1
|
Becher B, Derfuss T, Liblau R. Targeting cytokine networks in neuroinflammatory diseases. Nat Rev Drug Discov 2024; 23:862-879. [PMID: 39261632 DOI: 10.1038/s41573-024-01026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
In neuroinflammatory diseases, systemic (blood-borne) leukocytes invade the central nervous system (CNS) and lead to tissue damage. A causal relationship between neuroinflammatory diseases and dysregulated cytokine networks is well established across several preclinical models. Cytokine dysregulation is also observed as an inadvertent effect of cancer immunotherapy, where it often leads to neuroinflammation. Neuroinflammatory diseases can be separated into those in which a pathogen is at the centre of the immune response and those of largely unknown aetiology. Here, we discuss the pathophysiology, cytokine networks and therapeutic landscape of 'sterile' neuroinflammatory diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), neurosarcoidosis and immune effector cell-associated neurotoxicity syndrome (ICANS) triggered by cancer immunotherapy. Despite successes in targeting cytokine networks in preclinical models of neuroinflammation, the clinical translation of targeting cytokines and their receptors has shown mixed and often paradoxical responses.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Tobias Derfuss
- Department of Neurology and Biomedicine, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Roland Liblau
- Institute for inflammatory and infectious diseases, INSERM UMR1291 - CNRS UMR505, Toulouse, France.
| |
Collapse
|
2
|
Wang J, Brown K, Danehy C, Mérigeon E, Goralski S, Rice S, Torgbe K, Thomas F, Block D, Olsen H, Strome SE, Fitzpatrick EA. Fc multimers effectively treat murine models of multiple sclerosis. Front Immunol 2023; 14:1199747. [PMID: 37638040 PMCID: PMC10451071 DOI: 10.3389/fimmu.2023.1199747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Multiple Sclerosis (MS) is a chronic neurodegenerative disease with limited therapeutic options. Recombinant Fc multimers (rFc), designed to mirror many of the anti-inflammatory activities of Intravenous Immunoglobulin (IVIG), have been shown to effectively treat numerous immune-mediated diseases in rodents. In this study we used the experimental autoimmune encephalomyelitis (EAE) murine model of MS to test the efficacy of a rFc, M019, that consists of multimers of the Fc portion of IgG2, in inhibiting disease severity. We show that M019 effectively reduced clinical symptoms when given either pre- or post-symptom onset compared to vehicle treated EAE induced mice. M019 was effective in reducing symptoms in both SJL model of relapsing remitting MS as well as the B6 model of chronic disease. M019 binds to FcγR bearing-monocytes both in vivo and in vitro and prevented immune cell infiltration into the CNS of treated mice. The lack of T cell infiltration into the spinal cord was not due to a decrease in T cell priming; there was an equivalent frequency of Th17 cells in the spleens of M019 and vehicle treated EAE induced mice. Surprisingly, there was an increase in chemokines in the sera but not in the CNS of M019 treated mice compared to vehicle treated animals. We postulate that M019 interacts with a FcγR rich monocyte intermediary to prevent T cell migration into the CNS and demyelination.
Collapse
Affiliation(s)
- Jin Wang
- Dept. of Microbiology Immunology and Biochemistry, UTHSC, Memphis, TN, United States
| | - Kellie Brown
- Dept. of Microbiology Immunology and Biochemistry, UTHSC, Memphis, TN, United States
| | - Caroline Danehy
- College of Graduate Health Sciences, UTHSC, Memphis, TN, United States
| | | | | | - Samuel Rice
- College of Medicine, UTHSC, Memphis, TN, United States
| | - Kwame Torgbe
- Dept. of Pathology, UTHSC, Memphis, TN, United States
| | - Fridtjof Thomas
- Div. of Biostatistics, Dept. of Preventive Medicine, UTHSC, Memphis, TN, United States
| | | | | | - Scott E. Strome
- Dept. of Microbiology Immunology and Biochemistry, UTHSC, Memphis, TN, United States
| | | |
Collapse
|
3
|
Radandish M, Khalilian P, Esmaeil N. The Role of Distinct Subsets of Macrophages in the Pathogenesis of MS and the Impact of Different Therapeutic Agents on These Populations. Front Immunol 2021; 12:667705. [PMID: 34489926 PMCID: PMC8417824 DOI: 10.3389/fimmu.2021.667705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/31/2021] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating inflammatory disorder of the central nervous system (CNS). Besides the vital role of T cells, other immune cells, including B cells, innate immune cells, and macrophages (MФs), also play a critical role in MS pathogenesis. Tissue-resident MФs in the brain’s parenchyma, known as microglia and monocyte-derived MФs, enter into the CNS following alterations in CNS homeostasis that induce inflammatory responses in MS. Although the neuroprotective and anti-inflammatory actions of monocyte-derived MФs and resident MФs are required to maintain CNS tolerance, they can release inflammatory cytokines and reactivate primed T cells during neuroinflammation. In the CNS of MS patients, elevated myeloid cells and activated MФs have been found and associated with demyelination and axonal loss. Thus, according to the role of MФs in neuroinflammation, they have attracted attention as a therapeutic target. Also, due to their different origin, location, and turnover, other strategies may require to target the various myeloid cell populations. Here we review the role of distinct subsets of MФs in the pathogenesis of MS and different therapeutic agents that target these cells.
Collapse
Affiliation(s)
- Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Khalilian
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
4
|
Aqel SI, Yang X, Kraus EE, Song J, Farinas MF, Zhao EY, Pei W, Lovett-Racke AE, Racke MK, Li C, Yang Y. A STAT3 inhibitor ameliorates CNS autoimmunity by restoring Teff:Treg balance. JCI Insight 2021; 6:142376. [PMID: 33411696 PMCID: PMC7934926 DOI: 10.1172/jci.insight.142376] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Reestablishing an appropriate balance between T effector cells (Teff) and Tregs is essential for correcting autoimmunity. Multiple sclerosis (MS) is an immune-mediated chronic CNS disease characterized by neuroinflammation, demyelination, and neuronal degeneration, in which the Teff:Treg balance is skewed toward pathogenic Teffs Th1 and Th17 cells. STAT3 is a key regulator of Teff:Treg balance. Using the structure-based design, we have developed a potentially novel small-molecule prodrug LLL12b that specifically inhibits STAT3 and suppresses Th17 differentiation and expansion. Moreover, LLL12b regulates the fate decision between Th17 and Tregs in an inflammatory environment, shifting Th17:Treg balance toward Tregs and favoring the resolution of inflammation. Therapeutic administration of LLL12b after disease onset significantly suppresses disease progression in adoptively transferred, chronic, and relapsing-remitting experimental autoimmune encephalomyelitis. Disease relapses were also significantly suppressed by LLL12b given during the remission phase. Additionally, LLL12b shifts Th17:Treg balance of CD4+ T cells from MS patients toward Tregs and increases Teff sensitivity to Treg-mediated suppression. These data suggest that selective inhibition of STAT3 by the small molecule LLL12b recalibrates the effector and regulatory arms of CD4+ T responses, representing a potentially clinically translatable therapeutic strategy for MS.
Collapse
Affiliation(s)
- Saba I Aqel
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Xiaozhi Yang
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Emma E Kraus
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Jinhua Song
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Marissa F Farinas
- Neuroscience program, College of Arts and Sciences, OSU, Columbus, Ohio, USA
| | - Erin Y Zhao
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Wei Pei
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, OSU Wexner Medical Center, Columbus, Ohio, USA
| | - Michael K Racke
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA.,Quest Diagnostics, Secaucus, New Jersey, USA
| | - Chenglong Li
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Yuhong Yang
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA.,Department of Microbial Infection and Immunity, OSU Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
5
|
Zahoor I, Rui B, Khan J, Datta I, Giri S. An emerging potential of metabolomics in multiple sclerosis: a comprehensive overview. Cell Mol Life Sci 2021; 78:3181-3203. [PMID: 33449145 PMCID: PMC8038957 DOI: 10.1007/s00018-020-03733-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/14/2020] [Accepted: 12/07/2020] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the nervous system that primarily affects young adults. Although the exact etiology of the disease remains obscure, it is clear that alterations in the metabolome contribute to this process. As such, defining a reliable and disease-specific metabolome has tremendous potential as a diagnostic and therapeutic strategy for MS. Here, we provide an overview of studies aimed at identifying the role of metabolomics in MS. These offer new insights into disease pathophysiology and the contributions of metabolic pathways to this process, identify unique markers indicative of treatment responses, and demonstrate the therapeutic effects of drug-like metabolites in cellular and animal models of MS. By and large, the commonly perturbed pathways in MS and its preclinical model include lipid metabolism involving alpha-linoleic acid pathway, nucleotide metabolism, amino acid metabolism, tricarboxylic acid cycle, d-ornithine and d-arginine pathways with collective role in signaling and energy supply. The metabolomics studies suggest that metabolic profiling of MS patient samples may uncover biomarkers that will advance our understanding of disease pathogenesis and progression, reduce delays and mistakes in diagnosis, monitor the course of disease, and detect better drug targets, all of which will improve early therapeutic interventions and improve evaluation of response to these treatments.
