsified in Complete Freund's Adjuvant. Vaccinated mice presented significant lower clinical scores and lost less body weight. MOG35–55 immunization also determined less inflammation in lumbar spinal cord but did not change CD4+CD25+Foxp3+ T cells frequency in spleen and CNS. Infiltrating cells from the CNS stimulated with rhsp65 produced significantly higher levels of IL-10. These results suggest that the ability of pVAXhsp65 vaccination to control EAE development is associated with IL-10 induction.
Collapse
|
9
|
Intracerebral Hemorrhage in a Patient with Multiple Sclerosis Receiving Interferon Beta-1α. ARCHIVES OF NEUROSCIENCE 2016. [DOI: 10.5812/archneurosci.42758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
10
|
Insights from Mendelian Interferonopathies: Comparison of CANDLE, SAVI with AGS, Monogenic Lupus. J Mol Med (Berl) 2016; 94:1111-1127. [PMID: 27678529 DOI: 10.1007/s00109-016-1465-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 01/13/2023]
Abstract
Autoinflammatory disorders are sterile inflammatory conditions characterized by episodes of early-onset fever and disease-specific patterns of organ inflammation. Recently, the discoveries of monogenic disorders with strong type I interferon (IFN) signatures caused by mutations in proteasome degradation and cytoplasmic RNA and DNA sensing pathways suggest a pathogenic role of IFNs in causing autoinflammatory phenotypes. The IFN response gene signature (IGS) has been associated with systemic lupus erythematosus (SLE) and other autoimmune diseases. In this review, we compare the clinical presentations and pathogenesis of two IFN-mediated autoinflammatory diseases, CANDLE and SAVI, with Aicardi Goutières syndrome (AGS) and monogenic forms of SLE (monoSLE) caused by loss-of-function mutations in complement 1 (C1q) or the DNA nucleases, DNASE1 and DNASE1L3. We outline differences in intracellular signaling pathways that fuel a pathologic type I IFN amplification cycle. While IFN amplification is caused by predominantly innate immune cell dysfunction in SAVI, CANDLE, and AGS, autoantibodies to modified RNA and DNA antigens interact with tissues and immune cells including neutrophils and contribute to IFN upregulation in some SLE patients including monoSLE, thus justifying a grouping of "autoinflammatory" and "autoimmune" interferonopathies. Understanding of the differences in the cellular sources and signaling pathways will guide new drug development and the use of emerging targeted therapies.
Collapse
|
11
|
Tsareva E, Kulakova O, Boyko A, Favorova O. Pharmacogenetics of multiple sclerosis. Pharmacogenet Genomics 2016; 26:103-15. [DOI: 10.1097/fpc.0000000000000194] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
12
|
Interferon Beta: From Molecular Level to Therapeutic Effects. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:343-72. [DOI: 10.1016/bs.ircmb.2016.06.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
13
|
|
14
|
|
15
|
Nobile-Orazio E, Gallia F. Multifocal motor neuropathy: current therapies and novel strategies. Drugs 2014; 73:397-406. [PMID: 23516024 DOI: 10.1007/s40265-013-0029-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multifocal motor neuropathy (MMN) is a purely motor mononeuritis multiplex characterized by the presence of conduction block on motor but not on sensory nerves and by the presence of high titers of anti-GM1 antibodies. Several data point to a pathogenetic role of the immune system in this neuropathy, although this has not yet been proved. Several uncontrolled studies and randomized controlled trials have demonstrated the efficacy of therapy with high-dose intravenous immunoglobulin (IVIg) in MMN. However, this therapy has a short-lasting effect that needs to be maintained with periodic infusions. This can be partly overcome by the use of subcutaneous immunoglobulin (SCIg) at the same dose. The high cost and need for repeated infusions have led to the search for other immune therapies, the efficacy of which have not yet been confirmed in randomized trials. In addition, some therapies, including corticosteroids and plasma exchange, are not only ineffective but have been associated with clinical worsening. More recently, a number of novel therapies have been investigated in MMN, including interferon-β1a, the anti-CD20 monoclonal antibody rituximab and the complement inhibitor eculizumab. Preliminary data from open-label uncontrolled studies show that some patients improve after these therapies; however, randomized controlled trials are needed to confirm efficacy. Until then, IVIg (and SCIg) remains the mainstay of treatment in MMN, and the use of other immune therapies should only be considered for patients not responding to, or becoming resistant to, IVIg.
Collapse
Affiliation(s)
- Eduardo Nobile-Orazio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), 2nd Neurology, Humanitas Clinical and Research Center, IRCCS Humanitas Clinical Institute, Milan University, Via Manzoni 56, Rozzano, 20089 Milan, Italy.
| | | |
Collapse
|
16
|
Wang Y, Shaked I, Stanford SM, Zhou W, Curtsinger JM, Mikulski Z, Shaheen ZR, Cheng G, Sawatzke K, Campbell AM, Auger JL, Bilgic H, Shoyama FM, Schmeling DO, Balfour HH, Hasegawa K, Chan AC, Corbett JA, Binstadt BA, Mescher MF, Ley K, Bottini N, Peterson EJ. The autoimmunity-associated gene PTPN22 potentiates toll-like receptor-driven, type 1 interferon-dependent immunity. Immunity 2013; 39:111-22. [PMID: 23871208 DOI: 10.1016/j.immuni.2013.06.013] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 04/24/2013] [Indexed: 12/16/2022]
Abstract
Immune cells sense microbial products through Toll-like receptors (TLR), which trigger host defense responses including type 1 interferons (IFNs) secretion. A coding polymorphism in the protein tyrosine phosphatase nonreceptor type 22 (PTPN22) gene is a susceptibility allele for human autoimmune and infectious disease. We report that Ptpn22 selectively regulated type 1 IFN production after TLR engagement in myeloid cells. Ptpn22 promoted host antiviral responses and was critical for TLR agonist-induced, type 1 IFN-dependent suppression of inflammation in colitis and arthritis. PTPN22 directly associated with TNF receptor-associated factor 3 (TRAF3) and promotes TRAF3 lysine 63-linked ubiquitination. The disease-associated PTPN22W variant failed to promote TRAF3 ubiquitination, type 1 IFN upregulation, and type 1 IFN-dependent suppression of arthritis. The findings establish a candidate innate immune mechanism of action for a human autoimmunity "risk" gene in the regulation of host defense and inflammation.
