1
|
Aharoni R, Milo R, Arnon R. Glatiramer Acetate for the Treatment of Multiple Sclerosis: From First-Generation Therapy to Elucidation of Immunomodulation and Repair. Pharmacol Rev 2024; 76:1133-1158. [PMID: 39406508 DOI: 10.1124/pharmrev.124.000927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 10/18/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS), with a putative autoimmune origin and complex pathogenesis. Modification of the natural history of MS by reducing relapses and slowing disability accumulation was first attained in the 1990 s with the development of the first-generation disease-modifying therapies. Glatiramer acetate (GA), a copolymer of L-alanine, L-lysine, L-glutamic acid, and L-tyrosine, was discovered due to its ability to suppress the animal model of MS, experimental autoimmune encephalomyelitis. Extensive clinical trials and long-term assessments established the efficacy and the safety of GA. Furthermore, studies of the therapeutic processes induced by GA in animal models and in MS patients indicate that GA affects various levels of the innate and the adaptive immune response, generating deviation from proinflammatory to anti-inflammatory pathways. This includes competition for binding to antigen presenting cells; driving dendritic cells, monocytes, and B-cells toward anti-inflammatory responses; and stimulating T-helper 2 and T-regulatory cells. The immune cells stimulated by GA reach the CNS and secrete in situ anti-inflammatory cytokines alleviating the pathological processes. Furthermore, cumulative findings reveal that in addition to its immunomodulatory effect, GA promotes neuroprotective repair processes such as neurotrophic factors secretion, remyelination, and neurogenesis. This review aims to provide an overview of MS pathology diagnosis and treatment as well as the diverse mechanism of action of GA. SIGNIFICANCE STATEMENT: Understanding the complex MS immune pathogenesis provided multiple targets for therapeutic intervention, resulting in a plethora of agents, with various mechanisms of action, efficacy, and safety profiles. However, promoting repair beyond the body's limited spontaneous extent is still a major challenge. GA, one of the first approved disease-modifying therapies, induces diverse immunomodulatory effects. Furthermore, GA treatment results in elevated neurotrophic factors secretion, remyelination and neurogenesis, supporting the notion that immunomodulatory treatment can support in situ a growth-promoting and repair environment.
Collapse
Affiliation(s)
- Rina Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ron Milo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| | - Ruth Arnon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel (Ri.A., Ru.A.); and Department of Neurology, Barzilai Medical Center, Ashkelon, Israel, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel (R.M.)
| |
Collapse
|
2
|
Rahiman N, Zamani P, Arabi L, Alavizadeh SH, Nikpoor A, Mashreghi M, Badiee A, Jaafari MR. Novel liposomal glatiramer acetate: Preparation and immunomodulatory evaluation in murine model of multiple sclerosis. Int J Pharm 2023; 648:123620. [PMID: 37981250 DOI: 10.1016/j.ijpharm.2023.123620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/23/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
The frequent administration rate required for Glatiramer acetate (GA), a first-line therapy for Multiple sclerosis (MS), poses patient compliance issues. Only a small portion of the subcutaneously administered GA is available for phagocytosis by macrophages, as most of it is hydrolyzed at its administration site or excreted renally. To unravel these hurdles, we have prepared liposomal formulations of GA through thin film-hydration method plus extrusion. The clinical and histopathological efficacy of GA-loaded liposomes were assessed in prophylactic and therapeutic manners on murine model of MS (experimental autoimmune encephalomyelitis (EAE)). The selected GA liposomal formulation showed favorable size (275 nm on average), high loading efficiency, and high macrophage localization. Moreover, administration of GA-liposomes in mice robustly suppressed the inflammatory responses and decreased the inflammatory and demyelinated lesion regions in CNS compared to the free GA with subsequent reduction of the EAE clinical score. Our study indicated that liposomal GA could be served as a reliable nanomedicine-based platform to hopefully curb MS-related aberrant autoreactive immune responses with higher efficacy, longer duration of action, fewer administration frequencies, and higher delivery rate to macrophages. This platform has the potential to be introduced as a vaccine for MS after clinical translation and merits further investigations.