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA. .,Department of Neurology, Henry Ford Hospital, Education & Research Building, Room 4023, 2799 W Grand Blvd, Detroit, MI, 48202, USA.
| | - Bin Rui
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Junaid Khan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Indrani Datta
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA. .,Department of Neurology, Henry Ford Hospital, Education & Research Building, Room 4051, 2799 W Grand Blvd, Detroit, MI, 48202, USA.
| |
Collapse
|
6
|
Monaghan KL, Wan EC. The Role of Granulocyte-Macrophage Colony-Stimulating Factor in Murine Models of Multiple Sclerosis. Cells 2020; 9:cells9030611. [PMID: 32143326 PMCID: PMC7140439 DOI: 10.3390/cells9030611] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease that predominantly impacts the central nervous system (CNS). Animal models have been used to elucidate the underpinnings of MS pathology. One of the most well-studied models of MS is experimental autoimmune encephalomyelitis (EAE). This model was utilized to demonstrate that the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) plays a critical and non-redundant role in mediating EAE pathology, making it an ideal therapeutic target. In this review, we will first explore the role that GM-CSF plays in maintaining homeostasis. This is important to consider, because any therapeutics that target GM-CSF could potentially alter these regulatory processes. We will then focus on current findings related to the function of GM-CSF signaling in EAE pathology, including the cell types that produce and respond to GM-CSF and the role of GM-CSF in both acute and chronic EAE. We will then assess the role of GM-CSF in alternative models of MS and comment on how this informs the understanding of GM-CSF signaling in the various aspects of MS immunopathology. Finally, we will examine what is currently known about GM-CSF signaling in MS, and how this has promoted clinical trials that directly target GM-CSF.
Collapse
Affiliation(s)
- Kelly L. Monaghan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA;
| | - Edwin C.K. Wan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506, USA;
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Correspondence: ; Tel.:+1-304-293-6293
| |
Collapse
|
7
|
Glatigny S, Bettelli E. Experimental Autoimmune Encephalomyelitis (EAE) as Animal Models of Multiple Sclerosis (MS). Cold Spring Harb Perspect Med 2018; 8:cshperspect.a028977. [PMID: 29311122 DOI: 10.1101/cshperspect.a028977] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a multifocal demyelinating disease of the central nervous system (CNS) leading to the progressive destruction of the myelin sheath surrounding axons. It can present with variable clinical and pathological manifestations, which might reflect the involvement of distinct pathogenic processes. Although the mechanisms leading to the development of the disease are not fully understood, numerous evidences indicate that MS is an autoimmune disease, the initiation and progression of which are dependent on an autoimmune response against myelin antigens. In addition, genetic susceptibility and environmental triggers likely contribute to the initiation of the disease. At this time, there is no cure for MS, but several disease-modifying therapies (DMTs) are available to control and slow down disease progression. A good number of these DMTs were identified and tested using animal models of MS referred to as experimental autoimmune encephalomyelitis (EAE). In this review, we will recapitulate the characteristics of EAE models and discuss how they help shed light on MS pathogenesis and help test new treatments for MS patients.
Collapse
Affiliation(s)
- Simon Glatigny
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101.,Department of Immunology, University of Washington, Seattle, Washington 98109
| | - Estelle Bettelli
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101.,Department of Immunology, University of Washington, Seattle, Washington 98109
| |
Collapse
|
8
|
Urinary and Plasma Metabolomics Identify the Distinct Metabolic Profile of Disease State in Chronic Mouse Model of Multiple Sclerosis. J Neuroimmune Pharmacol 2018; 14:241-250. [DOI: 10.1007/s11481-018-9815-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
|
9
|
Tiwari S, Lapierre J, Ojha CR, Martins K, Parira T, Dutta RK, Caobi A, Garbinski L, Ceyhan Y, Esteban-Lopez M, El-Hage N. Signaling pathways and therapeutic perspectives related to environmental factors associated with multiple sclerosis. J Neurosci Res 2018; 96:1831-1846. [PMID: 30204260 PMCID: PMC7167107 DOI: 10.1002/jnr.24322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disorder of unknown etiology. Both genetic-susceptibility and environment exposures, including vitamin D deficiency, Epstein-Barr viral and Herpesvirus (HHV-6) infections are strongly implicated in the activation of T cells and MS-pathogenesis. Despite precise knowledge of how these factors could be operating alone or in combination to facilitate and aggravate the disease progression, it is clear that prolonged induction of inflammatory molecules and recruitment of other immune cells by the activated T cells results in demyelination and axonal damage. It is imperative to understand the risk factors associated with MS progression and how these factors contribute to disease pathology. Understanding of the underlying mechanisms of what factors triggers activation of T cells to attack myelin antigen are important to strategize therapeutics and therapies against MS. Current review provides a detailed literature to understand the role of both pathogenic and non-pathogenic factors on the impact of MS.
Collapse
Affiliation(s)
- Sneham Tiwari
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Jessica Lapierre
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Chet Raj Ojha
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Kyle Martins
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Tiyash Parira
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Rajib Kumar Dutta
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Allen Caobi
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Luis Garbinski
- Cell Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Yasemin Ceyhan
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Maria Esteban-Lopez
- Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| | - Nazira El-Hage
- Departments of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
10
|
Wang D, Ghosh D, Islam SMT, Moorman CD, Thomason AE, Wilkinson DS, Mannie MD. IFN-β Facilitates Neuroantigen-Dependent Induction of CD25+ FOXP3+ Regulatory T Cells That Suppress Experimental Autoimmune Encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2992-3007. [PMID: 27619998 PMCID: PMC5101178 DOI: 10.4049/jimmunol.1500411] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 08/07/2016] [Indexed: 12/11/2022]
Abstract
This study introduces a flexible format for tolerogenic vaccination that incorporates IFN-β and neuroantigen (NAg) in the Alum adjuvant. Tolerogenic vaccination required all three components, IFN-β, NAg, and Alum, for inhibition of experimental autoimmune encephalomyelitis (EAE) and induction of tolerance. Vaccination with IFN-β + NAg in Alum ameliorated NAg-specific sensitization and inhibited EAE in C57BL/6 mice in pretreatment and therapeutic regimens. Tolerance induction was specific for the tolerogenic vaccine Ag PLP178-191 or myelin oligodendrocyte glycoprotein (MOG)35-55 in proteolipid protein- and MOG-induced models of EAE, respectively, and was abrogated by pretreatment with a depleting anti-CD25 mAb. IFN-β/Alum-based vaccination exhibited hallmarks of infectious tolerance, because IFN-β + OVA in Alum-specific vaccination inhibited EAE elicited by OVA + MOG in CFA but not EAE elicited by MOG in CFA. IFN-β + NAg in Alum vaccination elicited elevated numbers and percentages of FOXP3+ T cells in blood and secondary lymphoid organs in 2D2 MOG-specific transgenic mice, and repeated boosters facilitated generation of activated CD44high CD25+ regulatory T cell (Treg) populations. IFN-β and MOG35-55 elicited suppressive FOXP3+ Tregs in vitro in the absence of Alum via a mechanism that was neutralized by anti-TGF-β and that resulted in the induction of an effector CD69+ CTLA-4+ IFNAR+ FOXP3+ Treg subset. In vitro IFN-β + MOG-induced Tregs inhibited EAE when transferred into actively challenged recipients. Unlike IFN-β + NAg in Alum vaccines, vaccination with TGF-β + MOG35-55 in Alum did not increase Treg percentages in vivo. Overall, this study indicates that IFN-β + NAg in Alum vaccination elicits NAg-specific, suppressive CD25+ Tregs that inhibit CNS autoimmune disease. Thus, IFN-β has the activity spectrum that drives selective responses of suppressive FOXP3+ Tregs.