Collapse
Affiliation(s)
- Yaya Wang
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Depaz R, Aboab J, Gout O. [Update on diagnosis and treatment of multiple sclerosis]. Rev Med Interne 2013; 34:628-35. [PMID: 23764421 DOI: 10.1016/j.revmed.2013.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 02/07/2013] [Indexed: 10/26/2022]
Abstract
Recent advances in neuroimaging have simplified the diagnostic criteria of multiple sclerosis. Indeed, the diagnosis of multiple sclerosis could be obtained during the first bout of disease flare, very early in the disease course. This is particularly important to shorten the diagnostic delay as early treatment may limit the occurrence of late irreversible disabilities. At the same time, major therapeutic advances have been obtained and new drugs that are well tolerated and more effective, despite the possible rare but potentially severe side effects are been developed. This article reviews the modern diagnostic and therapeutic strategies in multiple sclerosis in accordance with the recent obtained advances.
Collapse
Affiliation(s)
- R Depaz
- Fondation ophtalmologique Adolphe-de-Rothschild, 25-29, rue Manin, 75019 Paris, France.
| | | | | |
Collapse
|
18
|
Hartung HP, Haas J, Meergans M, Tracik F, Ortler S. [Interferon-β1b in multiple sclerosis therapy: more than 20 years clinical experience]. DER NERVENARZT 2013; 84:679-704. [PMID: 23669866 DOI: 10.1007/s00115-013-3781-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The introduction of interferon-β1b in 1993 in the USA and 2 years later in Europe made it possible for the first time to alter the course of the disease in patients with relapsing-remitting multiple sclerosis (MS). Subsequently, interferon-β1b was approved for the treatment of patients with active secondary progressive MS (1999) and early relapsing-remitting MS following a first demyelinating event (clinically isolated syndrome, CIS) (2006). Here we provide an overview of the clinical experience gathered during more than 20 years of interferon-β use focusing on long-term efficacy and safety and the impact of early initiation of treatment. Furthermore, the following aspects will be discussed: putative mechanisms of action of interferon-β, indications for a disease-modifying therapy, clinical relevance of neutralizing antibodies, importance of adherence in MS therapy, high versus low frequency therapy, combination therapies with interferon-β and safety of interferon-β in children and adolescents with MS and during pregnancy.
Collapse
Affiliation(s)
- H-P Hartung
- Neurologische Klinik im UKD, Medizinische Fakultät, Heinrich-Heine Universität, Moorenstr. 5, 40225 Düsseldorf, Deutschland.
| | | | | | | | | |
Collapse
|
19
|
Münzel EJ, Wimperis JZ, Williams A. Relapsing-remitting multiple sclerosis and chronic idiopathic neutropenia: a challenging combination. BMJ Case Rep 2013; 2013:bcr-2012-007936. [PMID: 23417376 DOI: 10.1136/bcr-2012-007936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We report the challenges of treating relapsing-remitting multiple sclerosis (MS) in a 31-year-old woman with long-standing chronic idiopathic neutropenia. The treatment with the disease-modifying therapy interferon-β was significantly complicated by a further fall in her generally low neutrophil count, to values below 0.5×10(9)/l, although this recovered rapidly when the treatment was stopped. We discuss the difficulties of balancing the risk of neutropenia with a risk of MS relapse.
Collapse
Affiliation(s)
- Eva Jolanda Münzel
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
20
|
Cox MB, Bowden NA, Scott RJ, Lechner-Scott J. Altered expression of the plasminogen activation pathway in peripheral blood mononuclear cells in multiple sclerosis: possible pathomechanism of matrix metalloproteinase activation. Mult Scler 2013; 19:1268-74. [PMID: 23401127 DOI: 10.1177/1352458513475493] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune disorder where a breakdown in the integrity of the blood-brain barrier is thought to allow lymphocytes to enter the central nervous system. OBJECTIVES The purpose of this study was to examine gene expression profiles between MS patients and healthy controls to identify genes intimately involved in the pathobiology of MS. METHODS Whole-genome gene expression analysis was performed using peripheral blood mononuclear cells from 39 healthy controls and 37 MS patients, 24 MS patients receiving no disease modifying therapy and 13 MS patients receiving interferon-beta (IFN-beta). Pathway analysis was performed to identify pathways dysregulated in MS. RESULTS Gene expression profiling of MS identified a signature of predominately immune associated genes. The plasminogen activation pathway contained an over-representation of significantly differentially expressed genes, including matrix metallopeptidase 9 (MMP9). Treatment with IFN-beta ameliorated the over-expression of MMP9, however the expression of two genes, plasminogen activator urokinase (PLAU) and serpin peptidase inhibitor, clade B (ovalbumin), member 2 (SERPINB2), forming part of the plasminogen activation pathway were not affected by IFN-beta therapy. CONCLUSIONS High expression levels of MMP9 have been associated with MS and the breakdown of the blood-brain barrier, while IFN-beta therapy decreases MMP9 expression. We confirm altered MMP9 expression in MS, and identify dysregulation within the plasminogen activation cascade, a pathway involved in the activation of MMP9.
Collapse
|