1
|
Giannessi F, Percario Z, Lombardi V, Sabatini A, Sacchi A, Lisi V, Battistini L, Borsellino G, Affabris E, Angelini DF. Macrophages treated with interferons induce different responses in lymphocytes via extracellular vesicles. iScience 2024; 27:109960. [PMID: 38832015 PMCID: PMC11144789 DOI: 10.1016/j.isci.2024.109960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/09/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024] Open
Abstract
Limited information exists regarding the impact of interferons (IFNs) on the information carried by extracellular vesicles (EVs). This study aimed at investigating whether IFN-α2b, IFN-β, IFN-γ, and IFN-λ1/2 modulate the content of EVs released by primary monocyte-derived macrophages (MDM). Small-EVs (sEVs) were purified by size exclusion chromatography from supernatants of MDM treated with IFNs. To characterize the concentration and dimensions of vesicles, nanoparticle tracking analysis was used. SEVs surface markers were examined by flow cytometry. IFN treatments induced a significant down-regulation of the exosomal markers CD9, CD63, and CD81 on sEVs, and a significant modulation of some adhesion molecules, major histocompatibility complexes and pro-coagulant proteins, suggesting IFNs influence biogenesis and shape the immunological asset of sEVs. SEVs released by IFN-stimulated MDM also impact lymphocyte function, showing significant modulation of lymphocyte activation and IL-17 release. Altogether, our results show that sEVs composition and activity are affected by IFN treatment of MDM.
Collapse
Affiliation(s)
- Flavia Giannessi
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Via Ardeatina 306-354, 00179 Rome, Italy
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Zulema Percario
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Valentina Lombardi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Andrea Sabatini
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Alessandra Sacchi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Veronica Lisi
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Luca Battistini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Via Ardeatina 306-354, 00179 Rome, Italy
| | - Giovanna Borsellino
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Via Ardeatina 306-354, 00179 Rome, Italy
| | - Elisabetta Affabris
- Laboratory of Molecular Virology and Antimicrobial Immunity, Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Daniela F. Angelini
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, Via Ardeatina 306-354, 00179 Rome, Italy
| |
Collapse
|
2
|
Soyfer EM, Fleischman AG. Myeloproliferative neoplasms - blurring the lines between cancer and chronic inflammatory disorder. Front Oncol 2023; 13:1208089. [PMID: 37361587 PMCID: PMC10288874 DOI: 10.3389/fonc.2023.1208089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Myeloproliferative Neoplasm (MPN) is a group of chronic blood cancers that arise from a hematopoietic stem cell (HSC) clone with somatic mutations causing constitutive activation of myeloid cytokine receptor signaling. In addition to elevated blood cell counts, MPN typically presents with increased inflammatory signaling and inflammation symptoms. Therefore, while being a clonally derived neoplasm, MPN has much in common with chronic non-cancerous inflammatory conditions, such as rheumatoid arthritis, lupus, and many more. MPN and chronic inflammatory disease (CID) share similar chronicity, symptoms, dependency on the immune system, environmental triggers, and treatments. Overall, we will highlight the similarities between an MPN and CID. We highlight that while MPN is classified as a cancer, its behavior is more aligned to that of a chronic inflammatory disease. We propose that MPN should inhabit a fluid/spectrum between auto-inflammatory disease and cancer.
Collapse
Affiliation(s)
- Eli M. Soyfer
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Angela G. Fleischman
- School of Medicine, University of California, Irvine, Irvine, CA, United States
- Division of Hematology/Oncology, University of California (UC) Irvine Health, Irvine, CA, United States
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
3
|
Albert C, Mikolajczak J, Liekfeld A, Piper SK, Scheel M, Zimmermann HG, Nowak C, Dörr J, Bellmann-Strobl J, Chien C, Brandt AU, Paul F, Hoffmann O. Fingolimod after a first unilateral episode of acute optic neuritis (MOVING) - preliminary results from a randomized, rater-blind, active-controlled, phase 2 trial. BMC Neurol 2020; 20:75. [PMID: 32126977 PMCID: PMC7052969 DOI: 10.1186/s12883-020-01645-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 02/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background Neuroprotection and promotion of remyelination represent important therapeutic gaps in multiple sclerosis (MS). Acute optic neuritis (ON) is a frequent MS manifestation. Based on the presence and properties of sphingosine-1-phosphate receptors (S1PR) on astrocytes and oligodendrocytes, we hypothesized that remyelination can be enhanced by treatment with fingolimod, a S1PR modulator currently licensed for relapsing-remitting MS. Methods MOVING was an investigator-driven, rater-blind, randomized clinical trial. Patients with acute unilateral ON, occurring as a clinically isolated syndrome or MS relapse, were randomized to 6 months of treatment with 0.5 mg oral fingolimod or subcutaneous IFN-β 1b 250 μg every other day. The change in multifocal visual evoked potential (mfVEP) latency of the qualifying eye was examined as the primary (month 6 vs. baseline) and secondary (months 3, 6 and 12 vs. baseline) outcome. In addition, full field visual evoked potentials, visual acuity, optical coherence tomography as well as clinical relapses and measures of disability, cerebral MRI, and self-reported visual quality of life were obtained for follow-up. The study was halted due to insufficient recruitment (n = 15), and available results are reported. Results Per protocol analysis of the primary endpoint revealed a significantly larger reduction of mfVEP latency at 6 months compared to baseline with fingolimod treatment (n = 5; median decrease, 15.7 ms) than with IFN-β 1b treatment (n = 4; median increase, 8.15 ms) (p < 0.001 for interaction). Statistical significance was maintained in the secondary endpoint analysis. Descriptive results are reported for other endpoints. Conclusion Preliminary results of the MOVING trial argue in support of a beneficial effect of fingolimod on optic nerve remyelination when compared to IFN-β treatment. Interpretation is limited by the small number of complete observations, an unexpected deterioration of the control group and a difference in baseline mfVEP latencies. The findings need to be confirmed in larger studies. Trial registration The trial was registered as EUDRA-CT 2011–004787-30 on October 26, 2012 and as NCT01647880 on July 24, 2012.