Collapse
Affiliation(s)
- Duncheng Wang
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834; and
| | - Debjani Ghosh
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834; and
| | - S M Touhidul Islam
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834; and
| | - Cody D Moorman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834; and
| | - Ashton E Thomason
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834; and
| | - Daniel S Wilkinson
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834; and
| | - Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834; and
- The Harriet and John Wooten Laboratory for Alzheimer's and Neurodegenerative Disease Research, Brody School of Medicine, East Carolina University, Greenville, NC 27834
| |
Collapse
|
11
|
Kudo D, Uno K, Aoyagi T, Akahori Y, Ishii K, Kanno E, Maruyama R, Kushimoto S, Kaku M, Kawakami K. Low-dose interferon-α treatment improves survival and inflammatory responses in a mouse model of fulminant acute respiratory distress syndrome. Inflammation 2014; 36:812-20. [PMID: 23407996 PMCID: PMC7088027 DOI: 10.1007/s10753-013-9607-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is accompanied by severe lung inflammation induced by various diseases. Despite the severity of symptoms, therapeutic strategies for this pathologic condition are still poorly developed. Interferon (IFN)-α is well known as an antiviral cytokine and low-dose IFN-α has been reported to show antiinflammatory effects. Therefore, we investigated how this cytokine affected ARDS in a mouse model. C57BL/6 mice received sequential intratracheal administration of α-galactosylceramide (α-GalCer) and lipopolysaccharide (LPS), which resulted in the development of fulminant ARDS. These mice were then treated intranasally with IFN-α and their survival, lung weight, pathological findings, and cytokine production were evaluated. Administration of low-dose IFN-α prolonged survival of fulminant ARDS mice, but higher doses of IFN-α did not. Histological analysis showed that low-dose IFN-α treatment improved findings of diffuse alveolar damage in fulminant ARDS mice, which was associated with reduction in the wet/dry (W/D) lung weight ratio. Furthermore, IFN-γ production in the lungs was significantly reduced in IFN-α-treated mice, compared with control mice, but tumor necrosis factor (TNF)-α production was almost equivalent for both groups. Low-dose IFN-α shows antiinflammatory and therapeutic effects in a mouse model of fulminant ARDS, and reduced production of IFN-γ in the lung may be involved in the beneficial effect of this treatment.
Collapse
Affiliation(s)
- Daisuke Kudo
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Owens T, Khorooshi R, Wlodarczyk A, Asgari N. Interferons in the central nervous system: A few instruments play many tunes. Glia 2013; 62:339-55. [DOI: 10.1002/glia.22608] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Trevor Owens
- Department of Neurobiology Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
| | - Nasrin Asgari
- Department of Neurobiology Research, Institute of Molecular Medicine; University of Southern Denmark; Odense Denmark
- Department of Neurology; Vejle Hospital; Denmark
| |
Collapse
|
13
|
Abstract
It is widely accepted that the main common pathogenetic pathway in multiple sclerosis (MS) involves an immune-mediated cascade initiated in the peripheral immune system and targeting CNS myelin. Logically, therefore, the therapeutic approaches to the disease include modalities aiming at downregulation of the various immune elements that are involved in this immunologic cascade. Since the introduction of interferons in 1993, which were the first registered treatments for MS, huge steps have been made in the field of MS immunotherapy. More efficious and specific immunoactive drugs have been introduced and it appears that the increased specificity for MS of these new treatments is paralleled by greater efficacy. Unfortunately, this seemingly increased efficacy has been accompanied by more safety issues. The immunotherapeutic modalities can be divided into two main groups: those affecting the acute stages (relapses) of the disease and the long-term treatments that are aimed at preventing the appearance of relapses and the progression in disability. Immunomodulating treatments may also be classified according to the level of the 'immune axis' where they exert their main effect. Since, in MS, a neurodegenerative process runs in parallel and as a consequence of inflammation, early immune intervention is warranted to prevent progression of relapses of MS and the accumulation of disability. The use of neuroimaging (MRI) techniques that allow the detection of silent inflammatory activity of MS and neurodegeneration has provided an important tool for the substantiation of the clinical efficacy of treatments and the early diagnosis of MS. This review summarizes in detail the existing information on all the available immunotherapies for MS, old and new, classifies them according to their immunologic mechanisms of action and proposes a structured algorithm/therapeutic scheme for the management of the disease.
Collapse
|
14
|
Abstract
In multiple sclerosis, type I interferon (IFN) is considered immune-modulatory, and recombinant forms of IFN-β are the most prescribed treatment for this disease. This is in contrast to most other autoimmune disorders, because type I IFN contributes to the pathologies. Even within the relapsing-remitting multiple sclerosis (RRMS) population, 30-50% of MS patients are non-responsive to this treatment, and it consistently worsens neuromyelitis optica, a disease similar to RRMS. In this article, we discuss the recent advances in the field of autoimmunity and introduce the theory explain how type I IFNs can be pro-inflammatory in disease that is predominantly driven by a Th17 response and are therapeutic when disease is predominantly Th1.
Collapse
Affiliation(s)
- Robert C Axtell
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305-5316, USA.
| | | |
Collapse
|
15
|
Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2012; 164:1079-106. [PMID: 21371012 DOI: 10.1111/j.1476-5381.2011.01302.x] [Citation(s) in RCA: 1014] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple sclerosis (MS). EAE is a complex condition in which the interaction between a variety of immunopathological and neuropathological mechanisms leads to an approximation of the key pathological features of MS: inflammation, demyelination, axonal loss and gliosis. The counter-regulatory mechanisms of resolution of inflammation and remyelination also occur in EAE, which, therefore can also serve as a model for these processes. Moreover, EAE is often used as a model of cell-mediated organ-specific autoimmune conditions in general. EAE has a complex neuropharmacology, and many of the drugs that are in current or imminent use in MS have been developed, tested or validated on the basis of EAE studies. There is great heterogeneity in the susceptibility to the induction, the method of induction and the response to various immunological or neuropharmacological interventions, many of which are reviewed here. This makes EAE a very versatile system to use in translational neuro- and immunopharmacology, but the model needs to be tailored to the scientific question being asked. While creating difficulties and underscoring the inherent weaknesses of this model of MS in straightforward translation from EAE to the human disease, this variability also creates an opportunity to explore multiple facets of the immune and neural mechanisms of immune-mediated neuroinflammation and demyelination as well as intrinsic protective mechanisms. This allows the eventual development and preclinical testing of a wide range of potential therapeutic interventions.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neurology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | | | | | | |
Collapse
|
16
|
Farooqi N, Gran B, Constantinescu CS. Are current disease-modifying therapeutics in multiple sclerosis justified on the basis of studies in experimental autoimmune encephalomyelitis? J Neurochem 2010; 115:829-44. [DOI: 10.1111/j.1471-4159.2010.06982.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
17
|
Exacerbated innate host response to SARS-CoV in aged non-human primates. PLoS Pathog 2010; 6:e1000756. [PMID: 20140198 PMCID: PMC2816697 DOI: 10.1371/journal.ppat.1000756] [Citation(s) in RCA: 262] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 01/06/2010] [Indexed: 12/15/2022] Open
Abstract
The emergence of viral respiratory pathogens with pandemic potential, such as severe acute respiratory syndrome coronavirus (SARS-CoV) and influenza A H5N1, urges the need for deciphering their pathogenesis to develop new intervention strategies. SARS-CoV infection causes acute lung injury (ALI) that may develop into life-threatening acute respiratory distress syndrome (ARDS) with advanced age correlating positively with adverse disease outcome. The molecular pathways, however, that cause virus-induced ALI/ARDS in aged individuals are ill-defined. Here, we show that SARS-CoV-infected aged macaques develop more severe pathology than young adult animals, even though viral replication levels are similar. Comprehensive genomic analyses indicate that aged macaques have a stronger host response to virus infection than young adult macaques, with an increase in differential expression of genes associated with inflammation, with NF-κB as central player, whereas expression of type I interferon (IFN)-β is reduced. Therapeutic treatment of SARS-CoV-infected aged macaques with type I IFN reduces pathology and diminishes pro-inflammatory gene expression, including interleukin-8 (IL-8) levels, without affecting virus replication in the lungs. Thus, ALI in SARS-CoV-infected aged macaques developed as a result of an exacerbated innate host response. The anti-inflammatory action of type I IFN reveals a potential intervention strategy for virus-induced ALI. Severe acute respiratory syndrome coronavirus (SARS-CoV) infection causes acute lung injury that may develop into the life-threatening acute respiratory distress syndrome (ARDS) in mostly elderly individuals. Although SARS-CoV infection can be fatal, most patients recover, suggesting that protective host responses are operational to combat the viral infection. Therefore, we used age as predisposing factor to obtain insight into the pathogenesis of SARS-CoV. In this study, we show that SARS-CoV-infected aged macaques developed significantly more pathology than young adult animals, which could not be contributed to differences in viral replication. Using comparative microarray analyses, it was shown that although the nature of the host response to SARS-CoV infection was similar in aged and young adult macaques, the severity was significantly different, with aged macaques displaying an increase in differential expression of genes associated with inflammation. Interestingly, type I IFN-β mRNA levels correlated negatively with gross pathology. Therapeutic treatment of aged macaques with type I IFN reduced pathology without affecting virus replication. However, pro-inflammatory gene expression was significantly diminished. Thus, modulation of the host response by type I IFNs provides a promising outlook for novel intervention strategies.
