1
|
Meta-Analysis of Two Human RNA-seq Datasets to Determine Periodontitis Diagnostic Biomarkers and Drug Target Candidates. Int J Mol Sci 2022; 23:ijms23105580. [PMID: 35628390 PMCID: PMC9145972 DOI: 10.3390/ijms23105580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 02/01/2023] Open
Abstract
Periodontitis is a chronic inflammatory oral disease that affects approximately 42% of adults 30 years of age or older in the United States. In response to microbial dysbiosis within the periodontal pockets surrounding teeth, the host immune system generates an inflammatory environment in which soft tissue and alveolar bone destruction occur. The objective of this study was to identify diagnostic biomarkers and the mechanistic drivers of inflammation in periodontitis to identify drugs that may be repurposed to treat chronic inflammation. A meta-analysis comprised of two independent RNA-seq datasets was performed. RNA-seq analysis, signal pathway impact analysis, protein-protein interaction analysis, and drug target analysis were performed to identify the critical pathways and key players that initiate inflammation in periodontitis as well as to predict potential drug targets. Seventy-eight differentially expressed genes, 10 significantly impacted signaling pathways, and 10 hub proteins in periodontal gingival tissue were identified. The top 10 drugs that may be repurposed for treating periodontitis were then predicted from the gene expression and pathway data. The efficacy of these drugs in treating periodontitis has yet to be investigated. However, this analysis indicates that these drugs may serve as potential therapeutics to treat inflammation in gingival tissue affected by periodontitis.
Collapse
|
2
|
DiSano KD, Gilli F, Pachner AR. Memory B Cells in Multiple Sclerosis: Emerging Players in Disease Pathogenesis. Front Immunol 2021; 12:676686. [PMID: 34168647 PMCID: PMC8217754 DOI: 10.3389/fimmu.2021.676686] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/11/2021] [Indexed: 11/25/2022] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Once thought to be primarily driven by T cells, B cells are emerging as central players in MS immunopathogenesis. Interest in multiple B cell phenotypes in MS expanded following the efficacy of B cell-depleting agents targeting CD20 in relapsing-remitting MS and inflammatory primary progressive MS patients. Interestingly, these therapies primarily target non-antibody secreting cells. Emerging studies seek to explore B cell functions beyond antibody-mediated roles, including cytokine production, antigen presentation, and ectopic follicle-like aggregate formation. Importantly, memory B cells (Bmem) are rising as a key B cell phenotype to investigate in MS due to their antigen-experience, increased lifespan, and rapid response to stimulation. Bmem display diverse effector functions including cytokine production, antigen presentation, and serving as antigen-experienced precursors to antibody-secreting cells. In this review, we explore the cellular and molecular processes involved in Bmem development, Bmem phenotypes, and effector functions. We then examine how these concepts may be applied to the potential role(s) of Bmem in MS pathogenesis. We investigate Bmem both within the periphery and inside the CNS compartment, focusing on Bmem phenotypes and proposed functions in MS and its animal models. Finally, we review how current immunomodulatory therapies, including B cell-directed therapies and other immunomodulatory therapies, modify Bmem and how this knowledge may be harnessed to direct therapeutic strategies in MS.
Collapse
Affiliation(s)
- Krista D. DiSano
- Department of Neurology, Geisel School of Medicine & Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | | | | |
Collapse
|
3
|
Atorvastatin ameliorates viral burden and neural stem/progenitor cell (NSPC) death in an experimental model of Japanese encephalitis. J Biosci 2020. [DOI: 10.1007/s12038-020-00052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
4
|
Emamnejad R, Sahraian M, Shakiba Y, Salehi Z, Masoomi A, Imani D, Najafi F, Laribi B, Shirzad H, Izad M. Circulating mesenchymal stem cells, stromal derived factor (SDF)-1 and IP-10 levels increased in clinically active multiple sclerosis patients but not in clinically stable patients treated with beta interferon. Mult Scler Relat Disord 2019; 35:233-238. [DOI: 10.1016/j.msard.2019.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 06/27/2019] [Accepted: 08/11/2019] [Indexed: 12/19/2022]
|
5
|
Cost of disease modifying therapies for multiple sclerosis: Is front-loading the answer? J Neurol Sci 2019; 404:19-28. [DOI: 10.1016/j.jns.2019.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 01/10/2023]
|
6
|
Wilbanks B, Maher LJ, Rodriguez M. Glial cells as therapeutic targets in progressive multiple sclerosis. Expert Rev Neurother 2019; 19:481-494. [PMID: 31081705 DOI: 10.1080/14737175.2019.1614443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Multiple sclerosis is a serious demyelinating disease of the central nervous system (CNS) with treatments generally restricted to immunosuppression to reduce attack rate and for symptom management. Glial cells may be useful targets for future CNS regenerative therapies to reverse disease. Areas covered: In this review, the authors cover currently available multiple sclerosis treatments and examine potential upcoming therapies targeting glial cells. The potential for new therapeutic approaches in the treatment of progressive multiple sclerosis is examined. Expert opinion: Microglia, astrocytes, and oligodendrocytes are each promising targets for the disease-altering treatment of multiple sclerosis. Though challenging, the opportunities presented have great potential for CNS regeneration and further investigation of glial cells in therapy is warranted. Patient-specific combinatorial therapy targeting the three glial cell types is expected to be the future of MS treatment.
Collapse
Affiliation(s)
- Brandon Wilbanks
- a Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - L J Maher
- a Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Moses Rodriguez
- b Departments of Neurology and Immunology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
7
|
Abstract
B cells play a vital function in multiple sclerosis (MS) pathogenesis through an array of effector functions. All currently approved MS disease-modifying therapies alter the frequency, phenotype, or homing of B cells in one way or another. The importance of this mechanism of action has been reinforced with the successful development and clinical testing of B-cell-depleting monoclonal antibodies that target the CD20 surface antigen. Ocrelizumab, a humanized anti-CD20 monoclonal antibody, was approved by the Food and Drug Administration (FDA) in March 2017 after pivotal trials showed dramatic reductions in inflammatory disease activity in relapsing MS as well as lessening of disability progression in primary progressive MS. These and other clinical studies place B cells at the center of the inflammatory cascade in MS and provide a launching point for development of therapies that target selective pathogenic B-cell populations.
Collapse
Affiliation(s)
- Joseph J Sabatino
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| | - Scott S Zamvil
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| | - Stephen L Hauser
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| |
Collapse
|
8
|
Napier J, Rose L, Adeoye O, Hooker E, Walsh KB. Modulating acute neuroinflammation in intracerebral hemorrhage: the potential promise of currently approved medications for multiple sclerosis. Immunopharmacol Immunotoxicol 2019; 41:7-15. [PMID: 30702002 DOI: 10.1080/08923973.2019.1566361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The secondary inflammatory injury following intracerebral hemorrhage (ICH) results in increased morbidity and mortality. White blood cells have been implicated as critical mediators of this inflammatory injury. Currently, no medications have been clinically proven to ameliorate or beneficially modulate inflammation, or to improve outcomes by any mechanism, following ICH. However, other neuroinflammatory conditions, such as multiple sclerosis, have approved pharmacologic therapies that modulate the inflammatory response and minimize the damage caused by inflammatory cells. Thus, there is substantial interest in existing therapies for neuroinflammation and their potential applicability to other acute neurological diseases such as ICH. In this review, we examined the mechanism of action of twelve currently approved medications for multiple sclerosis: alemtuzumab, daclizumab, dimethyl fumarate, fingolimod, glatiramer acetate, interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, ocrelizumab, rituximab, teriflunomide. We analyzed the existing literature pertaining to the effects of these medications on various leukocytes and also with emphasis on mechanisms of action during the acute period following initiation of therapy. As a result, we provide a valuable summary of the current body of knowledge regarding these therapies and evidence that supports or refutes their likely promise for treating neuroinflammation following ICH.