Collapse
Affiliation(s)
- Christian Albert
- Department of Neurology, Alexianer St. Josefs-Krankenhaus Potsdam, Allee nach Sanssouci 7, 14471, Potsdam, Germany
| | - Janine Mikolajczak
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Liekfeld
- Department of Ophthalmology, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Sophie K Piper
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Michael Scheel
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Hanna G Zimmermann
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Jan Dörr
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Oberhavel-Kliniken Hennigsdorf, Hennigsdorf, Germany
| | | | - Claudia Chien
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander U Brandt
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, University of California, Irvine, CA, USA
| | - Friedemann Paul
- Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Department of Neurology, Charité-Universitätmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olaf Hoffmann
- Department of Neurology, Alexianer St. Josefs-Krankenhaus Potsdam, Allee nach Sanssouci 7, 14471, Potsdam, Germany. .,Neurocure Clinical Research Center, Charite-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
4
|
Goin DE, Smed MK, Pachter L, Purdom E, Nelson JL, Kjærgaard H, Olsen J, Hetland ML, Zoffmann V, Ottesen B, Jawaheer D. Pregnancy-induced gene expression changes in vivo among women with rheumatoid arthritis: a pilot study. Arthritis Res Ther 2017; 19:104. [PMID: 28545501 PMCID: PMC5445464 DOI: 10.1186/s13075-017-1312-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Little is known about gene expression changes induced by pregnancy in women with rheumatoid arthritis (RA) and healthy women because the few studies previously conducted did not have pre-pregnancy samples available as baseline. We have established a cohort of women with RA and healthy women followed prospectively from a pre-pregnancy baseline. In this study, we tested the hypothesis that pregnancy-induced changes in gene expression among women with RA who improve during pregnancy (pregDASimproved) overlap substantially with changes observed among healthy women and differ from changes observed among women with RA who worsen during pregnancy (pregDASworse). METHODS Global gene expression profiles were generated by RNA sequencing (RNA-seq) from 11 women with RA and 5 healthy women before pregnancy (T0) and at the third trimester (T3). Among the women with RA, eight showed an improvement in disease activity by T3, whereas three worsened. Differential expression analysis was used to identify genes demonstrating significant changes in expression within each of the RA and healthy groups (T3 vs T0), as well as between the groups at each time point. Gene set enrichment was assessed in terms of Gene Ontology processes and protein networks. RESULTS A total of 1296 genes were differentially expressed between T3 and T0 among the 8 pregDASimproved women, with 161 genes showing at least two-fold change (FC) in expression by T3. The majority (108 of 161 genes) were also differentially expressed among healthy women (q<0.05, FC≥2). Additionally, a small cluster of genes demonstrated contrasting changes in expression between the pregDASimproved and pregDASworse groups, all of which were inducible by type I interferon (IFN). These IFN-inducible genes were over-expressed at T3 compared to the T0 baseline among the pregDASimproved women. CONCLUSIONS In our pilot RNA-seq dataset, increased pregnancy-induced expression of type I IFN-inducible genes was observed among women with RA who improved during pregnancy, but not among women who worsened. These findings warrant further investigation into expression of these genes in RA pregnancy and their potential role in modulation of disease activity. These results are nevertheless preliminary and should be interpreted with caution until replicated in a larger sample.
Collapse
Affiliation(s)
- Dana E Goin
- UCSF Benioff Children's Hospital Oakland, Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA, USA.,University of California, Berkeley, Berkeley, CA, USA
| | - Mette Kiel Smed
- Juliane Marie Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lior Pachter
- University of California, Berkeley, Berkeley, CA, USA.,California Institute of Technology, Pasadena, CA, USA
| | | | - J Lee Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,University of Washington, Seattle, WA, USA
| | - Hanne Kjærgaard
- Juliane Marie Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jørn Olsen
- University of California, Los Angeles, Los Angeles, CA, USA.,Aarhus University, Aarhus, Denmark
| | - Merete Lund Hetland
- DANBIO Registry and Copenhagen Centre for Arthritis Research, Centre for Rheumatology and Spine Diseases (VRR), Rigshospitalet, Glostrup, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vibeke Zoffmann
- Juliane Marie Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bent Ottesen
- Juliane Marie Center, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Damini Jawaheer
- UCSF Benioff Children's Hospital Oakland, Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA, USA. .,Aarhus University, Aarhus, Denmark. .,University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
5
|
Abstract
The immune system is essential for host defense against pathogen infections; however dysregulated immune response may lead to inflammatory or autoimmune diseases. Elevated activation of both innate immune cells and T cells such as Th17 cells are linked to many autoimmune diseases, including Multiple Sclerosis (MS), arthritis and inflammatory bowel disease (IBD). To keep immune homeostasis, the immune system develops a number of negative feedback mechanisms, such as the production of anti-inflammatory cytokine IL-10, to dampen excessive production of inflammatory cytokines and uncontrolled activation of immune cells. Our recent studies uncover a novel immunoregulatory function of interferon (IFN) pathways on the innate and antigen-specific immune response. Our results show that IFNα/β induced IL-10 production from macrophages and Th17 cells, which in turn negatively regulated Th17 function in autoimmune diseases such as Experimental Allergic Encephalomyelitis (EAE), an animal model of human MS. In a chronic colitis model resembling human IBD, we also found that IL-10 inhibited inflammasome/IL-1 pathway, and the pathogenicity of Th17 cells, leading to reduced chronic intestinal inflammation. Results from our and other studies further suggest that IL-10 produced by both macrophages and regulatory T cells may shift Th17 into more regulatory phenotypes, leading to reduced inflammatory response.
Collapse
Affiliation(s)
- Beichu Guo
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina 29425-5040, USA; Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, South Carolina 29425-5040, USA
| |
Collapse
|
6
|
Soluble VCAM-1 impairs human brain endothelial barrier integrity via integrin α-4-transduced outside-in signalling. Acta Neuropathol 2015; 129:639-52. [PMID: 25814153 PMCID: PMC4405352 DOI: 10.1007/s00401-015-1417-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/10/2015] [Accepted: 03/21/2015] [Indexed: 01/17/2023]
Abstract
Human brain microvascular endothelial cells forming the blood–brain barrier (BBB) release soluble vascular cell adhesion molecule-1 (sVCAM-1) under inflammatory conditions. Furthermore, sVCAM-1 serum levels in untreated patients with multiple sclerosis (MS) correlate with a breakdown of the BBB as measured by gadolinium-enhanced MRI. To date, it is unknown whether sVCAM-1 itself modulates BBB permeability. Here, we provide evidence that human brain endothelium expresses integrin α-4/β-1, the molecular binding partner of sVCAM-1, and that sVCAM-1 directly impairs BBB function by inducing intracellular signalling events through integrin α-4. Primary human brain microvascular endothelial cells showed low to moderate integrin α-4 and strong β-1 but no definite β-7 expression in vitro and in situ. Increased brain endothelial integrin α-4 expression was observed in active MS lesions in situ and after angiogenic stimulation in vitro. Exposure of cultured primary brain endothelial cells to recombinant sVCAM-1 significantly increased their permeability to the soluble tracer dextran, which was paralleled by formation of actin stress fibres and reduced staining of tight junction-associated molecules. Soluble VCAM-1 was also found to activate Rho GTPase and p38 MAP kinase. Chemical inhibition of these signalling pathways partially prevented sVCAM-1-induced changes of tight junction arrangement. Importantly, natalizumab, a neutralising recombinant monoclonal antibody against integrin α-4 approved for the treatment of patients with relapsing–remitting MS, partially antagonised the barrier-disturbing effect of sVCAM-1. In summary, we newly characterised sVCAM-1 as a compromising factor of brain endothelial barrier function that may be partially blocked by the MS therapeutic natalizumab.