Collapse
|
18
|
Mannie MD, Abbott DJ, Blanchfield JL. Experimental autoimmune encephalomyelitis in Lewis rats: IFN-beta acts as a tolerogenic adjuvant for induction of neuroantigen-dependent tolerance. THE JOURNAL OF IMMUNOLOGY 2009; 182:5331-41. [PMID: 19380780 DOI: 10.4049/jimmunol.0803756] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytokine-Ag fusion proteins represent a novel approach for induction of Ag-specific tolerance and may constitute an efficient therapy for autoimmune disease. This study addressed whether a fusion protein containing rat IFN-beta and the encephalitogenic 73-87 determinant of myelin basic protein (i.e., the neuroantigen, or NAg) could prevent or treat experimental autoimmune encephalomyelitis (EAE) in Lewis rats. The optimal structure of the fusion protein was comprised of the rat IFN-beta cytokine as the N-terminal domain with an enterokinase (EK) linker to the NAg domain. Both cytokine and NAg domains had full biological activity. Subcutaneous administration of 1 nmol of IFNbeta-NAg fusion protein in saline on days -21, -14, and -7 before encephalitogenic challenge on day 0 resulted in a substantial attenuation of EAE. In contrast, administration of IFN-beta or NAg alone did not affect susceptibility to EAE. The covalent attachment of IFN-beta and NAg was not necessary, because separate injections of IFN-beta and NAg at adjacent sites were as effective as injection of IFNbeta-NAg for prevention of disease. When treatment was initiated after disease onset, the rank order of inhibitory activity was as follows: the IFNbeta-NAg fusion protein > or = a mixture of IFN-beta plus NAg > IFN-beta > NAg. The novel finding that IFN-beta acts as a tolerogenic adjuvant as well as a tolerogenic fusion partner may have significance for development of tolerogenic vaccines.
Collapse
Affiliation(s)
- Mark D Mannie
- Department of Microbiology and Immunology, East Carolina University, Brody School of Medicine, Greenville, NC 27834, USA
| | | | | |
Collapse
|
19
|
Abstract
This article analyzes the conceptual and technological context in which, over a period of 50 years, exploration of the biological and clinical significance of type I interferon has evolved. The elaboration of techniques for production and purification of mouse and human interferons and the establishment of laboratory-size production units have been of crucial importance in this process. Animal experiments have been invaluable for elucidation of mechanisms underlying the in vivo antiviral, anti-tumour and immunomodulatory potential of interferon, but have been of limited help to define the areas of clinical applicability. Proof of principle for applications as they are established today has come from clinical trials performed quite independently of evidence from animal experiments.
Collapse
Affiliation(s)
- Alfons Billiau
- Rega Institute, University of Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| |
Collapse
|
20
|
Barca O, Seoane M, Ferré S, Prieto JM, Lema M, Señarís R, Arce VM. Mechanisms of interferon-beta-induced survival in fetal and neonatal primary astrocytes. Neuroimmunomodulation 2007; 14:39-45. [PMID: 17700039 DOI: 10.1159/000107287] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2006] [Accepted: 02/25/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We have previously shown that interferon-beta (IFN-beta) is a potent promoter of astrocyte survival. Although the mechanism(s) by which IFN-beta promotes astrocyte survival have not been completely elucidated, it has been shown that IFN-beta directly stimulates survival signaling pathways. In the present report, we took advantage of the differences in the susceptibility of fetal and neonatal astrocytes to apoptosis to further investigate the mechanism(s) underlying the antiapoptotic effect of IFN-beta. METHODS Primary monolayer cultures of cortical astrocytes were established from neonatal (3- to 6-day-old) or fetal (embryonic days: E15 or E17) Sprague-Dawley rat cerebral cortices. Apoptotic cell death was determined by fluorescent-microscopic analysis of staining patterns of cell DNA with Hoechst 33258, and determination of annexin V binding.Akt phosphorylation was detected by Western blottingusing a commercial kit that allows specific recognition of both non-phosphorylated and serine-phosphorylated Akt. RESULTS In the present work, we have found that primary astrocytes obtained from neonatal rats are resistant to apoptosis induced by serum starvation, though cell death may be induced by combining serum starvation with sodium butyrate treatment. This effect is counteracted by IFN-beta treatment through a mechanism that involves phosphatidylinositol 3-kinase stimulation. CONCLUSIONS IFN-beta can be considered as a neuroprotective agent and, therefore, part of its beneficial effects in multiple sclerosis (MS) treatment may depend on its capacity to protect astrocytes against the apoptotic cell death that occurs in the course of the MS lesions.
Collapse
Affiliation(s)
- Olga Barca
- Departamento de Fisioloxía, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | | | | | | | |
Collapse
|
21
|
Billiau A. Anti-inflammatory properties of Type I interferons. Antiviral Res 2006; 71:108-16. [PMID: 16626815 PMCID: PMC7114336 DOI: 10.1016/j.antiviral.2006.03.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 03/13/2006] [Accepted: 03/14/2006] [Indexed: 01/22/2023]
Abstract
The notion that Type I interferons (interferon-alpha and -beta) possess anti-inflammatory potential is supported by data from clinical application in multiple sclerosis, by studies on cultured immune-competent cells and by investigation of experimental diseases in whole animals. These observations deserve the attention of virologists for their potential role in the pathogenesis and clinical management of virus infections.
Collapse
Affiliation(s)
- Alfons Billiau
- Rega Institute, University of Leuven, Minderbroedersstraat 10, BE-3000 Leuven, Belgium.
| |
Collapse
|
22
|
Combination therapy with glatiramer acetate (copolymer-1) and a type I interferon (IFN-?) does not improve experimental autoimmune encephalomyelitis. Ann Neurol 2001. [DOI: 10.1002/1531-8249(200001)47:1<127::aid-ana22>3.0.co;2-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
23
|
Jacobs L, Rudick R, Simon J. Extended observations on MS patients treated with IM interferon-beta1a (Avonex): implications for modern MS trials and therapeutics. J Neuroimmunol 2000; 107:167-73. [PMID: 10854652 DOI: 10.1016/s0165-5728(00)00232-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Extended observations of the pivotal phase III clinical trial of interferon-beta1a (IFNbeta1a; Avonex, Biogen) in relapsing MS patients revealed that: (1) active treatment significantly slowed the accumulation of physical disability over time, reduced clinical exacerbations and MRI brain lesions; (2) clinical efficacy did not depend on disability endpoints; (3) active treatment benefited multiple MRI measures of brain lesions; (4) cerebral atrophy occurred over 2 years in relatively mildly disabled patients; and (5) Avonex could slow the development of atrophy after the first year of treatment. Data from this study were recently used to design a new outcome measure for MS clinical trials (the Multiple Sclerosis Functional Composite), and was also the basis for two ongoing studies of IFNbeta1a: one in patients with monosymptomatic onset of MS and the other in secondary progressive MS.
Collapse
Affiliation(s)
- L Jacobs
- Department of Neurology, State University of New York at Buffalo, Buffalo General Hospital, 100 High Street, Buffalo, NY 14203, USA.
| | | | | |
Collapse
|
24
|
Uhm JH, Dooley NP, Stuve O, Francis GS, Duquette P, Antel JP, Yong VW. Migratory behavior of lymphocytes isolated from multiple sclerosis patients: effects of interferon beta-1b therapy. Ann Neurol 1999; 46:319-24. [PMID: 10482262 DOI: 10.1002/1531-8249(199909)46:3<319::aid-ana7>3.0.co;2-n] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Previous reports by our group and by others have shown that in vitro treatment of T cells derived from healthy, normal subjects with interferon beta-1b (IFN-beta1b) reduces metalloproteinase (metalloproteinase type 9 [MMP-9]) activity with a consequent reduction in lymphocyte migration. In this study, we used a Boyden chamber assay to assess the migratory capacity of T cells derived from multiple sclerosis patients who either did or did not receive IFN-beta1b. Lymphocytes derived from patients treated for less than 2 years with IFN-beta migrated at a low rate that was indistinguishable from that of cells isolated from healthy donors. However, longer term treatment with IFN (>3.5 years) was associated with a reversion of the migration to a high level that did not differ statistically from that of cells isolated from untreated multiple sclerosis patients. For both high-migratory groups, migration could be reduced to control levels after the exogenous addition of TIMP-1, a relatively specific inhibitor of the MMP-9, implicating this protease in the process of T-cell migration. Our findings suggest that one of the mechanisms by which IFN-beta exerts its action is by reducing MMP-9 activity and thus the entry of inflammatory cells into the nervous system and, as such, MMPs may constitute potential therapeutic targets in inflammatory diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- J H Uhm
- Department of Neuroimmunology, Montreal Neurological Hospital and Institute, McGill University, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Yasuda CL, Al-Sabbagh A, Oliveira EC, Diaz-Bardales BM, Garcia AA, Santos LM. Interferon beta modulates experimental autoimmune encephalomyelitis by altering the pattern of cytokine secretion. Immunol Invest 1999; 28:115-26. [PMID: 10484677 DOI: 10.3109/08820139909061141] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The mechanism of action underlying the beneficial effect of IFNbeta in Multiple Sclerosis is poorly understood. Experimental Autoimmune Encephalomyelitis (EAE) is the experimental model for Multiple Sclerosis; therefore, we investigated the effects of recombinant mouse IFNbeta on the severity of EAE induced in SJL mice and on cytokine production by Th1 and Th2 lymphocytes. The results indicated that rmIFN beta reduced the disease activity with an I.P. dosage of 10,000 U/day every other day, and successfully treated EAE mice revealed reduced amounts of IFN gamma; no changes in the levels of IL4 were observed, although thera was a significant increase in IL10 and TGFbeta production. Beneficial effects on EAE are associated with inhibition of inflammatory cytokines and stimulation of anti-inflammatory cytokines.