Collapse
Affiliation(s)
- Jarred Napier
- a College of Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Lucas Rose
- a College of Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Opeolu Adeoye
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA.,c Gardner Neuroscience Institute , University of Cincinnati , Cincinnati , OH , USA
| | - Edmond Hooker
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Kyle B Walsh
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA.,c Gardner Neuroscience Institute , University of Cincinnati , Cincinnati , OH , USA
| |
Collapse
|
9
|
Furber KL, Van Agten M, Evans C, Haddadi A, Doucette JR, Nazarali AJ. Advances in the treatment of relapsing-remitting multiple sclerosis: the role of pegylated interferon β-1a. Degener Neurol Neuromuscul Dis 2017; 7:47-60. [PMID: 30050377 PMCID: PMC6053102 DOI: 10.2147/dnnd.s71986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a progressive, neurodegenerative disease with unpredictable phases of relapse and remission. The cause of MS is unknown, but the pathology is characterized by infiltration of auto-reactive immune cells into the central nervous system (CNS) resulting in widespread neuroinflammation and neurodegeneration. Immunomodulatory-based therapies emerged in the 1990s and have been a cornerstone of disease management ever since. Interferon β (IFNβ) was the first biologic approved after demonstrating decreased relapse rates, disease activity and progression of disability in clinical trials. However, frequent dosing schedules have limited patient acceptance for long-term therapy. Pegylation, the process by which molecules of polyethylene glycol are covalently linked to a compound, has been utilized to increase the half-life of IFNβ and decrease the frequency of administration required. To date, there has been one clinical trial evaluating the efficacy of pegylated IFN. The purpose of this article is to provide an overview of the role of IFN in the treatment of MS and evaluate the available evidence for pegylated IFN therapy in MS.
Collapse
Affiliation(s)
- Kendra L Furber
- Laboratory of Molecular Cell Biology, .,College of Pharmacy and Nutrition, .,Neuroscience Research Cluster, University of Saskatchewan,
| | - Marina Van Agten
- Laboratory of Molecular Cell Biology, .,College of Pharmacy and Nutrition, .,Neuroscience Research Cluster, University of Saskatchewan,
| | - Charity Evans
- College of Pharmacy and Nutrition, .,Cameco Multiple Sclerosis Neuroscience Research Center, City Hospital,
| | | | - J Ronald Doucette
- Neuroscience Research Cluster, University of Saskatchewan, .,Cameco Multiple Sclerosis Neuroscience Research Center, City Hospital, .,Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Adil J Nazarali
- Laboratory of Molecular Cell Biology, .,College of Pharmacy and Nutrition, .,Neuroscience Research Cluster, University of Saskatchewan, .,Cameco Multiple Sclerosis Neuroscience Research Center, City Hospital,
| |
Collapse
|
10
|
Buzzard K, Chan WH, Kilpatrick T, Murray S. Multiple Sclerosis: Basic and Clinical. ADVANCES IN NEUROBIOLOGY 2017; 15:211-252. [DOI: 10.1007/978-3-319-57193-5_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11
|
Cohan S. Therapeutic efficacy of monthly subcutaneous injection of daclizumab in relapsing multiple sclerosis. Biologics 2016; 10:119-38. [PMID: 27672308 PMCID: PMC5026217 DOI: 10.2147/btt.s89218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Despite the availability of multiple disease-modifying therapies for relapsing multiple sclerosis (MS), there remains a need for highly efficacious targeted therapy with a favorable benefit-risk profile and attributes that encourage a high level of treatment adherence. Daclizumab is a humanized monoclonal antibody directed against CD25, the α subunit of the high-affinity interleukin 2 (IL-2) receptor, that reversibly modulates IL-2 signaling. Daclizumab treatment leads to antagonism of proinflammatory, activated T lymphocyte function and expansion of immunoregulatory CD56(bright) natural killer cells, and has the potential to, at least in part, rectify the imbalance between immune tolerance and autoimmunity in relapsing MS. The clinical pharmacology, efficacy, and safety of subcutaneous daclizumab have been evaluated extensively in a large clinical study program. In pivotal studies, daclizumab demonstrated superior efficacy in reducing clinical and radiologic measures of MS disease activity compared with placebo or intramuscular interferon beta-1a, a standard-of-care therapy for relapsing MS. The risk of hepatic disorders, cutaneous events, and infections was modestly increased. The monthly subcutaneous self-injection dosing regimen of daclizumab may be advantageous in maintaining patient adherence to treatment, which is important for optimal outcomes with MS disease-modifying therapy. Daclizumab has been approved in the US and in the European Union and represents an effective new treatment option for patients with relapsing forms of MS, and is currently under review by other regulatory agencies.
Collapse
Affiliation(s)
- Stanley Cohan
- Providence Multiple Sclerosis Center
- Providence Brain and Spine Institute
- Providence Health & Services, Portland, OR, USA
| |
Collapse
|
12
|
Bittner S, Ruck T, Wiendl H, Grauer OM, Meuth SG. Targeting B cells in relapsing-remitting multiple sclerosis: from pathophysiology to optimal clinical management. Ther Adv Neurol Disord 2016; 10:51-66. [PMID: 28450895 DOI: 10.1177/1756285616666741] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease that is caused by an autoimmune response against central nervous system (CNS) structures. Traditionally considered a T-cell-mediated disorder, the contribution of B cells to the pathogenesis of MS has long been debated. Based on recent promising clinical results from CD20-depleting strategies by three therapeutic monoclonal antibodies in clinical phase II and III trials (rituximab, ocrelizumab and ofatumumab), targeting B cells in MS is currently attracting growing interest among basic researchers and clinicians. Many questions about the role of B and plasma cells in MS remain still unanswered, ranging from the role of specific B-cell subsets and functions to the optimal treatment regimen of B-cell depletion and monitoring thereafter. Here, we will assess our current knowledge of the mechanisms implicating B cells in multiple steps of disease pathology and examine current and future therapeutic approaches for the treatment of MS.
Collapse
Affiliation(s)
- Stefan Bittner
- Department of Neurology, University of Mainz, Mainz, Germany
| | - Tobias Ruck
- Department of Neurology, University of Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Oliver M Grauer
- Department of Neurology, University of Münster, Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Münster, Germany
| |
Collapse
|
13
|
Simpson S, Stewart N, van der Mei I, Blizzard L, Taylor BV. Synergetic and antagonistic effects of combined calcitriol and interferon-β treatment on cytokine production by stimulated PBMCs. J Neuroimmunol 2016; 297:148-55. [PMID: 27397088 DOI: 10.1016/j.jneuroim.2016.05.020] [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: 01/08/2015] [Revised: 04/04/2016] [Accepted: 05/25/2016] [Indexed: 11/26/2022]
Abstract
BACKGROUND We evaluated the effects of calcitriol and interferon-β on in vitro PBMC cytokine production from a cohort of 22 healthy adults not on medication. METHODS PBMCs were incubated with calcitriol and/or 100 or 400IU interferon-β or nothing, followed by stimulation with concanavalin A. RESULTS When combined, calcitriol and interferon-β appeared to potentiate the effects of one another on reducing IL-6. Calcitriol significantly reduced the production of IL-2, IL-4, IL-6, and IFN-γ, while interferon-β significantly reduced production of IL-6 and TNF-α, and increased IL-10. DISCUSSION This is the first study to evaluate the effects of combined calcitriol and interferon-β on cytokine production in PBMCs in vitro, demonstrating novel synergetic effects.