Collapse
|
7
|
Svenningsson A, Bergman J, Dring A, Vågberg M, Birgander R, Lindqvist T, Gilthorpe J, Bergenheim T. Rapid depletion of B lymphocytes by ultra-low-dose rituximab delivered intrathecally. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e79. [PMID: 25745637 PMCID: PMC4345631 DOI: 10.1212/nxi.0000000000000079] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/13/2015] [Indexed: 12/21/2022]
Abstract
Objective: We are conducting an open-label phase 1b study on the efficacy of intrathecal (IT) administration of rituximab, provided via an Ommaya reservoir, for the treatment of progressive multiple sclerosis (PMS). The objective of this initial study was to monitor B lymphocytes in peripheral blood (PB) and CSF from the first 10 patients 1 year posttreatment. Methods: Dose titration was performed with daily escalation from 1 mg to 25 mg IT rituximab (n = 3). Lymphocyte subpopulations were monitored daily during dose escalation in PB by flow cytometry and subsequently every 3 months for 1 year, after a total dose of 3 × 25 mg. PB B-lymphocyte subpopulations for the remaining patients (n = 7) were monitored at regular intervals. CSF lymphocyte subpopulations for all patients were monitored by flow cytometry every 2–3 months. Results: The PB B-lymphocyte count dropped rapidly after the first 2 injections (total dose of 3.5 mg IT rituximab) to undetectable levels. Three 25-mg doses given once per week depleted peripheral B lymphocytes entirely for the following 3–6 month period. Conclusions: Monoclonal antibodies seem to rapidly redistribute to the peripheral compartment following IT injection. Ultra-low doses of rituximab given IT are sufficient to cause complete depletion of peripheral B lymphocytes, indicating that low-dose IT treatment has the potential to be effective in both the CNS and systemic compartments. Classification of evidence: This study provides Class IV evidence that for patients with PMS, rituximab provided via an Ommaya reservoir depletes peripheral blood B lymphocytes.
Collapse
Affiliation(s)
- Anders Svenningsson
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| | - Joakim Bergman
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| | - Ann Dring
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| | - Mattias Vågberg
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| | - Richard Birgander
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| | - Thomas Lindqvist
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| | - Jonathan Gilthorpe
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| | - Tommy Bergenheim
- Department of Pharmacology and Clinical Neuroscience (A.S., J.B., A.D., M.V., J.G., T.B.) and Department of Radiation Sciences (R.B., T.L.), Diagnostic Radiology, Umeå University, Sweden
| |
Collapse
|
8
|
Berrih-Aknin S. Myasthenia Gravis: paradox versus paradigm in autoimmunity. J Autoimmun 2014; 52:1-28. [PMID: 24934596 DOI: 10.1016/j.jaut.2014.05.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Myasthenia Gravis (MG) is a paradigm of organ-specific autoimmune disease (AID). It is mediated by antibodies that target the neuromuscular junction. The purpose of this review is to place MG in the general context of autoimmunity, to summarize the common mechanisms between MG and other AIDs, and to describe the specific mechanisms of MG. We have chosen the most common organ-specific AIDs to compare with MG: type 1 diabetes mellitus (T1DM), autoimmune thyroid diseases (AITD), multiple sclerosis (MS), some systemic AIDs (systemic lupus erythematous (SLE), rheumatoid arthritis (RA), Sjogren's syndrome (SS)), as well as inflammatory diseases of the gut and liver (celiac disease (CeD), Crohn's disease (CD), and primary biliary cirrhosis (PBC)). Several features are similar between all AIDs, suggesting that common pathogenic mechanisms lead to their development. In this review, we address the predisposing factors (genetic, epigenetic, hormones, vitamin D, microbiota), the triggering components (infections, drugs) and their interactions with the immune system [1,2]. The dysregulation of the immune system is detailed and includes the role of B cells, Treg cells, Th17 and cytokines. We particularly focused on the role of TNF-α and interferon type I whose role in MG is very analogous to that in several other AIDS. The implication of AIRE, a key factor in central tolerance is also discussed. Finally, if MG is a prototype of AIDS, it has a clear specificity compared to the other AIDS, by the fact that the target organ, the muscle, is not the site of immune infiltration and B cell expansion, but exclusively that of antibody-mediated pathogenic mechanisms. By contrast, the thymus in the early onset subtype frequently undergoes tissue remodeling, resulting in the development of ectopic germinal centers surrounded by high endothelial venules (HEV), as observed in the target organs of many other AIDs.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center UM76, F-75013 Paris, France; INSERM U974, F-75013 Paris, France; CNRS FRE 3617, F-75013 Paris, France; Institute of Myology, F-75013 Paris, France.
| |
Collapse
|
9
|
Zhou H, Wang Y, Lian Q, Yang B, Ma Y, Wu X, Sun S, Liu Y, Sun B. Differential IL-10 production by DCs determines the distinct adjuvant effects of LPS and PTX in EAE induction. Eur J Immunol 2014; 44:1352-62. [PMID: 24496948 DOI: 10.1002/eji.201343744] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 01/07/2014] [Accepted: 01/24/2014] [Indexed: 02/06/2023]
Abstract
Pertussis toxin (PTX) is used as an adjuvant to induce EAE in mice as a model of MS. Although LPS and PTX are similar in their ability to mature dendritic cells (DCs) and in their adjuvant properties, here we demonstrate that LPS does not induce EAE development. We also demonstrate that DCs treated with PTX (PTX-DCs) are able to induce EAE, whereas DCs treated with LPS (LPS-DCs) fail to induce EAE. We determine that a key difference between LPS-DCs and PTX-DCs is that LPS-DCs produce larger amounts of IL-10. IL-10(-/-) -DCs treated with LPS promote stronger IFN-γ and IL-17 production and T-cell proliferation than WT DCs treated with LPS in a coculture system. Finally, we demonstrate that EAE can be successfully induced when IL-10(-/-) -DCs treated with LPS are used as an adjuvant, whereas the use of PTX-DCs overexpressing IL-10 as an adjuvant markedly controls EAE development. These results indicate that the inability of LPS to induce EAE is based on the induction of high levels of IL-10 in the targeted DCs, providing insight into the mechanisms responsible for the induction of EAE.