Collapse
Affiliation(s)
- C L Yasuda
- Department of Microbiology and Immunology, University of Campinas--UNICAMP, SP, Brazil
| | | | | | | | | | | |
Collapse
|
26
|
Okada AA, Keino H, Fukai T, Sakai J, Usui M, Mizuguchi J. Effect of type I interferon on experimental autoimmune uveoretinitis in rats. Ocul Immunol Inflamm 1998; 6:215-26. [PMID: 9924918 DOI: 10.1076/ocii.6.4.215.4024] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In order to establish a scientific basis for the potential use of type I interferons (IFNs) in clinical uveitis, we examined the effect of a preparation of IFN-alpha/beta on experimental autoimmune uveoretinitis (EAU). Lewis rats were immunized with bovine interphotoreceptor retinoid-binding protein (IRBP) and given daily intramuscular injections of 10(5) IU mouse natural IFN-alpha/beta. Intraocular inflammation was assessed by slit-lamp biomicroscopy and histopathological examination. Rats treated daily with IFN-alpha/beta starting on the day of IRBP immunization showed decreased intraocular inflammation as well as a slight delay in onset of inflammation when compared to control rats. This effect was also observed to a lesser extent in rats treated during either the induction phase of EAU only, or starting immediately after the onset of inflammation in the effector phase of EAU. Measurement of IRBP-stimulated splenocyte proliferation and serum anti-IRBP antibody subtypes did not reveal a significant difference between IFN-alpha/beta-treated rats and control rats. Measurement of cytokine production by IRBP-stimulated splenocytes in vitro showed significantly decreased TNF-alpha for IFN-alpha/beta-treated rats compared to control, but no difference for IFN-gamma, IL-2, IL-4, and IL-10. These results indicate that systemic administration of IFN-alpha/beta suppresses IRBP-induced EAU in rats, and suggest that such suppression may be mediated in part by a reduction in TNF-alpha production.
Collapse
Affiliation(s)
- A A Okada
- Department of Ophthalmology, Tokyo Medical College Hospital, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Cowden WB, Cullen FA, Staykova MA, Willenborg DO. Nitric oxide is a potential down-regulating molecule in autoimmune disease: inhibition of nitric oxide production renders PVG rats highly susceptible to EAE. J Neuroimmunol 1998; 88:1-8. [PMID: 9688317 DOI: 10.1016/s0165-5728(98)00040-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Rat strains vary in their susceptibility to experimental autoimmune encephalomyelitis (EAE) and in many cases, factors other than MHC antigens are thought to play a role in this. We found that PVG rats, which have a very low susceptibility to EAE, were rendered highly susceptible to clinical disease when treated with N-methylarginine (NMA) an inhibitor of nitric oxide synthase (NOS). The clinical course of the ensuing disease in NMA-treated PVG rats was in most cases fulminating in nature and accompanied by some mortality. Following immunisation with myelin basic protein (MBP)-complete Freund's adjuvant (CFA), PVG rats developed higher serum levels of the surrogate markers of nitric oxide production, reactive nitrogen intermediates (RNI; nitrite and nitrate), than did their Lewis counterparts. This in vivo finding was reflected in vitro, where the levels of RNI produced in 24, 48 and 72 h IFN-gamma-stimulated spleen cell cultures for PVG rats were significantly higher than those for Lewis rats. A mechanism by which increased NO production might protect PVG rats against clinical EAE was suggested by the finding that lymph node cells, isolated from NMA-treated MBP-immunised PVG rats, proliferated in response to MBP at a rate approximately 3 x greater than those from MBP-immunised, saline treated rats. Thus, the greater number of MBP-specific T cells generated in the NOS inhibitor-treated vs. untreated rats could account for their increased susceptibility to developing clinical EAE. The findings in this study suggest that NO plays a role in protecting PVG rats against developing EAE.
Collapse
MESH Headings
- Animals
- Cell Division/drug effects
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Enzyme Inhibitors/pharmacology
- Genetic Predisposition to Disease
- Interferon-gamma/pharmacology
- Lymph Nodes/cytology
- Lymph Nodes/drug effects
- Myelin Basic Protein/pharmacology
- Nitrates/blood
- Nitric Oxide/antagonists & inhibitors
- Nitric Oxide/physiology
- Nitric Oxide Synthase/antagonists & inhibitors
- Nitrites/blood
- Rats
- Rats, Inbred Lew
- Rats, Mutant Strains/genetics
- Rats, Mutant Strains/physiology
- Spleen/cytology
- Spleen/drug effects
- Spleen/metabolism
- omega-N-Methylarginine/pharmacology
Collapse
Affiliation(s)
- W B Cowden
- Division of Immunology and Cell Biology, The John Curtin School of Medical Research, The Australian National University, Canberra ACT.
| | | | | | | |
Collapse
|
28
|
van der Meide PH, de Labie MC, Ruuls SR, Groenestein RJ, Botman CA, Olsson T, Dijkstra CD. Discontinuation of treatment with IFN-beta leads to exacerbation of experimental autoimmune encephalomyelitis in Lewis rats. Rapid reversal of the antiproliferative activity of IFN-beta and excessive expansion of autoreactive T cells as disease promoting mechanisms. J Neuroimmunol 1998; 84:14-23. [PMID: 9600704 DOI: 10.1016/s0165-5728(97)00207-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
IFN-beta has recently been shown to exert remarkable beneficial effects on disease development in patients with early stage relapsing-remitting MS. The specific immune mechanism(s) by which IFN-beta ameliorates this human demyelinating disease is at present undefined. One potential mechanism may reside in the antiproliferative activity of IFN-beta which may inhibit the expansion of autoaggressive T cells thereby limiting disease progression. In the present study we investigated whether the administration of recombinant rat IFN-beta (rrIFN-beta) to Lewis rats with actively induced experimental autoimmune encephalomyelitis (EAE) inhibits the expansion of encephalitogenic T cells in lymphoid organs and as such may contribute to suppression of disease activity in this widely used animal model for MS. Our data show that daily administrations of > or = 3 x 10(5) u rrIFN-beta to EAE rats, starting two days before MBP sensitization and continued for 10 days led to a dramatic and dose-dependent reduction in encephalitogenic T cells in both spleen and inguinal lymph nodes at day 8 post-immunization (p.i.). However, the rrIFN-beta-mediated reduction in effector T cells did not ameliorate paralytic disease as expected but significantly enhanced the severity of EAE. Analyses of lymphoid organs in the remission phase of EAE revealed strongly elevated numbers of encephalitogenic T cells in rrIFN-beta-treated versus control rats suggesting a rapid reversal of the antiproliferative action of rrIFN-beta followed by an overshoot in the subsequent expansion of these effector T cells. In conformity with higher numbers of encephalitogenic T cells and worsening of disease, animals also showed significantly greater perivascular inflammation in the CNS. The relevance of our findings in relation to the beneficial effects of IFN-beta in MS is discussed.
Collapse
Affiliation(s)
- P H van der Meide
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Interferons were initially characterized for their ability to 'interfere' with viral replication, slow cell proliferation, and profoundly alter immunity. They are a group of hormone-like molecules synthesized and secreted by macrophages, monocytes, T lymphocytes, glia, and neurons. These cytokines have been shown to have several regulatory roles and diverse biological activities, including control of cellular and humoral immune responses, inflammation, and tumor regression. In addition, there are many reports indicating that interferon-alpha (IFN-alpha) participates in the regulation of various cellular and humoral processes such as the endocrine system modulates behavior, brain activity, temperature, glucose sensitive neurons, feeding pattern and opiate activity. Therefore, IFN-alpha can be considered as a physiological modulator, with only one of its functions being the ability to hinder viral replication intracellularly.
Collapse
Affiliation(s)
- N Dafny
- Department of Neurobiology and Anatomy, The University of Texas Medical School at Houston, 77225, USA
| |
Collapse
|
30
|
Ruuls SR, Sedgwick JD. Cytokine-directed therapies in multiple sclerosis and experimental autoimmune encephalomyelitis. Immunol Cell Biol 1998; 76:65-73. [PMID: 9553778 DOI: 10.1046/j.1440-1711.1998.00715.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Secreted and cell surface molecules that control the nature of immune responses or directly mediate tissue damage are important targets for therapeutic intervention of autoimmune inflammatory disorders. The precise aetiology of multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system is unknown and therapeutic interventions for this disease are limited. However, as it is believed that MS is autoimmune in nature, it seems likely that targeting of the immune system, either to deviate it down a more benign pathway or to directly block tissue damaging effector mechanisms, may enable this disease to be controlled. In this overview we examine three alternative approaches to therapy of MS: cytokine-directed immune deviation, immunoregulation by type I interferons and blockade of TNF. Underlying rationales for the application of these approaches to the treatment of MS are based largely on studies in the disease model experimental autoimmune encephalomyelitis. These studies are reviewed.