Collapse
Affiliation(s)
- Steve Simpson
- Menzies Institute for Medical Research, University of Tasmania, Australia.
| | - Niall Stewart
- School of Medicine, University of Tasmania, Australia; School of Pharmacy, University of Tasmania, Australia
| | - Ingrid van der Mei
- Menzies Institute for Medical Research, University of Tasmania, Australia
| | - Leigh Blizzard
- Menzies Institute for Medical Research, University of Tasmania, Australia
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Australia
| |
Collapse
|
14
|
Abstract
Discussions of multiple sclerosis (MS) pathophysiology tend to focus on T cells and B cells of the adaptive immune response. The innate immune system is less commonly considered in this context, although dendritic cells, monocytes, macrophages and microglia - collectively referred to as myeloid cells - have prominent roles in MS pathogenesis. These populations of myeloid cells function as antigen-presenting cells and effector cells in neuroinflammation. Furthermore, a vicious cycle of interactions between T cells and myeloid cells exacerbates pathology. Several disease-modifying therapies are now available to treat MS, and insights into their mechanisms of action have largely focused on the adaptive immune system, but these therapies also have important effects on myeloid cells. In this Review, we discuss the evidence for the roles of myeloid cells in MS and the experimental autoimmune encephalomyelitis model of MS, and consider how interactions between myeloid cells and T cells and/or B cells promote MS pathology. Finally, we discuss the direct and indirect effects of existing MS medications on myeloid cells.
Collapse
Affiliation(s)
- Manoj K Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
15
|
Kavrochorianou N, Markogiannaki M, Haralambous S. IFN-β differentially regulates the function of T cell subsets in MS and EAE. Cytokine Growth Factor Rev 2016; 30:47-54. [DOI: 10.1016/j.cytogfr.2016.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/21/2016] [Indexed: 12/30/2022]
|
16
|
Espejo C, Brieva L, Ruggiero G, Río J, Montalban X, Martínez-Cáceres EM. IFN-β treatment modulates the CD28/CTLA-4-mediated pathway for IL-2 production in patients with relapsing -remitting multiple sclerosis. Mult Scler 2016; 10:630-5. [PMID: 15584487 DOI: 10.1191/1352458504ms1094oa] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system probably mediated by Th1 lymphocytes. IFN-b is an established therapy for relapsing MS patients, although the mechanisms underlying its efficacy are yet to be well characterized. We determined IL-2 production, CD25 expression and T-cell proliferation from relapsing -remitting MS patients before and three months after starting therapy. A decrease in the percentage of CD80-induced IL-2-producing cells was observed after in vivo IFN-b treatment. These data support that one of the immunomodulatory effects of IFN-b treatment in MS may be a limitation of the autoimmune response modifying the CD80:CD28/CTLA-4 pathway.
Collapse
Affiliation(s)
- C Espejo
- Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
17
|
Yin H, Zhou H, Kang Y, Zhang X, Duan X, Alnabhan R, Liang S, Scott DA, Lamont RJ, Shang J, Wang H. Syk negatively regulates TLR4-mediated IFNβ and IL-10 production and promotes inflammatory responses in dendritic cells. Biochim Biophys Acta Gen Subj 2015; 1860:588-98. [PMID: 26708990 DOI: 10.1016/j.bbagen.2015.12.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND While Syk has been shown to associate with TLR4, the immune consequences of Syk-TLR interactions and related molecular mechanisms are unclear. METHODS Gain- and loss-of-function approaches were utilized to determine the regulatory function of Syk and elucidate the related molecular mechanisms in TLR4-mediated inflammatory responses. Cytokine production was measured by ELISA and phosphorylation of signaling molecules determined by Western blotting. RESULTS Syk deficiency in murine dendritic cells resulted in the enhancement of LPS-induced IFNβ and IL-10 but suppression of pro-inflammatory cytokines (TNFα, IL-6). Deficiency of Syk enhanced the activity of PI3K and elevated the phosphorylation of PI3K and Akt, which in turn, lead to the phospho-inactivation of the downstream, central gatekeeper of the innate response, GSK3β. Inhibition of PI3K or Akt abrogated the ability of Syk deficiency to enhance IFNβ and IL-10 in Syk deficient cells, confirmed by the overexpression of Akt (Myr-Akt) or constitutively active GSK3β (GSK3 S9A). Moreover, neither inhibition of PI3K-Akt signaling nor neutralization of de novo synthesized IFNβ could rescue TNFα and IL-6 production in LPS-stimulated Syk deficient cells. Syk deficiency resulted in decreased phosphorylation of IKKβ and the NF-κB p65 subunit, further suggesting a divergent influence of Syk on pro- and anti-inflammatory TLR responses. CONCLUSIONS Syk negatively regulates TLR4-mediated production of IFNβ and IL-10 and promotes inflammatory responses in dendritic cells through divergent regulation of downstream PI3K-Akt and NF-κB signaling pathways. GENERAL SIGNIFICANCE Syk may represent a novel target for manipulating the direction or intensity of the innate response, depending on clinical necessity.
Collapse
Affiliation(s)
- Hui Yin
- Department of Infectious Diseases, Henan Provincial People's Hospital (Zhengzhou University People's Hospital), Zhengzhou, Henan 450001, China
| | - Huaxin Zhou
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Yi Kang
- Department of Infectious Diseases, Henan Provincial People's Hospital (Zhengzhou University People's Hospital), Zhengzhou, Henan 450001, China
| | - Xiaoju Zhang
- Department of Infectious Diseases, Henan Provincial People's Hospital (Zhengzhou University People's Hospital), Zhengzhou, Henan 450001, China
| | - Xiaoxian Duan
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Ridab Alnabhan
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Shuang Liang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA
| | - Jia Shang
- Department of Infectious Diseases, Henan Provincial People's Hospital (Zhengzhou University People's Hospital), Zhengzhou, Henan 450001, China.
| | - Huizhi Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY 40202, USA.
| |
Collapse
|
18
|
Boivin N, Baillargeon J, Doss PMIA, Roy AP, Rangachari M. Interferon-β suppresses murine Th1 cell function in the absence of antigen-presenting cells. PLoS One 2015; 10:e0124802. [PMID: 25885435 PMCID: PMC4401451 DOI: 10.1371/journal.pone.0124802] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Interferon (IFN)-β is a front-line therapy for the treatment of the relapsing-remitting form of multiple sclerosis. However, its immunosuppressive mechanism of function remains incompletely understood. While it has been proposed that IFN-β suppresses the function of inflammatory myelin antigen-reactive T cells by promoting the release of immunomodulatory cytokines such as IL-27 from antigen-presenting cells (APCs), its direct effects on inflammatory CD4+ Th1 cells are less clear. Here, we establish that IFN-β inhibits mouse IFN-γ+ Th1 cell function in the absence of APCs. CD4+ T cells express the type I interferon receptor, and IFN-β can suppress Th1 cell proliferation under APC-free stimulation conditions. IFN-β-treated myelin antigen-specific Th1 cells are impaired in their ability to induce severe experimental autoimmune encephalomyelitis (EAE) upon transfer to lymphocyte-deficient Rag1-/- mice. Polarized Th1 cells downregulate IFN-γ and IL-2, and upregulate the negative regulatory receptor Tim-3, when treated with IFN-β in the absence of APCs. Further, IFN-β treatment of Th1 cells upregulates phosphorylation of Stat1, and downregulates phosphorylation of Stat4. Our data indicate that IFN-γ-producing Th1 cells are directly responsive to IFN-β and point to a novel mechanism of IFN-β-mediated T cell suppression that is independent of APC-derived signals.
Collapse
Affiliation(s)
- Nicolas Boivin
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Joanie Baillargeon
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
| | - Prenitha Mercy Ignatius Arokia Doss
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Andrée-Pascale Roy
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Graduate Programme in Microbiology and Immunology, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
| | - Manu Rangachari
- Department of Neuroscience, Centre de recherche du CHU de Québec—Université Laval, Québec QC, Canada G1V 4G2
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec QC, Canada G1V 0A6
- * E-mail:
| |
Collapse
|
19
|
|
20
|
Lundström W, Hermanrud C, Sjöstrand M, Brauner S, Wahren-Herlenius M, Olsson T, Karrenbauer V, Hillert J, Fogdell-Hahn A. Interferon beta treatment of multiple sclerosis increases serum interleukin-7. Mult Scler 2014; 20:1727-36. [PMID: 24821684 DOI: 10.1177/1352458514532700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Interleukin-7 (IL-7) is a non-redundant cytokine for T-cell development and survival. The IL-7 signaling pathway has been genetically and functionally associated with several autoimmune diseases including multiple sclerosis (MS). OBJECTIVE The objective of this paper is to elucidate the effect of the widely used immunomodulatory MS therapy interferon beta (IFNβ) on IL-7 homeostasis. METHODS Swedish MS patients were screened for IL-7 concentration in serum and blood cell counts. IL-7 receptor alpha chain (IL-7Rα) expression was determined by semi-quantitative real-time polymerase chain reaction (PCR) and flow cytometry. RESULTS IFNβ treatment led to significantly increased serum IL-7 levels (mean: 17 pg/ml) compared with healthy controls (mean: 7.6 pg/ml) and natalizumab-treated patients (mean: 5.3 pg/ml). In vitro and in vivo, peripheral blood leukocytes showed decreased IL-7Rα expression and IL-7 consumption upon IFNβ exposure, suggesting that their IL-7 responsiveness is impaired during treatment. CONCLUSIONS MS patients undergoing IFNβ treatment have increased serum IL-7 levels and decreased IL-7 consumption. Given IL-7's important role in T-cell immunity, this relationship may be highly relevant for IFNβ's treatment efficacy.