Collapse
Affiliation(s)
- Haiyan Zhou
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Valatas V, Vakas M, Kolios G. The value of experimental models of colitis in predicting efficacy of biological therapies for inflammatory bowel diseases. Am J Physiol Gastrointest Liver Physiol 2013; 305:G763-85. [PMID: 23989010 DOI: 10.1152/ajpgi.00004.2013] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
During the last decade, biological therapies have an increasing share in the modern therapeutics of various diseases including inflammatory bowel diseases (IBD). Animal models of IBD have often been used to identify the targets of biological therapies, to test their relevance to disease pathogenesis, to assess their therapeutic efficacy in vivo, and to check for drug toxicity. In the field of inflammatory diseases the majority of biologics under development have failed to reach the clinic. This review examines the ability of preclinical data from animal models of IBD to predict success or failure of biologics in human IBD. Specifically, it describes the murine models of IBD, the mechanism of disease induction, the phenotype of the disease, its relevance to human IBD, and the specific immunological features of disease pathogenesis in each model and mainly compares the results of the phase II and III trials of biologics in IBD with preclinical data obtained from studies in animal models. Finally, it examines the possible reasons for low success in translation from bench to bedside and offers some suggestions to improve translation rates.
Collapse
Affiliation(s)
- Vassilis Valatas
- Dept. of Gastroenterology, Univ. Hospital of Heraklion, PO Box 1352, Voutes, Heraklion, GR-71100, Crete, Greece.
| | | | | |
Collapse
|
11
|
Hartung HP, Haas J, Meergans M, Tracik F, Ortler S. [Interferon-β1b in multiple sclerosis therapy: more than 20 years clinical experience]. DER NERVENARZT 2013; 84:679-704. [PMID: 23669866 DOI: 10.1007/s00115-013-3781-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The introduction of interferon-β1b in 1993 in the USA and 2 years later in Europe made it possible for the first time to alter the course of the disease in patients with relapsing-remitting multiple sclerosis (MS). Subsequently, interferon-β1b was approved for the treatment of patients with active secondary progressive MS (1999) and early relapsing-remitting MS following a first demyelinating event (clinically isolated syndrome, CIS) (2006). Here we provide an overview of the clinical experience gathered during more than 20 years of interferon-β use focusing on long-term efficacy and safety and the impact of early initiation of treatment. Furthermore, the following aspects will be discussed: putative mechanisms of action of interferon-β, indications for a disease-modifying therapy, clinical relevance of neutralizing antibodies, importance of adherence in MS therapy, high versus low frequency therapy, combination therapies with interferon-β and safety of interferon-β in children and adolescents with MS and during pregnancy.
Collapse
Affiliation(s)
- H-P Hartung
- Neurologische Klinik im UKD, Medizinische Fakultät, Heinrich-Heine Universität, Moorenstr. 5, 40225 Düsseldorf, Deutschland.
| | | | | | | | | |
Collapse
|
12
|
Koh M, Shinohara J, Hongo Y, Okazaki T, Takitani K, Tamai H. Case treated with triple therapy of lamivudine, interferon-β and prednisolone for acute exacerbation of chronic hepatitis B during pregnancy. Hepatol Res 2013; 43:425-9. [PMID: 23560863 PMCID: PMC3638365 DOI: 10.1111/j.1872-034x.2012.01077.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We herein report a case of a pregnant Chinese woman who suffered an acute exacerbation of hepatitis B. The patient's liver enzymes became elevated toward the end of the first trimester. She was treated with lamivudine, interferon (IFN)-β and steroids early in the second trimester. After this treatment regimen was initiated, aminotransferase levels rapidly normalized within 4 weeks. IFN-β and steroids were administrated for 2 weeks in the second trimester, while the administration of lamivudine continued until delivery. The spontaneous delivery of a female baby weighing 2984 g occurred at 37 weeks of gestation. A neonatal examination revealed no congenital anomalies, and fetal growth was found to be within normal reference ranges. The infant received simultaneous active and passive hepatitis B virus immunization within 12 h of delivery and completed the hepatitis B vaccine schedule at 2, 3 and 5 months of age. The infant was successfully prevented from contracting hepatitis B virus. This case suggests that combination therapy with lamivudine, IFN-β and steroids may be safely used during the second trimester to treat acute exacerbations of hepatitis B.
Collapse
Affiliation(s)
- Maki Koh
- Department of PediatricsHirakata
| | | | - Yasushi Hongo
- Department of Gastroenterology and HepatologyHirakata
| | - Tadashi Okazaki
- Department of Obstetrics and Gynecology, Hirakata City HospitalHirakata
| | | | - Hiroshi Tamai
- Department of Pediatrics, Osaka Medical CollageTakatsuki, Japan
| |
Collapse
|
13
|
Fitzgerald DC, Fonseca-Kelly Z, Cullimore ML, Safabakhsh P, Saris CJM, Zhang GX, Rostami A. Independent and interdependent immunoregulatory effects of IL-27, IFN-β, and IL-10 in the suppression of human Th17 cells and murine experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 190:3225-34. [PMID: 23455508 DOI: 10.4049/jimmunol.1200141] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IFN-β, IL-27, and IL-10 have been shown to exert a range of similar immunoregulatory effects in murine and human experimental systems, particularly in Th1- and Th17-mediated models of autoimmune inflammatory disease. In this study we sought to translate some of our previous findings in murine systems to human in vitro models and delineate the interdependence of these different cytokines in their immunoregulatory effects. We demonstrate that human IL-27 upregulates IL-10 in T cell-activated PBMC cultures and that IFN-β drives IL-27 production in activated monocytes. IFN-β-driven IL-27 is responsible for the upregulation of IL-10, but not IL-17 suppression, by IFN-β in human PBMCs. Surprisingly, IL-10 is not required for the suppression of IL-17 by either IL-27 or IFN-β in this model or in de novo differentiating Th17 cells, nor is IL-27 signaling required for the suppression of experimental autoimmune encephalomyelitis (EAE) by IFN-β in vivo. Furthermore, and even more surprisingly, IL-10 is not required for the suppression of Th17-biased EAE by IL-27, in sharp contrast to Th1-biased EAE. In conclusion, IFN-β and IL-27 both induce human IL-10, both suppress human Th17 responses, and both suppress murine EAE. However, IL-27 signaling is not required for the therapeutic effect of IFN-β in EAE. Suppression of Th17-biased EAE by IL-27 is IL-10-independent, in contrast to its mechanism of action in Th1-biased EAE. Taken together, these findings delineate a complex set of interdependent and independent immunoregulatory mechanisms of IFN-β, IL-27, and IL-10 in human experimental models and in murine Th1- and Th17-driven autoimmunity.