Collapse
Affiliation(s)
- S R Ruuls
- Centenary Institute of Cancer Medicine and Cell Biology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | |
Collapse
|
31
|
Abstract
In this review we delineate the rationale for immunotherapy in multiple sclerosis and describe the various levels at which immune intervention, according to a modern model of the immune system organization, is feasible. Current and future immunosuppressive and immunomodulating therapeutic approaches at the level of antigen presentation and at the lymphocyte and cytokine network levels are discussed.
Collapse
Affiliation(s)
- D M Karussis
- Department of Neurology, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | | |
Collapse
|
32
|
Stuve O, Chabot S, Jung SS, Williams G, Yong VW. Chemokine-enhanced migration of human peripheral blood mononuclear cells is antagonized by interferon beta-1b through an effect on matrix metalloproteinase-9. J Neuroimmunol 1997; 80:38-46. [PMID: 9413258 DOI: 10.1016/s0165-5728(97)00134-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The increased migration of peripheral blood mononuclear cells (PBMNCs) across a fibronectin (FN) matrix in response to the chemokines RANTES, MIP-1 alpha and MCP-1 is antagonized by interferon-beta-1b (IFN beta-1b). MCP-1 treatment of PBMNCs elevates their mRNA level and secretion of a matrix degrading enzyme, matrix metalloproteinase (MMP)-9, which is abrogated by IFN beta-1b. The clinical benefits of IFN beta-1b treatment in multiple sclerosis patients may in part be a result of this drug's ability to decrease the migration of PBMNCs in spite of a chemotactic gradient. Furthermore, the elevation of MMP-9 production by PBMNCs may be an important mechanism of action of chemokines.
Collapse
Affiliation(s)
- O Stuve
- Department of Neurology and Neurosurgery, McGill University, Montreal, Que., Canada
| | | | | | | | | |
Collapse
|
33
|
Abstract
Interferon-beta (IFN-beta) is the first therapeutic intervention shown to alter the natural history of multiple sclerosis (MS), a relapsing then progressive inflammatory degenerative disease of the CNS. Since publication of the first randomized placebo-controlled trial of IFN-beta, and subsequent acquisition of US and European product licences for use in relapsing-remitting MS, the hopes and expectations of patients have been elevated greatly only to be dampened as more critical analysis of the trial results, in conjunction with the cost of treatment, led to marked limitations on prescription in several countries. IFN-beta is not a cure. Here we review what is known about the mechanisms of action of IFN-beta in demyelinating disease, and propose a possible model of action of IFN-beta in the treatment of MS.
Collapse
Affiliation(s)
- G L Hall
- University of Cambridge Neurology Unit, Addenbrooke's Hospital, UK
| | | | | |
Collapse
|
34
|
Stüve O, Dooley NP, Uhm JH, Antel JP, Francis GS, Williams G, Yong VW. Interferon beta-1b decreases the migration of T lymphocytes in vitro: effects on matrix metalloproteinase-9. Ann Neurol 1996; 40:853-63. [PMID: 9007090 DOI: 10.1002/ana.410400607] [Citation(s) in RCA: 274] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In multiple sclerosis (MS), the influx of activated T lymphocytes into the brain parenchyma leads to the subsequent damage of oligodendrocytes, the cells that produce central nervous system (CNS) myelin. We report here that interferon beta-1b (IFNbeta-1b), a drug shown to be efficacious in the treatment of patients with MS, decreases the in vitro migration of activated T lymphocytes through fibronectin (FN), a major component of the basement membrane that surrounds cerebral endothelium. At 1,000 IU/ml, IFNbeta-1b reduced the migratory rate to that of unactivated T cells. In contrast, IFNgamma at 1,000 IU/ml, which caused a similar decrease (25%) in the proliferation rate of T lymphocytes as IFNbeta-1b, did not affect migration. All T-lymphocyte subsets and natural killer (NK) cells were demonstrated by flow cytometry to be equally affected by IFNbeta-1b treatment. 125I-Western blot analyses revealed that IFNbeta-1b treatment resulted in a marked reduction of the ability of T cells to cleave FN. The substrate-degrading capability of T lymphocytes was shown to be due predominantly to the activity of a 92-kd matrix metalloproteinase, MMP-9, whose levels were decreased by IFNbeta-1b. We suggest that the clinical benefits of IFNbeta-1b treatment in MS patients may be in part a result of the ability of this drug to significantly decrease MMP-9 activity, leading to a reduction of T-lymphocyte infiltration into the CNS.
Collapse
Affiliation(s)
- O Stüve
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
35
|
Nelson PA, Akselband Y, Dearborn SM, Al-Sabbagh A, Tian ZJ, Gonnella PA, Zamvil SS, Chen Y, Weiner HL. Effect of oral beta interferon on subsequent immune responsiveness. Ann N Y Acad Sci 1996; 778:145-55. [PMID: 8610968 DOI: 10.1111/j.1749-6632.1996.tb21123.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Oral administration of myelin antigens reduces the incidence and severity of EAE in rat and mouse models and decreases the frequency of MBP-reactive cells and the frequency of attacks in some patients with multiple sclerosis. Low-dose oral tolerance has been shown to be mediated by Th2-type regulatory cells that secrete TGFbeta and IL-4/IL-10. Adjuvants and cytokines may modulate oral tolerance. The addition of betaIFN to the experimental therapy regimen, either orally or by intraperitoneal injection, has been shown to enhance the suppressive effects of oral myelin antigens when either are fed the suboptimal dosing regimen to suppress EAE. The current studies were conducted to elucidate the mechanism of the observed in vivo synergy of betaIFN and antigen feeding. Analysis of the in vitro proliferative response and cytokine production by lymphocytes from fed animals in response to specific antigen in culture shows that the synergistic effect may be related to both independent suppression of the immune response by oral betaIFN and enhanced production of TGFbeta and IL-4/IL-10. There was an unexpected increase in the production of gammaIFN by lymphocytes in vitro after three doses of oral betaIFN in vivo. These observations have important implications for the use of cytokines to modulate oral tolerance.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antigens/administration & dosage
- Antigens/immunology
- Cattle
- Cells, Cultured
- Crosses, Genetic
- Cytokines/biosynthesis
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Female
- Guinea Pigs
- Immune Tolerance
- Injections, Intraperitoneal
- Interferon Type I/administration & dosage
- Interferon Type I/pharmacology
- Interferon-beta/administration & dosage
- Interferon-beta/pharmacology
- Lymphocyte Activation
- Mice
- Mice, Inbred Strains
- Mycobacterium tuberculosis/immunology
- Myelin Basic Protein/administration & dosage
- Myelin Basic Protein/immunology
- Ovalbumin/immunology
- Rats
- Rats, Inbred Lew
Collapse
Affiliation(s)
- P A Nelson
- AutoImmune Inc., Lexington, Massachusetts 02173, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Vriesendorp FJ, Flynn RE, Khan M, Pappolla MA, Brod SA. Oral administration of type I interferon modulates the course of experimental allergic neuritis. Autoimmunity 1996; 24:157-65. [PMID: 9020408 DOI: 10.3109/08916939608995361] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We investigated the effect of oral administration of type I interferon (IFN) in experimental allergic neuritis (EAN) in Lewis rats immunized with bovine peripheral nerve myelin. Starting at 7 days preceding immunization, rats were fed daily until sacrifice either with 5000 U rat IFN-alpha/beta or mock-IFN. The clinical severity of EAN was significantly reduced in IFN-alpha/beta fed animals compared to mock-IFN fed controls. Demyelination, but not inflammation, was decreased in IFN-alpha/beta fed compared to mock-IFN fed rats at day 20 after immunization. In situ IFN-gamma production and inflammation were reduced when evaluated by immunocytochemistry at day 13 after immunization. Spleen cells from IFN-alpha/beta fed compared to mock-IFN fed EAN rats showed significantly reduced proliferation to stimulation with Con A or peripheral nerve myelin. IFN-gamma production in draining lymph node cells was significantly reduced after stimulation with bovine peripheral nerve myelin. Our data suggest that oral administration of IFN-alpha/beta reduces the severity of EAN, possibly by a reduction in IFN-gamma production.