Collapse
Affiliation(s)
- Wangko Lundström
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christina Hermanrud
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Sjöstrand
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Susanna Brauner
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | | | - Tomas Olsson
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Virginija Karrenbauer
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Fogdell-Hahn
- Center for Molecular Medicine, Karolinska Institutet, SE-17176 Stockholm, Sweden
| |
Collapse
|
21
|
Menezes SM, Decanine D, Brassat D, Khouri R, Schnitman SV, Kruschewsky R, López G, Alvarez C, Talledo M, Gotuzzo E, Vandamme AM, Galvão-Castro B, Liblau R, Weyenbergh JV. CD80+ and CD86+ B cells as biomarkers and possible therapeutic targets in HTLV-1 associated myelopathy/tropical spastic paraparesis and multiple sclerosis. J Neuroinflammation 2014; 11:18. [PMID: 24472094 PMCID: PMC3922160 DOI: 10.1186/1742-2094-11-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/13/2014] [Indexed: 01/14/2023] Open
Abstract
Background Human T-cell lymphotropic virus (HTLV-1) is the causative agent of the incapacitating, neuroinflammatory disease HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Currently, there are no disease-modifying therapies with long-term clinical benefits or validated biomarkers for clinical follow-up in HAM/TSP. Although CD80 and CD86 costimulatory molecules play prominent roles in immune regulation and reflect disease status in multiple sclerosis (MS), data in HAM/TSP are lacking. Methods Using flow cytometry, we quantified ex vivo and in vitro expression of CD80 and CD86 in PBMCs of healthy controls, HTLV-1-infected individuals with and without HAM/TSP, and MS patients. We hypothesized ex vivo CD80 and CD86 expressions and their in vitro regulation by interferon (IFN)-α/β mirror similarities between HAM/TSP and MS and hence might reveal clinically useful biomarkers in HAM/TSP. Results Ex vivo expression of CD80 and CD86 in T and B cells increased in all HTLV-1 infected individuals, but with a selective defect for B cell CD86 upregulation in HAM/TSP. Despite decreased total B cells with increasing disease duration (p = 0.0003, r = −0.72), CD80+ B cells positively correlated with disease severity (p = 0.0017, r = 0.69) in HAM/TSP. B cell CD80 expression was higher in women with HAM/TSP, underscoring that immune markers can reflect the female predominance observed in most autoimmune diseases. In contrast to MS patients, CD80+ (p = 0.0001) and CD86+ (p = 0.0054) lymphocytes expanded upon in vitro culture in HAM/TSP patients. The expansion of CD80+ and CD86+ T cells but not B cells was associated with increased proliferation in HTLV-1 infection. In vitro treatment with IFN-β but not IFN-α resulted in a pronounced increase of B cell CD86 expression in healthy controls, as well as in patients with neuroinflammatory disease (HAM/TSP and MS), similar to in vivo treatment in MS. Conclusions We propose two novel biomarkers, ex vivo CD80+ B cells positively correlating to disease severity and CD86+ B cells preferentially induced by IFN-β, which restores defective upregulation in HAM/TSP. This study suggests a role for B cells in HAM/TSP pathogenesis and opens avenues to B cell targeting (with proven clinical benefit in MS) in HAM/TSP but also CD80-directed immunotherapy, unprecedented in both HAM/TSP and MS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Johan Van Weyenbergh
- Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
22
|
Calder CJ, Duddy M, Bar-Or A. B-cell subsets: cellular interactions and relevance in multiple sclerosis. Expert Rev Clin Immunol 2014; 3:73-83. [DOI: 10.1586/1744666x.3.1.73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Buzzard KA, Broadley SA, Butzkueven H. What do effective treatments for multiple sclerosis tell us about the molecular mechanisms involved in pathogenesis? Int J Mol Sci 2012. [PMID: 23202920 PMCID: PMC3497294 DOI: 10.3390/ijms131012665] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Multiple sclerosis is a potentially debilitating disease of the central nervous system. A concerted program of research by many centers around the world has consistently demonstrated the importance of the immune system in its pathogenesis. This knowledge has led to the formal testing of a number of therapeutic agents in both animal models and humans. These clinical trials have shed yet further light on the pathogenesis of MS through their sometimes unexpected effects and by their differential effects in terms of impact on relapses, progression of the disease, paraclinical parameters (MRI) and the adverse events that are experienced. Here we review the currently approved medications for the commonest form of multiple sclerosis (relapsing-remitting) and the emerging therapies for which preliminary results from phase II/III clinical trials are available. A detailed analysis of the molecular mechanisms responsible for the efficacy of these medications in multiple sclerosis indicates that blockade or modulation of both T- and B-cell activation and migration pathways in the periphery or CNS can lead to amelioration of the disease. It is hoped that further therapeutic trials will better delineate the pathogenesis of MS, ultimately leading to even better treatments with fewer adverse effects.
Collapse
Affiliation(s)
- Katherine A. Buzzard
- Department of Neurology, Royal Melbourne Hospital, Royal Parade, Parkville VIC 3050, Australia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +61-3-8344-1802; Fax: +61-3-9348-1707
| | - Simon A. Broadley
- School of Medicine, Griffith University, Gold Coast Campus, QLD 4222, Australia; E-Mail:
- Department of Neurology, Gold Coast Hospital, 108 Nerang Street, Southport QLD 4215, Australia
| | - Helmut Butzkueven
- Melbourne Brain Centre at the Royal Melbourne Hospital, Department of Medicine, University of Melbourne, Royal Parade, Parkville VIC 3010, Australia; E-Mail:
| |
Collapse
|
24
|
Aung LL, Brooks A, Greenberg SA, Rosenberg ML, Dhib-Jalbut S, Balashov KE. Multiple sclerosis-linked and interferon-beta-regulated gene expression in plasmacytoid dendritic cells. J Neuroimmunol 2012; 250:99-105. [PMID: 22688425 DOI: 10.1016/j.jneuroim.2012.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/14/2012] [Accepted: 05/18/2012] [Indexed: 01/05/2023]
Abstract
The cause of multiple sclerosis (MS) is not known and the mechanism of interferon-beta, a disease-modifying treatment, is not well-understood. We studied gene expression in plasmacytoid dendritic cells (pDCs), antigen-presenting cells implicated in MS pathogenesis. PDCs were separated from healthy donors and MS patients at two time points: before and after initiation of treatment with interferon-beta. Expression of selected MS-linked and interferon-beta-regulated genes was validated with single assays. We have identified 60 genes which were abnormally expressed in MS patients and were corrected after treatment. These genes could be studied as potential MS biomarkers and possible therapeutic targets in MS.