Collapse
Affiliation(s)
- Denise C Fitzgerald
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, Northern Ireland, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
14
|
Gordon RA, Grigoriev G, Lee A, Kalliolias GD, Ivashkiv LB. The interferon signature and STAT1 expression in rheumatoid arthritis synovial fluid macrophages are induced by tumor necrosis factor α and counter-regulated by the synovial fluid microenvironment. ACTA ACUST UNITED AC 2013; 64:3119-28. [PMID: 22614743 DOI: 10.1002/art.34544] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Type I interferons (IFNs) have emerged as potential activators of the IFN signature and elevated STAT-1 expression in rheumatoid arthritis (RA) synovium, but mechanisms that induce synovial IFN expression are unknown. Recently, tumor necrosis factor α (TNFα) was shown to induce a delayed IFN response in macrophages. We undertook this study to test whether TNFα, classically thought to activate inflammatory NF-κB target genes in RA, also contributes to the "IFN signature" in RA synovial macrophages. METHODS Synovial fluid (SF) macrophages purified from 24 patients with RA and 18 patients with spondylarthritides (SpA) were lysed immediately after isolation or were cultured ex vivo in the absence or presence of blockade of endogenous type I IFN or TNFα. Expression of IFN-inducible target genes was measured by quantitative reverse transcription-polymerase chain reaction, and expression of their corresponding proteins was measured by enzyme-linked immunosorbent assay. RESULTS Expression of an IFN signature and STAT1 in RA synovial macrophages was suppressed when type I IFNs or TNFα were blocked, whereas TNFα blockade did not affect expression of IFN response genes or STAT1 in SpA synovial macrophages. RA SF suppressed the IFN signature in RA synovial macrophages and in TNFα-, IFNα-, and IFNβ-stimulated control macrophages. Type I IFNs suppressed expression of IL8 and MMP9 in RA synovial macrophages and in TNFα-stimulated control macrophages. CONCLUSION Our findings identify a new function of TNFα in RA synovitis by implicating TNFα as a major inducer of the RA synovial IFN response. The results suggest that the expression of IFN response genes in RA synovium is regulated by interplay between TNFα and opposing homeostatic factors expressed in the synovial microenvironment.
Collapse
|
15
|
Chakravarty SD, Harris ME, Schreiner AM, Crow MK. Sarcoidosis triggered by interferon-Beta treatment of multiple sclerosis: a case report and focused literature review. Semin Arthritis Rheum 2012; 42:206-12. [PMID: 22572392 DOI: 10.1016/j.semarthrit.2012.03.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 03/12/2012] [Accepted: 03/18/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To report a rare case of sarcoidosis induced by chronic interferon-beta (a type I interferon) therapy of multiple sclerosis and to review previously reported cases. METHODS We describe a patient with a prior diagnosis of multiple sclerosis, who developed noncaseating granulomas in her skin and pulmonary lymph nodes, consistent with sarcoidosis, while being treated with recombinant interferon-beta. A retrospective review of the literature was performed using the PubMed database. RESULTS In our patient, sarcoidosis developed after 3 years of continuous recombinant interferon-beta therapy, dosed 3 times a week. The patient presented with progressive dyspnea on exertion, diffuse arthralgias, low-grade fevers, with an acute onset of rash. The diagnosis of sarcoidosis was secured by finding typical, well-formed, noncaseating granulomas on skin and endobronchial biopsies, with other possible etiologies for granulomatous conditions excluded beforehand. Following the withdrawal of recombinant interferon-beta and a course of corticosteroids combined with disease-modifying anti-rheumatic drug therapy, the patient's clinical presentation resolved. Excluding ours, only 4 additional cases of sarcoidosis developing after interferon-beta therapy have been reported, with 2 of those cases in the context of underlying multiple sclerosis. CONCLUSIONS Developing sarcoidosis during treatment of multiple sclerosis with recombinant interferon-beta represents an exceedingly rare and paradoxical adverse event. The occurrence of sarcoidosis with the use of this agent is perhaps due to a dysregulation in the modulatory role played by interferon-beta (and more generally type I interferon) expression in chronic inflammation.
Collapse
Affiliation(s)
- Soumya D Chakravarty
- Division of Rheumatology, Hospital for Special Surgery and Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
16
|
Gough DJ, Messina NL, Clarke CJP, Johnstone RW, Levy DE. Constitutive type I interferon modulates homeostatic balance through tonic signaling. Immunity 2012; 36:166-74. [PMID: 22365663 DOI: 10.1016/j.immuni.2012.01.011] [Citation(s) in RCA: 349] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/16/2012] [Accepted: 01/31/2012] [Indexed: 02/07/2023]
Abstract
Interferons (IFNs) were discovered as cytokines induced during and protecting from viral infection. They have been documented to play essential roles in numerous physiological processes beyond antiviral and antimicrobial defense, including immunomodulation, cell cycle regulation, cell survival, and cell differentiation. Recent data have also uncovered a potentially darker side to IFN, including roles in inflammatory diseases, such as autoimmunity and diabetes. IFN can have effects in the absence of acute infection, highlighting a physiologic role for constitutive IFN. Type I IFNs are constitutively produced at vanishingly low quantities and yet exert profound effects, mediated in part through modulation of signaling intermediates required for responses to diverse cytokines. We review evidence for a yin-yang of IFN function through its role in modulating crosstalk between multiple cytokines by both feedforward and feedback regulation of common signaling intermediates and postulate a homeostatic role for IFN through tonic signaling in the absence of acute infection.
Collapse
Affiliation(s)
- Daniel J Gough
- New York University Medical Center, New York, NY 10016, USA
| | | | | | | | | |
Collapse
|
17
|
Type I IFN promotes IL-10 production from T cells to suppress Th17 cells and Th17-associated autoimmune inflammation. PLoS One 2011; 6:e28432. [PMID: 22163016 PMCID: PMC3232207 DOI: 10.1371/journal.pone.0028432] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 11/08/2011] [Indexed: 02/07/2023] Open
Abstract
Whereas the immune system is essential for host defense against pathogen infection or endogenous danger signals, dysregulated innate and adaptive immune cells may facilitate harmful inflammatory or autoimmune responses. In the CNS, chronic inflammation plays an important role in the pathogenesis of neurodegenerative diseases such as multiple sclerosis (MS). Our previous study has demonstrated a critical role for the type I IFN induction and signaling pathways in constraining Th17-mediated experimental autoimmune encephalomyelitis (EAE), an animal model of human MS. However, it remains unknown if self-reactive Th17 cells can be reprogrammed to have less encephalitogenic activities or even have regulatory effects through modulation of innate pathways. In this study, we investigated the direct effects of type I IFN on Th17 cells. Our data show that IFNβ treatment of T cells cultured under Th17 polarizing conditions resulted in reduced production of IL-17, but increased production of IL-10. We also found that IFNβ induced IL-10 production by antigen specific T cells derived from immunized mice. Furthermore, IFNβ treatment could suppress the encephalitogenic activity of myelin-specific T cells, and ameliorate clinical symptoms of EAE in an adoptive transfer model. Together, results from this study suggest that IFNβ may induce antigen-specific T cells to produce IL-10, which in turn negatively regulate Th17-mediate inflammatory and autoimmune response.