Collapse
Affiliation(s)
- F J Vriesendorp
- Department of Neurology, University of Texas Health Science Center-Houston 77030, USA
| | | | | | | | | |
Collapse
|
37
|
Arnason BG, Dayal A, Qu ZX, Jensen MA, Genç K, Reder AT. Mechanisms of action of interferon-beta in multiple sclerosis. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 1996; 18:125-48. [PMID: 8984676 DOI: 10.1007/bf00792613] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- B G Arnason
- Department of Neurology and the Brain Research Institute, University of Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
38
|
Yu M, Nishiyama A, Trapp BD, Tuohy VK. Interferon-beta inhibits progression of relapsing-remitting experimental autoimmune encephalomyelitis. J Neuroimmunol 1996; 64:91-100. [PMID: 8598395 DOI: 10.1016/0165-5728(95)00160-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The results of two phase III clinical trials have recently shown that interferon-beta (IFNbeta) is effective in the treatment of relapsing-remitting multiple sclerosis (RRMS). Treatment with IFNbeta results in a significant decrease in the rate of clinical relapse and a marked delay in progression to disability compared to placebo-treated control patients. In the present study, we demonstrate similar therapeutic effects after treating (SWR X SJL)F1 mice with IFNbeta at the onset of clinical signs of experimental autoimmune encephalomyelitis (EAE), a disease animal model widely used in MS studies. EAE was actively induced by immunization of (SWR X SJL)F1 mice with the immunodominant encephalitogenic peptide 139-151 of myelin proteolipid protein (PLP). In blinded testing, mice treated with IFNbeta at EAE onset showed a delay in progression to clinical disability as determined by marked improvement with time in mean clinical score, significant delay in onset of relapse, and significant decrease in exacerbation frequency compared to placebo-treated control mice. The therapeutic effect of IFNbeta was accompanied by a significant inhibition of delayed-type hypersensitivity (DTH) but not proliferation in response to the priming PLP 139-151. In addition, IFNbeta treatment resulted in an overall decrease in severity of both inflammation and demyelination in the central nervous system. These results mimic in an autoimmune animal model the effectiveness of IFNbeta treatment observed in MS. Moreover, our study suggests that anti-viral properties of IFNbeta are not essential for producing therapeutic effects in autoimmune demyelinating disease, and that the efficacy of IFNbeta in the treatment of MS may be due to inhibition of autoreactivity.
Collapse
Affiliation(s)
- M Yu
- Department of Immunology, FFb-1, Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
39
|
Brod SA, Scott M, Burns DK, Phillips JT. Modification of acute experimental autoimmune encephalomyelitis in the Lewis rat by oral administration of type 1 interferons. J Interferon Cytokine Res 1995; 15:115-22. [PMID: 8590314 DOI: 10.1089/jir.1995.15.115] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The effect of orally administered type 1 interferons on the severity of acute experimental autoimmune encephalomyelitis (EAE), a T cell-mediated autoimmune disease, was examined by inoculation of Lewis rats with guinea pig myelin basic protein (GPMBP) and complete Freund's adjuvant. Rats were fed either rat species-specific or human recombinant type 1 interferon (IFN) or mock IFN daily for 7 days preceding immunization and for 21 days thereafter. There was a significant decrease in the clinical score and inflammatory foci in animals fed 5000 units IFN compared with mock-treated animals. There was a significant decrease in clinical score and number of inflammatory foci in spinal cord in animals fed orally 5000 units human recombinant IFN-alpha PO compared with SC 5000 units recombinant human IFN-alpha. Oral administration of type 1 interferon, as opposed to subcutaneous administration, inhibited the secretion of IFN-gamma from ConA-activated draining popliteal lymph node cells compared with mock-fed animals. These experiments demonstrate that acute EAE is more effectively inhibited by equivalent amounts of orally in contrast to parenterally administered IFN-alpha. These results suggest that type 1 IFNs are active by the oral route and have significant clinical and histologic effects in acute autoimmune disease.
Collapse
Affiliation(s)
- S A Brod
- Department of Neurology, University of Texas Health Science Center at Houston 77030, USA
| | | | | | | |
Collapse
|
40
|
Duong TT, Finkelman FD, Singh B, Strejan GH. Effect of anti-interferon-gamma monoclonal antibody treatment on the development of experimental allergic encephalomyelitis in resistant mouse strains. J Neuroimmunol 1994; 53:101-7. [PMID: 8051292 DOI: 10.1016/0165-5728(94)90069-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Immunization with myelin basic protein (MBP) in complete Freund's adjuvant failed to induce experimental allergic encephalomyelitis (EAE) in six resistant mouse strains studied: A/J, BALB/c C3H/HeJ, AKR, NZW and DBA/2. However, treatment of challenged mice with anti-interferon-gamma (IFN-gamma) monoclonal antibody (mAb) induced severe EAE in mice of all strains except AKR. Furthermore, anti-IFN-gamma mAb treatment led to increased disease incidence and severity in BALB/c mice challenged with the MBP peptide87-103, known to be encephalitogenic for the susceptible SJL strain. In three strains tested, anti-IFN-gamma mAb enhanced passively induced EAE in the A/J and C3H/HeJ but not in the BALB/c mice. All mice with clinically overt EAE had widespread histological lesions characterized by mononuclear cell infiltrates and focal demyelination. The results indicate that resistant strains are genetically capable of developing EAE, and that IFN-gamma can contribute to disease resistance.
Collapse
Affiliation(s)
- T T Duong
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada
| | | | | | | |
Collapse
|
41
|
Abstract
Although multiple sclerosis (MS) is generally believed to be an immune-mediated disease, conventional therapy with ACTH, corticosteroids, or immunosuppressive drugs is unsatisfactory. Aside from their unpredictable therapeutic effects, these agents are potentially hazardous and can only be given for short periods of time. There is an urgent need for less toxic yet effective immunotherapy, that that can be administered early in the disease and continued indefinitely. Clinical trials of the interferons (IFNs) have not only led to a promising new approach to treatment, but have also stimulated basic research in the immunological mechanisms of underlying disease activity. Administration of IFN-gamma promotes exacerbations of MS, whereas recombinant IFN-beta has been shown, in controlled clinical trials, to suppress them. Other ongoing studies are likely to provide further information about its long-term therapeutic value. More importantly, laboratory studies performed in conjunction with these clinical trials have provided fresh insights into the pathogenesis of MS by revealing immunoregulatory mechanisms in which endogenous IFN-gamma, TNF-alpha, and other cytokines appear to play central roles. The 'Decade of the Brain' may therefore see answers both to the therapeutic dilemma of MS, and to more basic questions about the function of IFNs and other cytokines in activation and regulation of the disease process.
Collapse
Affiliation(s)
- H S Panitch
- Neurology Service, Baltimore VA Medical Center, MD
| | | |
Collapse
|
42
|
Duong TT, Finkelman FD, Strejan GH. Effect of interferon-gamma on myelin basic protein-specific T cell line proliferation in response to antigen-pulsed accessory cells. Cell Immunol 1992; 145:311-23. [PMID: 1280534 DOI: 10.1016/0008-8749(92)90334-l] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We have shown previously that treatment of SJL/J mice with anti-interferon-gamma monoclonal antibody (mAb) exacerbated experimental allergic encephalomyelitis (EAE) only if administered at the time of encephalitogenic challenge. Here we investigate the role of interferon-gamma (IFN-gamma) and anti-IFN-gamma mAb in the early events of T cell activation in vitro. Pretreatment of murine peritoneal exudate cells (PEC) with IFN-gamma led to a significant increase in their ability to activate myelin basic protein (MBP)-specific, short-term T cell lines. When exogenous IFN-gamma was added to cocultures of T cells and MBP-pulsed PEC, the antigen-specific T cell proliferation was considerably reduced. Anti-IFN-gamma mAb added to these cultures neutralized the inhibitory effect of the exogenous IFN-gamma on T cell proliferation but had no visible effect on class II MHC expression by the antigen-pulsed PEC present in the same cultures. A reduction in T cell proliferation was also observed when the T cells were treated with IFN-gamma prior to coculture with the MBP-pulsed PEC. These results demonstrate that, on one hand, IFN-gamma enhances the ability of PEC to induce antigen-specific T cell proliferation but, on the other hand, acts on the T cells themselves by inhibiting their proliferation in response to the antigen-pulsed PEC. This may explain why treatment with anti-IFN-gamma antibody in vivo induces EAE exacerbation.
Collapse
Affiliation(s)
- T T Duong
- Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Canada
| | | | | |
Collapse
|
43
|
Nakano T, Awaki E, Araga S, Takai H, Inoue K, Takahashi K. Recurrent herpes zoster myelitis treated with human interferon alpha: a case report. Acta Neurol Scand 1992; 85:372-5. [PMID: 1320320 DOI: 10.1111/j.1600-0404.1992.tb04064.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recurrent herpes zoster myelitis is very rare. However, a case was recently observed in our hospital. A 43-year-old woman developed myelitis 2 weeks after development of shingles. Her condition was improved by methylprednisolone. Seven months later, she developed myelitis after development of shingles again. Antibody against varicella-zoster (VZV), increased in the serum, but was negative in the cerebrospinal fluid. Methylprednisolone was not sufficiently effective against this attack. The refractory sensory disturbance was improved by human interferon alpha (IFN-alpha). Natural killer cell activity, the helper T-cell/suppressor T-cell ratio and the kappa/lambda ratio of B-cells increased with clinical improvement. In this case, delayed-type hypersensitivity after VZV infection played a role in the occurrence of myelopathy and clinical improvement resulted from the immunosuppressive effects of IFN-alpha.