Collapse
Affiliation(s)
- Latt Latt Aung
- Department of Neurology, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| | | | | | | | | | | |
Collapse
|
25
|
Ireland SJ, Blazek M, Harp CT, Greenberg B, Frohman EM, Davis LS, Monson NL. Antibody-independent B cell effector functions in relapsing remitting Multiple Sclerosis: Clues to increased inflammatory and reduced regulatory B cell capacity. Autoimmunity 2012; 45:400-14. [DOI: 10.3109/08916934.2012.665529] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Regulation of suppressors of cytokine signaling as a therapeutic approach in autoimmune diseases, with an emphasis on multiple sclerosis. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:635721. [PMID: 22132325 PMCID: PMC3206360 DOI: 10.1155/2011/635721] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 09/09/2011] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating, presumably autoimmune disease of the central nervous system (CNS). Among the available MS therapies, interferon (IFN)β and the recently introduced statins have been reported to exert their immunomodulatory effects through the induction of SOCS1 and SOCS3 in various inflammatory cell subsets. The SOCS proteins negatively regulate cytokine and Toll-like receptors- (TLR-) induced signaling in the inflammatory cells. SOCS1 and SOCS3 have been reported to play an important role in the regulation of Th17-cell differentiation through their effects on the cells of the innate and adaptive immune systems. IFNβ and statins inhibit Th17-cell differentiation directly and indirectly via induction of SOCS1 and SOCS3 expression in monocytes, dendritic cells (DCs), and B-cells. Due to their rapid induction and degradation, and SOCS-mediated regulation of multiple cytokine-signaling pathways, they represent an attractive therapeutic target in the autoimmune diseases, and particularly relapsing remitting (RR) MS.
Collapse
|
27
|
Abstract
Multiple sclerosis (MS) is characterized by autoimmune inflammation and subsequent neurodegeneration. It is believed that early in the disease course, proinflammatory T cells that are activated in the periphery by antigen presentation cross the blood-brain barrier (BBB) into the CNS directed by various chemotaxic agents. However, to date, there has been no formal demonstration of a specific precipitating antigen. Once inside the CNS, activated T cells including T helper-1 (T(h)1), T(h)17, γδ and CD8+ types are believed to secrete proinflammatory cytokines. Decreased levels of T(h)2 cells also correlate with relapses and disease progression in MS, since T(h)2-derived cytokines are predominantly anti-inflammatory. In healthy tissue, inflammatory effects are opposed by specific subsets of regulatory T cells (T(regs)) including CD4+, CD25+ and FoxP3+ cells that have the ability to downregulate the activity of proinflammatory T cells, allowing repair and recovery to generally follow inflammatory insult. Given their function, the pathogenesis of MS most likely involves deficits of T(reg) function, which allow autoimmune inflammation and resultant neurodegeneration to proceed relatively unchecked. Interferons (IFNs) are naturally occurring cytokines possessing a wide range of anti-inflammatory properties. Recombinant forms of IFNβ are widely used as first-line treatment in relapsing forms of MS. The mechanism of action of IFNβ is complex, involving effects at multiple levels of cellular function. IFNβ appears to directly increase expression and concentration of anti-inflammatory agents while downregulating the expression of proinflammatory cytokines. IFNβ treatment may reduce the trafficking of inflammatory cells across the BBB and increase nerve growth factor production, leading to a potential increase in neuronal survival and repair. IFNβ can also increase the number of CD56bright natural killer cells in the peripheral blood. These cells are efficient producers of anti-inflammatory mediators, and may have the ability to curb neuron inflammation. The mechanistic effects of IFNβ manifest clinically as reduced MRI lesion activity, reduced brain atrophy, increased time to reach clinically definite MS after the onset of neurological symptoms, decreased relapse rate and reduced risk of sustained disability progression. The mechanism of action of IFNβ in MS is multifactorial and incompletely understood. Ongoing and future studies will increase our understanding of the actions of IFNβ on the immune system and the CNS, which will in turn aid advances in the management of MS.
Collapse
Affiliation(s)
- Bernd C Kieseier
- Department of Neurology, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
28
|
Baker DP, Pepinsky RB, Brickelmaier M, Gronke RS, Hu X, Olivier K, Lerner M, Miller L, Crossman M, Nestorov I, Subramanyam M, Hitchman S, Glick G, Richman S, Liu S, Zhu Y, Panzara MA, Davar G. PEGylated interferon beta-1a: meeting an unmet medical need in the treatment of relapsing multiple sclerosis. J Interferon Cytokine Res 2011; 30:777-85. [PMID: 20836711 DOI: 10.1089/jir.2010.0092] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis is a chronic autoimmune disease of the central nervous system for which a number of disease-modifying therapies are available, including interferon beta (Avonex®, Rebif®, and Betaseron/Betaferon®), glatiramer acetate (Copaxone®), and an anti-VLA4 monoclonal antibody (Tysabri®). Despite the availability and efficacy of these protein and peptide drugs, there remains a significant number of patients who are untreated, including those with relatively mild disease who choose not to initiate therapy, those wary of injections or potential adverse events associated with therapy, and those who have stopped therapy due to perceived lack of efficacy. Since these drugs have side effects that may affect a patient's decision to initiate and to remain on treatment, there is a need to provide a therapy that is safe and efficacious but that requires a reduced dosing frequency and hence a concomitant reduction in the frequency of side effects. Here we describe the development of a PEGylated form of interferon beta-1a that is currently being tested in a multicenter, randomized, double-blind, parallel-group, placebo-controlled study in relapsing multiple sclerosis patients, with the aim of determining the safety and efficacy of 125 microg administered via the subcutaneous route every 2 or 4 weeks.
Collapse
|
29
|
Ramgolam VS, Sha Y, Marcus KL, Choudhary N, Troiani L, Chopra M, Markovic-Plese S. B Cells as a Therapeutic Target for IFN-β in Relapsing–Remitting Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2011; 186:4518-26. [DOI: 10.4049/jimmunol.1000271] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
30
|
Boster A, Ankeny DP, Racke MK. The Potential Role of B Cell-Targeted Therapies in Multiple Sclerosis. Drugs 2010; 70:2343-2356. [DOI: 10.2165/11585230-000000000-00000] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
31
|
Boyko AN. Clinical effects and tolerability of high-dose, high-frequency recombinant interferon beta-1a in patients with multiple sclerosis: maximizing therapy through long-term adherence. Expert Opin Biol Ther 2010; 10:653-66. [PMID: 20218924 DOI: 10.1517/14712591003702361] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD High-dose, high-frequency IFN beta-1a in multiple sclerosis (MS) can prevent lesion formation, decrease the frequency/severity of relapses and delay progression of disability, with a proven safety profile. Rates of non-adherence are high. There are drugs under investigation that may have greater efficacy and different safety profiles from existing therapies. AREAS COVERED IN THIS REVIEW Evidence supporting the efficacy of IFN beta-1a, factors contributing to non-adherence, and strategies to combat non-adherence. It is hoped that these strategies, coupled with future advances in pharmacogenetics, might lead to better outcomes. The PubMed database was searched using the terms "multiple sclerosis" and "interferon beta-1a", for papers published between 1998 and 2010. Relevant manuscripts and pivotal papers from clinical trials were cited. Searches of abstracts from congresses were also performed to obtain recent findings. WHAT THE READER WILL GAIN An overview of early pivotal trials, comparative studies with other treatments, and recent studies assessing the development of this therapy. TAKE HOME MESSAGE Long-term treatment with IFN beta-1a has benefits in MS and a good safety profile. Although adherence outside of clinical trials can be poor, injection devices, better tolerated drug formulations and education regarding treatment expectations are some of the strategies employed to help patients to adhere to treatment in the hope of improving outcomes.