Collapse
|
18
|
Avnir Y, Turjeman K, Tulchinsky D, Sigal A, Kizelsztein P, Tzemach D, Gabizon A, Barenholz Y. Fabrication principles and their contribution to the superior in vivo therapeutic efficacy of nano-liposomes remote loaded with glucocorticoids. PLoS One 2011; 6:e25721. [PMID: 21998684 PMCID: PMC3188566 DOI: 10.1371/journal.pone.0025721] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Accepted: 09/09/2011] [Indexed: 11/18/2022] Open
Abstract
We report here the design, development and performance of a novel formulation of liposome- encapsulated glucocorticoids (GCs). A highly efficient (>90%) and stable GC encapsulation was obtained based on a transmembrane calcium acetate gradient driving the active accumulation of an amphipathic weak acid GC pro-drug into the intraliposome aqueous compartment, where it forms a GC-calcium precipitate. We demonstrate fabrication principles that derive from the physicochemical properties of the GC and the liposomal lipids, which play a crucial role in GC release rate and kinetics. These principles allow fabrication of formulations that exhibit either a fast, second-order (t(1/2) ~1 h), or a slow, zero-order release rate (t(1/2) ~ 50 h) kinetics. A high therapeutic efficacy was found in murine models of experimental autoimmune encephalomyelitis (EAE) and hematological malignancies.
Collapse
Affiliation(s)
- Yuval Avnir
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Keren Turjeman
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Deborah Tulchinsky
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Alex Sigal
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Pablo Kizelsztein
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Dina Tzemach
- Laboratory of Experimental Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Alberto Gabizon
- Laboratory of Experimental Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
19
|
Sandberg-Wollheim M, Alteri E, Moraga MS, Kornmann G. Pregnancy outcomes in multiple sclerosis following subcutaneous interferon beta-1a therapy. Mult Scler 2011; 17:423-30. [PMID: 21220368 DOI: 10.1177/1352458510394610] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Women with multiple sclerosis (MS) are advised to discontinue interferon-beta therapy before trying to conceive. Unplanned pregnancies occur and risks related to exposure remain unclear. METHODS To determine pregnancy outcomes following interferon-beta therapy, we examined pregnancies from a global drug safety database containing individual case safety reports received in the post-marketing setting and safety data from clinical trials of subcutaneous interferon beta-1a in MS. RESULTS One thousand and twenty-two cases of exposure to subcutaneous interferon beta-1a during pregnancy were retrieved; 679 had a documented outcome. In cases for which exposure duration was available (n = 231), mean time of foetal exposure to subcutaneous interferon beta-1a before treatment discontinuation was 28 days; most pregnancies (199/231; 86.1%) were exposed for ≤ 45 days. To avoid bias, only outcomes for prospective data (n = 425) in pregnancies exposed to interferon beta-1a in utero were analysed further. Of these, 324 (76.2%) resulted in normal live births and four (0.9%) in live births with congenital anomalies (3 [0.7%] were 'major'). Four (0.9%) pregnancies resulted in stillbirths (1 [0.2%] with foetal defects). There were 5 (1.2%) ectopic pregnancies, 49 (11.5%) spontaneous abortions and 39 (9.2%) elective terminations. Most pregnancies exposed to subcutaneous interferon beta-1a in utero were associated with normal live births. The rates of spontaneous abortion and major congenital anomalies in live births were in line with those observed in the general population. CONCLUSIONS These data should be taken into account when considering options for women with MS who become pregnant or who are planning pregnancy while on treatment with subcutaneous interferon beta-1a.
Collapse
|
20
|
Crow MK. Type I interferon in organ-targeted autoimmune and inflammatory diseases. Arthritis Res Ther 2010; 12 Suppl 1:S5. [PMID: 21303493 PMCID: PMC2991778 DOI: 10.1186/ar2886] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A significant role for IFNα in the pathogenesis of systemic lupus erythematosus is well supported, and clinical trials of anti-IFNα monoclonal antibodies are in progress in this disease. In other autoimmune diseases characterized by substantial inflammation and tissue destruction, the role of type I interferons is less clear. Gene expression analysis of peripheral blood cells from patients with rheumatoid arthritis and multiple sclerosis demonstrate an interferon signature similar to but less intense than that seen in patients with lupus. In both of those diseases, presence of the interferon signature has been associated with more significant clinical manifestations. At the same time, evidence supports an anti-inflammatory and beneficial role of IFNβ locally in the joints of patients with rheumatoid arthritis and in murine arthritis models, and many patients with multiple sclerosis show a clinical response to recombinant IFNβ. As can also be proposed for type I diabetes mellitus, type I interferon appears to contribute to the development of autoimmunity and disease progression in multiple autoimmune diseases, while maintaining some capacity to control established disease - particularly at local sites of inflammation. Recent studies in both rheumatoid arthritis and multiple sclerosis suggest that quantification of type I interferon activity or target gene expression might be informative in predicting responses to distinct classes of therapeutic agents.
Collapse
Affiliation(s)
- Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, Weill Cornell Medical College, 535 East 70th Street, New York, NY 10021, USA.
| |
Collapse
|
21
|
Prinz M, Kalinke U. New lessons about old molecules: how type I interferons shape Th1/Th17-mediated autoimmunity in the CNS. Trends Mol Med 2010; 16:379-86. [PMID: 20591737 DOI: 10.1016/j.molmed.2010.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/14/2010] [Accepted: 06/03/2010] [Indexed: 01/02/2023]
Abstract
Type I interferons (IFN-alpha and IFN-beta) were discovered more than five decades ago and are widely used for the treatment of human autoimmune diseases such as multiple sclerosis (MS). Despite their highly beneficial features, the precise mechanism of action remains speculative. Given the frequent side effects of IFN-alpha/beta therapy, understanding its action in an in vivo setting is vital to further improve this therapeutic approach. Major advances in our understanding of the IFN biology have recently been made and are particularly based on the combination of powerful genome-wide expression analysis in humans with gene-targeting techniques available for basic research. The recent discovery of a novel T-cell subset, Th17 cells, sheds new light on type I IFNs in MS.