Collapse
Affiliation(s)
- T Nakano
- Division of Neurology, Tottori University, Yonago, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Duong TT, St Louis J, Gilbert JJ, Finkelman FD, Strejan GH. Effect of anti-interferon-gamma and anti-interleukin-2 monoclonal antibody treatment on the development of actively and passively induced experimental allergic encephalomyelitis in the SJL/J mouse. J Neuroimmunol 1992; 36:105-15. [PMID: 1732276 DOI: 10.1016/0165-5728(92)90042-j] [Citation(s) in RCA: 126] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SJL/J mice challenged with myelin basic protein (MBP) in complete Freund's adjuvant (CFA) developed only mild chronic-relapsing experimental allergic encephalomyelitis (EAE) with very low incidence. However, treatment of challenged mice with anti-interferon-gamma (IFN-gamma) monoclonal antibody (mAb) determined severe disease in all cases. Similarly, in passive EAE, the addition of anti-IFN-gamma to the in vitro MBP-activated cells at the time of transfer led to significant disease exacerbation in all recipients. The disease enhancing effect was observed only when the mAb was given at the time of active challenge or of passive transfer, but not at later times. Anti-interleukin-2 (IL-2) antibody had only a marginal effect in the active induction, but drastically reduced the manifestations of passive EAE, even when mixed with a disease-enhancing dose of anti-IFN-gamma. These findings support the notion that IL-2 is required for disease induction whereas IFN-gamma plays a disease-limiting role early in the development of EAE.
Collapse
Affiliation(s)
- T T Duong
- Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Canada
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Veljić J, Marić D, Janković BD. Changes of experimental allergic encephalomyelitis by methionine-enkephalin injected into lateral ventricles of the rat brain. Int J Neurosci 1991; 59:81-9. [PMID: 1774141 DOI: 10.3109/00207459108985451] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Our previous investigations have shown that the opioid peptide methionine-enkephalin (Met-Enk) modulates in vivo a variety of humoral and cell-mediated immune performances. In this study, rats bearing polyethylene cannulae permanently inserted into the lateral ventricles of the brain were used. Experimental allergic encephalomyelitis (EAE) was induced with guinea pig spinal cord in complete Freund's adjuvant injected into the left hind foot pad. The following groups of cannulated rats were tested: nontreated with Met-Enk or saline, intracerebroventricularly (i.c.v.) injected with saline, and i.c.v. treated with low (1 microgram/kg) dose of Met-Enk and high (1 mg/kg) dose of Met-Enk. Intact noncannulated rats sensitized for EAE served as an additional control. The results showed that i.c.v. treatment with 1 microgram/kg of Met-Enk significantly increased the incidence and severity of EAE. In contrast, injections of 1 mg/kg of Met-Enk produced a moderate decline of clinical EAE, but marked diminution of inflammatory lesions in the brain. Interestingly, histopathology of EAE was more pronounced in control rats treated i.c.v. with saline. On the other hand, control cannulated rats noninjected with saline exhibited a striking decrease of neurological and histopathological signs of the disease, thus indicating a suppressive effect of stress (surgical procedure) on EAE. In conclusion, the present study showing the central effect of Met-Enk on EAE when peptide was applied in the cerebral cavity, and earlier studies which revealed the peripheral effect on EAE when Met-Enk was administered intraperitoneally, suggests that Met-Enk exerts its immunomodulatory action both centrally and peripherally.
Collapse
Affiliation(s)
- J Veljić
- Immunology Research Center, Belgrade, Yugoslavia
| | | | | |
Collapse
|
47
|
Janković BD, Marić D. Enkephalins and autoimmunity: differential effect of methionine-enkephalin on experimental allergic encephalomyelitis in Wistar and Lewis rats. J Neurosci Res 1987; 18:88-94. [PMID: 3500326 DOI: 10.1002/jnr.490180115] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To evaluate the immunomodulating activity of methionine-enkephalin (Met-Enk) and leucine-enkephalin (Leu-Enk) in the development of experimental allergic encephalomyelitis (EAE), two strains of rats, one highly susceptible to EAE (Lewis rats) and the other relatively resistant to the disease (Wistar rats) were used. The animals were given daily injections of either a high dose (5 mg/kg b.w.) or a low dose (0.2 mg/kg b.w.) of these opioid peptides, after receiving guinea pig spinal cord in combination with immunological adjuvants. A major conclusion from this study is that Met-Enk is a potent immunomodulator/regulator of the autoimmune disease, whereas Leu-Enk does not affect EAE. The high dose of 5 mg/kg b.w. of Met-Enk completely or significantly inhibited neurological signs and markedly diminished occurrence and intensity of histological lesions in the brain, and cervical, thoracic, and lumbar spinal cord. The loss of body weight, which accompanies EAE, was also prevented by a high dose of Met-Enk. In contrast, the low dose of 0.2 mg/kg b.w. of Met-Enk potentiated neurological and histopathological features of the disease. Nonimmunized rats injected with enkephalins for a period of 17 consecutive days showed neither neurological nor histological signs of EAE, nor signs of intoxication due to the application of enkephalins. Thus, these data establish a link between methionine-enkephalin and EAE, and suggest that this opioid pentapeptide might be important in the pathogenesis and prevention of the inflammatory autoimmune disease.
Collapse
Affiliation(s)
- B D Janković
- Immunology Research Center, Belgrade, Yugoslavia
| | | |
Collapse
|
48
|
Jacobs L, Salazar AM, Herndon R, Reese PA, Freeman A, Josefowicz R, Cuetter A, Husain F, Smith WA, Ekes R. Multicentre double-blind study of effect of intrathecally administered natural human fibroblast interferon on exacerbations of multiple sclerosis. Lancet 1986; 2:1411-3. [PMID: 2878272 PMCID: PMC7134842 DOI: 10.1016/s0140-6736(86)92730-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In this randomised, double-blind, placebo-controlled, 2-year multicentre study intrathecally administered natural human fibroblast interferon (IFN-B) was effective in reducing exacerbations of multiple sclerosis (MS) in patients with exacerbating/remitting disease. The mean reduction in exacerbation rate of 34 patients who received IFN-B (recipients) was significantly greater during the study than that of 35 patients who received placebo (p less than 0.04). The prestudy exacerbation rates were comparable in recipients and controls, but the rate at the end of the study was significantly lower in recipients than in controls (p less than 0.001). IFN-B was given by nine or ten lumbar punctures over the first 6 months of the study, and patient observations continued for 2 years. IFN-B was well tolerated in 95% of the recipients, and the side-effects experienced were clearly acceptable for the benefits achieved. Low doses of indomethacin reduced the toxicity of IFN-B and played an important role in successful double-blinding.
Collapse
|
49
|
Abreu SL, Thampoe I, Kaplan P. Interferon in experimental autoimmune encephalomyelitis: intraventricular administration. JOURNAL OF INTERFERON RESEARCH 1986; 6:627-32. [PMID: 2437223 DOI: 10.1089/jir.1986.6.627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Highly purified rat fibroblast-derived interferon (RfIFN), was administered intraventricularly to Lewis rats affected with experimental autoimmune encephalomyelitis (EAE). Its effects on both the active and passive forms of EAE were studied. A method was devised to deliver, via a syringe pump, RfIFN (sp. act. 10(8) U/mg) into the third ventricle of control and experimental animals. Rats with either the active form of EAE (produced by injection with myelin basic protein (MBP) in Freund's complete adjuvant) or with adoptively transferred EAE were treated with RfIFN either therapeutically and/or prophylactically. In no instance was a significant difference observed, on the course of EAE, between those animals receiving RfIFN and those receiving "mock" IFN. The pharmacokinetics of the infused RfIFN were also studied.
Collapse
|
50
|
Hertz F, Deghenghi R. Effect of rat and beta-human interferons on hyperacute experimental allergic encephalomyelitis in rats. AGENTS AND ACTIONS 1985; 16:397-403. [PMID: 2413742 DOI: 10.1007/bf01982879] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human interferon-beta (human IFN-beta) and rat interferon (rat IFN) were evaluated on experimental allergic encephalomyelitis (EAE) in rats, a delayed cellular reaction resembling human multiple sclerosis (MS). Rat IFN was active by intravenous and intracerebroventricular routes. It decreased the severity of clinical symptoms of paralysis during the 22 days of the assay. Human IFN-beta, on the contrary, had no effect when similarly tested in this rat model. Cyclophosphamide delayed the onset of paralysis, but levamisole enhanced the severity of the EAE.
Collapse
|