Collapse
Affiliation(s)
- Alexey N Boyko
- Russian State Medical University, City Hospital #11, Department of Neurology and Neurosurgery, Dvitsev 6, 127018 Moscow, Russia.
| |
Collapse
|
32
|
Mix E, Stefan K, Höppner J, Klauer T, Zettl UK, Strauss U, Meyer-Rienecker HJ, Rolfs A. Lymphocyte Subpopulations, Oxidative Burst and Apoptosis in Peripheral Blood Cells of Patients with Multiple Sclerosis–Effect of Interferon-β. Autoimmunity 2009; 36:291-305. [PMID: 14567559 DOI: 10.1080/0891693031000152697] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
At present, the most efficient therapeutical treatment of multiple sclerosis (MS) is achieved by IFN-beta. However, its in vivo effects remain incompletely understood. If applied parenterally, the hydrophobic IFN-beta acts primarily on blood cells with probable selectivity for functionally different lymphocyte subpopulations, monocytes and granulocytes. We have investigated the expression of the activation marker interleukin-2 receptor-alpha (CD25) on CD3+ T cells, CD19+ B cells, foetal-type gamma(delta)+CD3+ T cells and foetal-type CD5+CD19+ B cells of the peripheral blood. In addition, the oxidative burst activity and apoptosis have been determined in mononuclear and polymorphonuclear blood cells, respectively. The study accompanied a phase III trial with IFN-beta1b (BETAFERON, Schering). Two groups of MS patients with relapsing-remitting course of the disease have been investigated at 8 time points (days 0, 5, 15, 31, 60, 90, 180 and 270 after starting therapy): (1) verum group (n = 8) with application of 8 Mill. units IFN-beta1 b every other day, and (2) placebo group (n = 4) with application of placebo for 3 months and therapy as in (1) from day 90 onward. The main results were: (1) Activated T cells decreased until day 180 in the verum group and return thereafter to pre-treatment values, whereas in the placebo group the values remained relatively stable over the whole observation period. (2) Activated B cells increased between days 90 and 270 in both groups, i.e. after verum application in both groups. (3) Foetal-type B cells were more activated than total B and T cells with increase over time in both groups. (4) Foetal-type T cells exerted relatively stable intra-individual levels with generally low CD25 expression, but punctual CD25 peaks in both groups. (5) The spontaneous oxidative burst was higher in lymphocytes, more variable in monocytes and faster increasing in granulocytes in the verum group than in the placebo group. (6) Apoptosis of mononuclear cells and granulocytes showed similar variations in the verum and placebo groups with the exception of a selective increase over time of the proportion of granulocytes undergoing induced apoptosis in the verum group. It is concluded that IFN-beta has the following main effects on the immune system of MS patients: (1) the T cell immunity is systemically and reversibly suppressed, (2) the foetal-type lymphocytes, which are responsible for the first line of defence of infections, are stimulated in the long range, (3) the oxidative burst activity is increased in lymphocytes and granulocytes and instable in monocytes, and (4) the inducibility of apoptosis in granulocytes is increased. Re-examination of the altered blood cell parameters after long-term IFN-beta therapy is warranted.
Collapse
Affiliation(s)
- Eilhard Mix
- Department of Neurology, University of Rostock, Gehlsheimer Str. 20, P.O. Box 100888, 18055 Rostock, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Interferon-β therapy up-regulates BDNF secretion from PBMCs of MS patients through a CD40-dependent mechanism. J Neuroimmunol 2009; 211:114-9. [DOI: 10.1016/j.jneuroim.2009.04.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Revised: 04/04/2009] [Accepted: 04/08/2009] [Indexed: 01/10/2023]
|
34
|
Hartung HP. High-dose, high-frequency recombinant interferon beta-1a in the treatment of multiple sclerosis. Expert Opin Pharmacother 2009; 10:291-309. [PMID: 19236200 DOI: 10.1517/14656560802677882] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND There is at present no cure for multiple sclerosis (MS), and existing therapies are designed primarily to prevent lesion formation, decrease the rate and severity of relapses and delay the resulting disability by reducing levels of inflammation. OBJECTIVE The aim of this review was to assess the treatment of relapsing MS with particular focus on subcutaneous (s.c.) interferon (IFN) beta-1a. METHOD The literature on IFN beta-1a therapy of MS was reviewed based on a PubMed search (English-language publications from 1990) including its pharmacodynamics and pharmacokinetics, clinical efficacy in relapsing MS as shown in placebo-controlled studies and in comparative trials, efficacy in secondary progressive MS, safety and tolerability, and the impact of neutralizing antibodies. CONCLUSION The literature suggests that high-dose, high-frequency s.c. IFN beta-1a offers an effective option for treating patients with relapsing MS, with proven long-term safety and tolerability, and has a favourable benefit-to-risk ratio compared with other forms of IFN beta.
Collapse
Affiliation(s)
- Hans-Peter Hartung
- Heinrich-Heine-University, Department of Neurology, Moorenstreet 5, D-40225 Düsseldorf, Germany.
| |
Collapse
|
35
|
Martinez-Forero I, Pelaez A, Villoslada P. Pharmacogenomics of multiple sclerosis: in search for a personalized therapy. Expert Opin Pharmacother 2008; 9:3053-67. [DOI: 10.1517/14656560802515553] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
36
|
Abstract
Multiple sclerosis is an inflammatory demyelinating disease of the central nervous system with no clear etiology. Until recently, most studies have emphasized the role of T cells in the pathogenesis of multiple sclerosis. Data suggesting that B cells play a role in the pathogenesis of multiple sclerosis have been accumulating for the past five decades, demonstrating that the cerebrospinal fluid and central nervous system tissues of multiple sclerosis patients contain B cells, plasma cells, antibodies, and immunoglobulins. Data suggest that B cells are involved in antigen capture and presentation to T cells, cytokine production, antibody secretion, demyelination, tissue damage, and remyelination in multiple sclerosis. These advances in the understanding of B-cell and antibody roles in the pathophysiology of multiple sclerosis provide a strong rationale for B-cell-targeted therapies.
Collapse
|
37
|
Miyazaki Y, Iwabuchi K, Kikuchi S, Fukazawa T, Niino M, Hirotani M, Sasaki H, Onoé K. Expansion of CD4+CD28- T cells producing high levels of interferon-{gamma} in peripheral blood of patients with multiple sclerosis. Mult Scler 2008; 14:1044-55. [PMID: 18573819 DOI: 10.1177/1352458508092809] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD4(+) T cells that lack surface expression of the CD28 co-stimulatory molecule (CD4(+)CD28(-) T cells) were expanded in peripheral blood of patients with multiple sclerosis (MS) [5.20 +/- 1.67% vs 13.00 +/- 2.68% (healthy controls (HC) versus patients with MS)]. Both the CD4(+)CD28(+) and CD4(+)CD28(-) T-cell populations of patients with MS produced higher levels of interferon (IFN)-gamma compared with those in HC. In particular, the proportion of IFN-gamma(+) cells among CD4(+)CD28(-) T cells from patients with MS was considerably high. However, expression of co-stimulatory molecules including inducible costimulator (ICOS), activating natural killer receptors, or members of tumor necrosis factor receptor family that replace CD28 in CD4(+)CD28(-) T cells of patients with MS could not be identified. A unique subpopulation bearing the CD45RA(high)CCR7(-) phenotype was identified among the CD4(+)CD28(-) T cells of some patients with MS. Because only MS samples contained this CD45RA(high)CCR7(-) population attributed to terminally differentiated effector memory cells and lacked naive CD45RA(high)CCR7(+) cells, we suggest that CD4(+)CD28(-) T cells of patients with MS represent a cell population which is in more differentiated state than healthy subjects. In patients treated with IFN-beta-1b, IFN-gamma production from CD4(+)CD28(+) T cells was suppressed compared with that in untreated patients. On the contrary, in the CD4(+)CD28(-) population, production of IFN-gamma in IFN-beta-1b-treated patients was not significantly suppressed compared with that in untreated patients with MS. Thus, an additional treatment strategy that specifically targets this cell population may enhance the beneficial effect of IFN-beta on MS.