Collapse
Affiliation(s)
- Marco Prinz
- Department of Neuropathology, University of Freiburg, Breisacher Str. 64, D-79106 Freiburg, Germany.
| | | |
Collapse
|
22
|
Lundkvist M, Greiner E, Hillert J, Fogdell-Hahn A. Multiple sclerosis patients lacking oligoclonal bands in the cerebrospinal fluid are less likely to develop neutralizing antibodies against interferon beta. Mult Scler 2010; 16:796-800. [PMID: 20534645 DOI: 10.1177/1352458510373112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Multiple sclerosis patients without cerebrospinal fluid oligoclonal IgG bands have been proposed to constitute an immunogenetically distinct subgroup of multiple sclerosis that may also differ in terms of prognosis. A proportion of patients with multiple sclerosis receiving IFNbeta develop neutralizing antibodies, which interfere with treatment efficacy. Evidence suggests that the likelihood of developing neutralizing antibodies is partly genetically determined. Here, we hypothesized that absence of oligoclonal IgG bands reflects a property of B-cell responses in oligoclonal IgG band-negative patients characterized by a lessened propensity to develop neutralizing antibodies. We aimed to compare the development of neutralizing antibodies against IFNbeta between oligoclonal IgG band-negative and oligoclonal IgG band-positive multiple sclerosis patients. Treatment, oligoclonal IgG band and neutralizing antibody information was obtained for 2219 patients from the Swedish multiple sclerosis registry and the Swedish neutralizing antibody registry. Additional data on genotype was available for 532 patients. A correlation was found between oligoclonal IgG band negativity and neutralizing antibody negativity (p = 0.02). This difference was confined to neutralizing antibodies against IFNbeta-1a, since oligoclonal IgG band-negative patients were, to a lesser extent, neutralizing antibody positive compared with oligoclonal IgG band-positive patients if treated with IFNbeta-1a (12% vs. 23%; p = 0.005). No difference was observed for IFNbeta-1b-treated patients (44% vs. 46%). We propose that oligoclonal IgG band-negative patients differ immunologically from oligoclonal IgG band-positive patients, potentially influenced by distinct HLA-DRB1 alleles.
Collapse
Affiliation(s)
- M Lundkvist
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
23
|
Abstract
Type I interferons are pleiotropic cytokines with antiviral, antitumor and immunoregulatory functions. An aspect of their complex biology is the paradox that, depending on context, type I interferons can be anti-inflammatory and tissue protective or can be proinflammatory and promote autoimmunity. Along these lines, the activation of type I interferon pathways is effective in suppressing disease activity in patients with multiple sclerosis and in animal models of arthritis and colitis, while there is an expectation that blockade of the same pathways will be beneficial in the treatment of patients with systemic lupus erythematosus.
Collapse
Affiliation(s)
- George D Kalliolias
- Arthritis and Tissue Degeneration Program and Department of Medicine, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | | |
Collapse
|
24
|
Molle C, Goldman M, Goriely S. Critical Role of the IFN-Stimulated Gene Factor 3 Complex in TLR-Mediated IL-27p28 Gene Expression Revealing a Two-Step Activation Process. THE JOURNAL OF IMMUNOLOGY 2010; 184:1784-92. [DOI: 10.4049/jimmunol.0902005] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Yoshida H, Nakaya M, Miyazaki Y. Interleukin 27: a double-edged sword for offense and defense. J Leukoc Biol 2009; 86:1295-303. [DOI: 10.1189/jlb.0609445] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
26
|
De Clercq E. Antiviral drug discovery: Ten more compounds, and ten more stories (part B). Med Res Rev 2009; 29:571-610. [DOI: 10.1002/med.20149] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
27
|
Perrin Ross A. The Importance of Early Treatment. J Neurosci Nurs 2009. [DOI: 10.1097/jnn.0b013e3181a23ea9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
28
|
Goriely S, Cavoy R, Goldman M. Interleukin-12 family members and type I interferons in Th17-mediated inflammatory disorders. Allergy 2009; 64:702-9. [PMID: 19383026 DOI: 10.1111/j.1398-9995.2009.02039.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cytokines produced by antigen-presenting cells govern the fate of helper T-cell responses. Herein, we review the impact of interleukin (IL)-23 and IL-27 on the outcome of T-helper (Th) 17 cell responses and discuss their impact in the pathogenesis of T-cell-mediated inflammatory disorders of autoimmune or allergic origin. We then discuss how type I interferons might influence the course of autoimmune diseases by tipping the balance between IL-12 family members.
Collapse
Affiliation(s)
- S Goriely
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi, Belgium
| | | | | |
Collapse
|
29
|
Boasso A. Type I interferon in HIV treatment: from antiviral drug to therapeutic target. HIV THERAPY 2009; 3:269-282. [PMID: 32280376 PMCID: PMC7147345 DOI: 10.2217/hiv.09.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Type I interferons (IFNs) are soluble molecules that exert potent antiviral activity and are currently used for the treatment of a panel of viral infections. In the case of HIV, the use of type I IFN has had limited success, and has almost been abandoned. During the last decade, a series of studies has highlighted how HIV infection may cause overactivation of type I IFN production, which contributes to the exhaustion of the immune system and to disease progression. This review describes the transition from the proposed use of type I IFN as antiviral drugs in HIV infection, to the idea that blocking their activity or production may provide an immunologic benefit of much greater importance than their antiviral activity.
Collapse
Affiliation(s)
- Adriano Boasso
- >Department of Immunology, Division of Investigative Science, Faculty of Medicine, Imperial College, Chelsea & Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK. Tel.: +44 208 746 5993; ;
| |
Collapse
|
30
|
Mirandola SR, Hallal DEM, Farias AS, Oliveira EC, Brandão CO, Ruocco HH, Damasceno BP, Santos LMB. Interferon-beta modifies the peripheral blood cell cytokine secretion in patients with multiple sclerosis. Int Immunopharmacol 2009; 9:824-30. [PMID: 19289181 DOI: 10.1016/j.intimp.2009.03.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 02/23/2009] [Accepted: 03/06/2009] [Indexed: 11/19/2022]
Abstract
Immunotherapy with Interferon-beta (IFNbeta) results in remarkably beneficial effects in patients with relapsing-remitting multiple sclerosis (MS), although the mechanisms by which it exerts these beneficial effects remain poorly understood. An investigation was made of the effects of IFNbeta on pro-inflammatory and anti-inflammatory cytokine production in peripheral blood cells in MS patients, both untreated and those undergoing immunotherapy, as well as in healthy controls. Results show a significant increase in the production of pro-inflammatory cytokines such as TNFalpha, IFNgamma and IL-12 in the plasma and in the supernatant of leukocyte cultures from MS patients with the untreated disease; IFNbeta administration significantly reduced the levels of TNFalpha and IFNgamma, with no changes in the level of IL-12. The Interferon-beta therapy also led to a significant increase in the production of IL-10, as well as a slight increase in that of TGFbeta. The reduction in pro-inflammatory cytokine production in the treated MS patient group, accompanied by a simultaneous increase in the production of anti-inflammatory cytokines and the reduction of relapse rates suggests that the beneficial effects of IFNbeta immunotherapy result, at least in part, from the modulation of cytokine patterns.