Collapse
Affiliation(s)
- Y Miyazaki
- Division of Immunobiology, Research Section of Pathophysiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wiesemann E, Deb M, Trebst C, Hemmer B, Stangel M, Windhagen A. Effects of interferon-β on co-signaling molecules: upregulation of CD40, CD86 and PD-L2 on monocytes in relation to clinical response to interferon-β treatment in patients with multiple sclerosis. Mult Scler 2007; 14:166-76. [DOI: 10.1177/1352458507081342] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Interferon-beta (IFN-β) reduces disease activity in a subgroup of patients with relapsing remitting multiple sclerosis (MS). The mechanism of action as well as the pathophysiological basis of responsiveness to IFN-β is not well understood. Since T-cell activation plays an important part in the pathophysiology of MS, we here investigated the effect of IFN-β on the expression of co-signaling pathways (CD28—CD80/CD86, CD154—CD40, ICOS—ICOSL, PD-1—PD-L1/2) in MS patients and correlated the results with the clinical response to IFN-β in individual patients. Expression of co-signaling molecules was measured by flow cytometry in vitro on peripheral blood mononuclear cells after incubation with IFN-β, and in vivo in whole blood samples of 32 untreated and 24 IFN-β treated MS patients, including 13 patients longitudinal. IFN-β treatment induced upregulation of CD40, CD80, CD86, PD-L1 and PD-L2 on monocytes as well as PD-L1 on CD4+-T-cells in vitro and in vivo. IFN-β treated MS patients were grouped into responders and non-responders on the basis of Kurtzkés EDSS (expanded disability status scale) progression and relapse rate. Upregulation of CD40, CD86 and PD-L2 on monocytes was associated with treatment response to IFN-β ( P < 0.001, P = 0.028 and P = 0.028, respectively). Our results show that IFN-β upregulates co-stimulatory as well as co-inhibitory molecules in vitro and in vivo implicating that modulation of the balance between positive and negative co-stimulatory signals might be an important part of the mechanism of action of IFN-β in MS. Upregulation of the expression of CD40, CD86 and PD-L2 may be useful as a predictive marker for clinical response to IFN-β treatment at early timepoints during IFN-β therapy. Multiple Sclerosis 2008; 14: 166—176. http://msj.sagepub.com
Collapse
Affiliation(s)
- Elke Wiesemann
- Department of Neurology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Milani Deb
- Department of Neurology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Corinna Trebst
- Department of Neurology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der isar, Technische Universität Munich, Germany
| | - Martin Stangel
- Department of Neurology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Anja Windhagen
- Department of Neurology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany,
| |
Collapse
|
39
|
Neuhaus O, Kieseier BC, Hartung HP. Pharmacokinetics and pharmacodynamics of the interferon-betas, glatiramer acetate, and mitoxantrone in multiple sclerosis. J Neurol Sci 2007; 259:27-37. [PMID: 17391705 DOI: 10.1016/j.jns.2006.05.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 04/25/2006] [Accepted: 05/01/2006] [Indexed: 11/15/2022]
Abstract
Five disease-modifying agents are currently approved for long-term treatment of multiple sclerosis (MS), namely three interferon-beta preparations, glatiramer acetate, and mitoxantrone(1). Pharmacokinetics describes the fate of drugs in the human body by studying their absorption, distribution, metabolism and excretion. Pharmacodynamics is dedicated to the mechanisms of action of drugs. The understanding of the pharmacokinetics and pharmacodynamics of the approved disease-modifying agents against MS is of importance as it might contribute to the development of future derivatives with a potentially higher efficacy and a more favourable safety profile. This article reviews data thus far present both on the pharmacokinetics as well as on the putative mechanisms of action of the interferon-betas, glatiramer acetate, and mitoxantrone in the immunopathogenesis of MS.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Department of Neurology, Heinrich Heine University, Düsseldorf, Germany.
| | | | | |
Collapse
|
40
|
Kantor AB, Deng J, Waubant E, Lin H, Becker CH, Lacy JR, Perrone AM, Bennett D, Goelz SE. Identification of short-term pharmacodynamic effects of interferon-beta-1a in multiple sclerosis subjects with broad- based phenotypic profiling. J Neuroimmunol 2007; 188:103-16. [DOI: 10.1016/j.jneuroim.2007.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 04/26/2007] [Accepted: 05/07/2007] [Indexed: 11/29/2022]
|
41
|
Gok B, Okutan O, Beskonakli E, Palaoglu S, Erdamar H, Sargon MF. Effect of immunomodulation with human interferon-beta on early functional recovery from experimental spinal cord injury. Spine (Phila Pa 1976) 2007; 32:873-80. [PMID: 17426631 DOI: 10.1097/01.brs.0000259841.40358.8f] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
STUDY DESIGN Electron and light microscopic changes, neutrophil infiltration, and lipid peroxidation in the spinal cord and early neurologic examination were studied in rats. OBJECTIVE To examine the effects of immunomodulator treatment with recombinant human interferon-beta after spinal cord contusion injury. SUMMARY OF BACKGROUND DATA Immunomodulator treatment with interferon-beta has been the subject of extensive studies, but mainly in relation to multiple sclerosis. Recently, it was reported that interferon-beta possessed significant neuroprotection after experimental transient ischemic stroke. However, to our knowledge, there have been no previous reports about the neuroprotective effect of interferon-beta after spinal cord injury. METHODS Rats were randomly allocated into 5 groups. Group 1 was control and after clinical examination, normal spinal cord samples were obtained. Group 2 was introduced 50 g/cm contusion injury. Group 3 was vehicle, immediately after trauma 1 mL of physiologic saline was injected. Group 4 was given 30 mg/kg methylprednisolone sodium succinate intraperitoneally immediately after trauma. Group 5 was given 1 x 10(7) IU interferon-beta immediately and 0.5 x 10(7) IU interferon-beta 4 hours after trauma. Animals were examined by inclined plane and Basso-Beattie-Bresnahan scale 24 hours after trauma. Spinal cord samples obtained following clinical evaluations. Neutrophil infiltration was evaluated by myeloperoxidase activity and lipid peroxidation was estimated by thiobarbituric acid test. Electron and light microscopic results were also performed to determine the effects of interferon-beta on tissue structure. RESULTS Interferon-beta treatment improved neurologic outcome, which was supported by decreased myeloperoxidase activity and lipid peroxidation. Electron and light microscopic results also showed preservation of tissue structure in the treatment group. CONCLUSIONS Immunomodulator treatment with interferon-beta possesses obvious neuroprotection after acute contusion injury to the rat spinal cord.
Collapse
Affiliation(s)
- Beril Gok
- Department of Neurological Surgery, Ankara Ataturk Research and Education Hospital, Ankara, Turkey.
| | | | | | | | | | | |
Collapse
|
42
|
Sorensen PS. The gap between effect of drugs and effectiveness of treatments. J Neurol Sci 2007; 259:128-32. [PMID: 17362994 DOI: 10.1016/j.jns.2006.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2006] [Accepted: 10/16/2006] [Indexed: 11/16/2022]
Abstract
Despite profound effects on the immune system, drugs for multiple sclerosis (MS) therapy have shown only moderate treatment effectiveness. The approved drugs, interferon (IFN)-beta and glatiramer acetate, have a number of effects on the immune system that could interfere with the disease processes in MS but are only able to reduce the relapse rate by 30% and have little or no effect on disease progression. The new targeted immune therapies, campath-1H and natalizumab, have shown immense treatment effectiveness as for inflammation-related disease manifestations, i.e. relapses and MRI activity, but an effect on long-term disease progression has not yet been demonstrated. There are several explanations of the gap between drug effects and treatment effectiveness of which some are related to the properties of the immune system and some are related to the properties of drugs used for treatment of MS. To fill the gap we need to have drugs that both effectively and safely eliminate the inflammation and in addition have neuroprotective properties. However, this may not be obtained from a single drug but may require combinations of drugs with different actions on the disease processes.
Collapse
Affiliation(s)
- Per Soelberg Sorensen
- Copenhagen Multiple Sclerosis Research Center, Department of Neurology 2082, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
43
|
Stevens SL, Stenzel-Poore MP. Toll-like receptors and tolerance to ischaemic injury in the brain. Biochem Soc Trans 2007; 34:1352-5. [PMID: 17073817 DOI: 10.1042/bst0341352] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Ischaemic tolerance in the brain is a powerful adaptive defence that involves an endogenous programme of neuroprotection culminating in marked protection against brain injury from ischaemia. A range of preconditioning stimuli exist that differ in ligand and target characteristics but share the common feature of causing mild stress or insult without inducing overt injury. The protective phenotype that emerges confers tolerance to subsequent exposure to injurious insults. Tolerance to injury is the result of genomic reprogramming, an adaptation comprising regulatory processes that countermand injurious effectors and invoke novel neuroprotective pathways. TLRs (Toll-like receptors) play important roles in sensing potential danger/insult in the form of pathogens as well as endogenous stress molecules that occur in response to mild injury (e.g. heat-shock proteins). Recent studies suggest that TLRs are novel and potent preconditioning targets that offer substantial promise to protect the brain from ischaemic injury.
Collapse
Affiliation(s)
- S L Stevens
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, 3181 Sam Jackson Park Road, Portland, OR, USA
| | | |
Collapse
|
44
|
Jensen J, Langkilde AR, Frederiksen JL, Sellebjerg F. CD8+ T cell activation correlates with disease activity in clinically isolated syndromes and is regulated by interferon-beta treatment. J Neuroimmunol 2006; 179:163-72. [PMID: 16919783 DOI: 10.1016/j.jneuroim.2006.06.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2005] [Revised: 06/02/2006] [Accepted: 06/30/2006] [Indexed: 10/24/2022]
Abstract
An increased percentage of blood CD8+ T cells from patients with clinically isolated syndromes (CIS) suggestive of multiple sclerosis (MS) was found to express CD26 and CD69. The percentage of CD26 or CD69 positive CD8+ T cells was higher in patients with MRI evidence of disease dissemination in space or with active MRI lesions than in the remaining patients. Treatment of MS with interferon (IFN)-beta resulted in a decrease in the percentage of CD26 and CD71 positive CD8+ T cells and an increase in the percentage of CD8+ T cells that expressed interleukin (IL)-10 and IL-13. CD8+ T cell activation in MS may be linked to disease activity already at disease onset, and is regulated by treatment with IFN-beta.
Collapse
Affiliation(s)
- J Jensen
- The MS Clinic, Department of Neurology, University of Copenhagen, Glostrup Hospital, Glostrup, Denmark
| | | | | | | |
Collapse
|
45
|
Marckmann S, Wiesemann E, Hilse R, Trebst C, Stangel M, Windhagen A. Interferon-beta up-regulates the expression of co-stimulatory molecules CD80, CD86 and CD40 on monocytes: significance for treatment of multiple sclerosis. Clin Exp Immunol 2005; 138:499-506. [PMID: 15544628 PMCID: PMC1809242 DOI: 10.1111/j.1365-2249.2004.02624.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Interferon (IFN)-beta reduces the biological activity of multiple sclerosis (MS), a presumably T cell-mediated autoimmune disease of central nervous system (CNS) myelin. Co-stimulatory molecules are necessary for full T cell activation and differential expression of co-stimulatory molecules on antigen-presenting cells is thought to influence the type of effector T cell response (Th1/Th2). In this study we investigated the effects of IFN-beta on the expression of co-stimulatory molecules on lymphocytes and monocytes as a potential mechanism of action of IFN-beta in MS. Peripheral blood mononuclear cells (PBMCs) were stimulated with IFN-beta in vitro and expression of CD80, CD86, CD40 and HLA was examined by flow cytometry and reverse-transcription polymerase chain reaction. Whereas IFN-beta had no effect on the expression of these molecules on T and B lymphocytes there was a significant increase on monocytes. Correspondingly, the expression of mRNA increased after 6-18 h. This in vitro response was also observed in untreated MS patients and patients receiving treatment with IFN-beta. The increase of co-stimulatory molecules on monocytes was not mediated by interleukin (IL)-10. When IFN-beta-stimulated monocytes were used to stimulate autologous T cells an increased secretion of IL-13 was observed. In biopsies taken from IFN-beta-induced skin reactions after subcutaneous injection increased expression of CD80 mRNA was detected, indicating that IFN-beta also up-regulates this co-stimulatory molecule in vivo. These data provide the background for further studies of IFN-beta-induced changes of co-stimulatory molecules in MS patients.
Collapse
Affiliation(s)
- S Marckmann
- Department of Neurology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Affiliation(s)
- Ludwig Kappos
- Department of Neurology, University Hospitals, Kantonsspital, CH-4031 Basel, Switzerland.
| |
Collapse
|
47
|
Abstract
Significant advances in magnetic resonance imaging (MRI) technology and treatment of multiple sclerosis (MS) have been made during the past decade. These advances have revealed evidence of profound heterogeneity in MS. There is a clear need to revisit the key issues in MS pathogenesis and treatment strategies, taking new data into consideration. This paper provides an overview of recent progress in MS research, including (a) a review of clinical, pathologic, and immunologic aspects of MS, (b) a discussion of the mechanism of action of currently available disease-modifying drugs for MS, (c) an account of the role of MRI in clinical management and clinical trials in MS, and (d) an overview of some emerging treatments for MS.
Collapse
Affiliation(s)
- Jingwu Zhang
- Baylor-Methodist Multiple Sclerosis Center, Department of Neurology, Baylor College of Medicine, Houston, Texas, USA.
| | | |
Collapse
|
48
|
Abstract
Multiple sclerosis (MS) is one of the most common chronic neurological diseases in young adults in western countries. An important aspect of treatment of this disease is the use of interferons (IFNs). These are molecules with antiviral, immunomodulatory, antiproliferative and hormonal activities. IFNbeta, a class I IFN, has been used extensively in the therapy of MS, particularly in its relapsing-remitting (RRMS) phase, the most frequent clinical form of the disease. Although the available evidence from published clinical trials is difficult to evaluate because of methodological differences, an unbiased review of the data reveals sufficient evidence to conclude that treatment with IFNbeta in RRMS is both efficacious and safe, at least over the periods so far investigated (up to 4-6 years). While there is no reason to suspect that IFNbeta should not continue to be efficacious and safe over the longer term, studies investigating these questions over longer periods and including greater numbers of patients are needed.
Collapse
Affiliation(s)
- Oscar Fernández
- Institute of Neurosciences, Neurology Service, Hospital Regional Universitario Carlos Haya, Málaga, Spain.
| |
Collapse
|
49
|
Stickler M, Valdes AM, Gebel W, Razo OJ, Faravashi N, Chin R, Rochanayon N, Harding FA. The HLA-DR2 haplotype is associated with an increased proliferative response to the immunodominant CD4(+) T-cell epitope in human interferon-beta. Genes Immun 2004; 5:1-7. [PMID: 14735143 DOI: 10.1038/sj.gene.6364027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human CD4(+) T-cell epitopes were identified in interferon-beta (IFN-beta)-1b. A prominent peptide epitope region was found that induced a proliferative response in 16% of all donors tested. Responses corresponded to the presence of the HLA-DR2 haplotype. Responsive donors expressing the HLA-DQ6 allele showed an increased level of proliferation to the epitope as compared to peptide-responsive HLA-DQ6 negative donors. A similar result was found for HLA-DR15-expressing donors. PBMC from donors expressing HLA-DR15 were more likely to proliferate in response to IFN-beta in a whole-protein in vitro assay than donors who did not carry this haplotype. It is striking that the common DQ6 allele HLA-DQB1(*)0602 is found in linkage disequilibrium with HLA-DRB1(*)1501, and this combination defines the HLA genotype associated with the development of multiple sclerosis. The HLA association between a response to IFN-beta and MS might explain the prevalence of neutralizing antibody development, and may underlie the etiology of the disease.
Collapse
Affiliation(s)
- M Stickler
- Genencor International, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Miller A, Glass-Marmor L, Abraham M, Grossman I, Shapiro S, Galboiz Y. Bio-markers of disease activity and response to therapy in multiple sclerosis. Clin Neurol Neurosurg 2004; 106:249-54. [PMID: 15177778 DOI: 10.1016/j.clineuro.2004.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Ariel Miller
- Division of Neuroimmunology and Multiple Sclerosis Center, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, 7 Michal Street, Haifa 34362, Israel.
| | | | | | | | | | | |
Collapse
|