Collapse
Affiliation(s)
- Sandra R Mirandola
- Neuroimmunology-Unit, Department of Genetics, Evolution and Bioagents, UNICAMP, Campinas, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
31
|
The implications of immunogenicity for protein-based multiple sclerosis therapies. J Neurol Sci 2008; 275:7-17. [DOI: 10.1016/j.jns.2008.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 07/31/2008] [Accepted: 08/05/2008] [Indexed: 11/21/2022]
|
32
|
Guo B, Chang EY, Cheng G. The type I IFN induction pathway constrains Th17-mediated autoimmune inflammation in mice. J Clin Invest 2008; 118:1680-90. [PMID: 18382764 DOI: 10.1172/jci33342] [Citation(s) in RCA: 283] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 02/13/2008] [Indexed: 01/17/2023] Open
Abstract
IFN-beta, a type I IFN, is widely used for the treatment of MS. However, the mechanisms behind its therapeutic efficacy are not well understood. Using a murine model of MS, EAE, we demonstrate that the Th17-mediated development of autoimmune disease is constrained by Toll-IL-1 receptor domain-containing adaptor inducing IFN-beta-dependent (TRIF-dependent) type I IFN production and its downstream signaling pathway. Mice with defects in TRIF or type I IFN receptor (IFNAR) developed more severe EAE. Notably, these mice exhibited marked CNS inflammation, as manifested by increased IL-17 production. In addition, IFNAR-dependent signaling events were essential for negatively regulating Th17 development. Finally, IFN-beta-mediated IL-27 production by innate immune cells was critical for the immunoregulatory role of IFN-beta in the CNS autoimmune disease. Together, our findings not only may provide a molecular mechanism for the clinical benefits of IFN-beta in MS but also demonstrate a regulatory role for type I IFN induction and its downstream signaling pathways in limiting Th17 development and autoimmune inflammation.
Collapse
Affiliation(s)
- Beichu Guo
- Department of Microbiology, Immunology, and Molecular Genetics, 8-240 Factor Building, University of California, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
33
|
Hooks JJ, Nagineni CN, Hooper LC, Hayashi K, Detrick B. IFN-beta provides immuno-protection in the retina by inhibiting ICAM-1 and CXCL9 in retinal pigment epithelial cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:3789-96. [PMID: 18322185 DOI: 10.4049/jimmunol.180.6.3789] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The retinal pigment epithelial (RPE) cell is a potent regulatory cell that facilitates normal physiologic processes and plays a critical role in a variety of retinal diseases. We evaluated IFN-beta production in human RPE cells through TLR signaling and investigated the effects of IFN-beta on RPE cells. RPE cells treated with poly(I:C) or infected with an RNA virus produce IFN-beta. Kinetic studies revealed that IFN-beta levels continue to increase over a 48-h period and this was associated with the up-regulation of IRF-7 gene expression, a known positive feedback molecule for IFN-beta production. Microarray analysis revealed that in IFN-beta treated cells, 480 genes of 22,283 genes were up or down-regulated by >2-fold. We hypothesize that IFN-beta induction during TLR signaling in the retina is an immunosuppressive factor produced to limit immunopathologic damage. Cytokine activation of RPE cells results in the production of the chemokines, CXCL9 and CXCL10, and the adhesion molecule, ICAM-1. Pretreatment of RPE cells with IFN-beta resulted in inhibition of ICAM-1 production and elimination of CXCL9 production. This treatment did not alter CXCL10 production. Anti-IFN-beta Ab blocked the inhibitory action of IFN-beta. Real time PCR analysis revealed that IFN-beta treatment inhibited gene expression of sICAM-1 and CXCL9. The results indicate a critical role for RPE cell derived IFN-beta in the down-regulation of CXCL9 and ICAM-1 expression in the retina and suggest that the inhibition of CXCL9 is an immuno-suppressive mechanism that protects the retina from excessive inflammation.
Collapse
Affiliation(s)
- John J Hooks
- Immunology and Virology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
34
|
Gold R, Rieckmann P. Fortschritte im Verständnis von Pathogenese und Immuntherapie der Multiplen Sklerose. DER NERVENARZT 2007; 78 Suppl 1:15-24; quiz 25. [PMID: 17668159 DOI: 10.1007/s00115-007-2327-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In this article recent advances in research on the pathogenesis of multiple sclerosis (MS) are summarized. New evidence from molecular histopathology is discussed focussing on neurodegenerative aspects. In addition findings with a direct effect on therapeutic decisions are presented which have contributed to improved immunotherapy. During the last decade important advances in immunotherapy have proven especially useful for patients with relapsing-remitting MS. Escalating algorithms are available for both relapses and long-term immunotherapy. Novel therapeutic approaches with monoclonal antibodies have increasing importance, yet side effects are not completely understood. The pathogenetic insights presented here may open new avenues for novel immunotherapies and lead to individualized MS therapy in the future. Limitations are given for primary progressive MS due to the lack of suitable tissue specimens and experimental models. Neuroprotective treatment strategies aiming at the protection of glial and neuronal cells are still in early stages of development.
Collapse
MESH Headings
- Alemtuzumab
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/adverse effects
- Antibodies, Neoplasm/therapeutic use
- Cerebral Cortex/drug effects
- Cerebral Cortex/pathology
- Clinical Trials as Topic
- Daclizumab
- Glatiramer Acetate
- Glucocorticoids/adverse effects
- Glucocorticoids/therapeutic use
- Humans
- Immunoglobulin G/adverse effects
- Immunoglobulin G/therapeutic use
- Immunosuppressive Agents/adverse effects
- Immunosuppressive Agents/therapeutic use
- Immunotherapy/adverse effects
- Immunotherapy/methods
- Interferon-beta/adverse effects
- Interferon-beta/therapeutic use
- Mitoxantrone/adverse effects
- Mitoxantrone/therapeutic use
- Multiple Sclerosis, Chronic Progressive/drug therapy
- Multiple Sclerosis, Chronic Progressive/etiology
- Multiple Sclerosis, Chronic Progressive/pathology
- Multiple Sclerosis, Relapsing-Remitting/drug therapy
- Multiple Sclerosis, Relapsing-Remitting/etiology
- Multiple Sclerosis, Relapsing-Remitting/pathology
- Natalizumab
- Nerve Fibers, Myelinated/drug effects
- Nerve Fibers, Myelinated/pathology
- Neurons/drug effects
- Neurons/pathology
- Neuroprotective Agents/adverse effects
- Neuroprotective Agents/therapeutic use
- Oligodendroglia/drug effects
- Oligodendroglia/pathology
- Peptides/adverse effects
- Peptides/therapeutic use
- Recombinant Proteins/adverse effects
- Recombinant Proteins/therapeutic use
Collapse
Affiliation(s)
- R Gold
- Neurologische Klinik, St. Josef Hospital, Klinikum der Ruhr-Universität, Gudrunstrasse 56, 44791, Bochum, Germany.
| | | |
Collapse
|