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aminreza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Zhang G, Yang F, Li J, Chen S, Kong Y, Mo C, Leng X, Liu Y, Xu Y, Wang Y. A quinazoline derivative suppresses B cell hyper-activation and ameliorates the severity of systemic lupus erythematosus in mice. Front Pharmacol 2023; 14:1159075. [PMID: 37256224 PMCID: PMC10225574 DOI: 10.3389/fphar.2023.1159075] [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: 02/14/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Background: Aberrant autoreactive B cell responses contribute to the pathogenesis of systemic lupus erythematosus (SLE). Currently, there is no safe and effective drug for intervention of SLE. Quinazoline derivative (N4-(4-phenoxyphenethyl)quinazoline-4,6-diamine, QNZ) is a NF-κB inhibitor and has potent anti-inflammatory activity. However, it is unclear whether QNZ treatment can modulate B cell activation and SLE severity. Methods: Splenic CD19+ B cells were treated with QNZ (2, 10, or 50 nM) or paeoniflorin (200 μM, a positive control), and their activation and antigen presentation function-related molecule expression were examined by flow cytometry. MRL/lpr lupus-prone mice were randomized and treated intraperitoneally with vehicle alone, 0.2 mg/kg/d QNZ or 1 mg/kg/d FK-506 (tacrolimus, a positive control) for 8 weeks. Their body weights and clinical symptoms were measured and the frequency of different subsets of splenic and lymph node activated B cells were quantified by flow cytometry. The degrees of kidney inflammation and glycogen deposition were examined by hematoxylin and eosin (H&E) and PAS staining. The levels of serum autoantibodies and renal IgG, complement C3 deposition were examined by ELISA and immunofluorescence. Results: QNZ treatment significantly inhibited the activation and antigen presentation-related molecule expression of B cells in vitro. Similarly, treatment with QNZ significantly mitigated the SLE activity by reducing the frequency of activated B cells and plasma cells in MRL/lpr mice. Conclusion: QNZ treatment ameliorated the severity of SLE in MRL/lpr mice, which may be associated with inhibiting B cell activation, and plasma cell formation. QNZ may be an excellent candidate for the treatment of SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Gan Zhang
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Fan Yang
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Juan Li
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Shan Chen
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Yuhang Kong
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Chunfen Mo
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Xiao Leng
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Yang Liu
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Ying Xu
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
| | - Yantang Wang
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
- Department of Pharmacology, School of Pharmacy, Chengdu Medical College, Chengdu, China
| |
Collapse
|
4
|
Alomar HA, Nadeem A, Ansari MA, Attia SM, Bakheet SA, Al-Mazroua HA, Alhazzani K, Assiri MA, Alqinyah M, Almudimeegh S, Ahmad SF. Mitogen-activated protein kinase inhibitor PD98059 improves neuroimmune dysfunction in experimental autoimmune encephalomyelitis in SJL/J mice through the inhibition of nuclear factor-kappa B signaling in B cells. Brain Res Bull 2023; 194:45-53. [PMID: 36646144 DOI: 10.1016/j.brainresbull.2023.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Multiple sclerosis (MS) is a severe autoimmune disease leading to demyelination, followed by consequent axonal degeneration, causing sensory, motor, cognitive, and visual symptoms. Experimental autoimmune encephalomyelitis (EAE) is the most well-studied animal model of MS. Most current MS treatments are not completely effective, and severe side effects remain a great challenge. In this study, we report the therapeutic efficacy of PD98059, a potent mitogen-activated protein kinase inhibitor, on proteolipid protein (PLP)139-151-induced EAE in SJL/J mice. Following the induction of EAE, mice were intraperitoneally treated with PD98059 (5 mg/kg for 14 days) daily from day 14 to day 28. This study investigated the effects of PD98059 on C-C motif chemokine receptor 6 (CCR6), CD14, NF-κB p65, IκBα, GM-CSF, iNOS, IL-6, TNF-α in CD45R+ B lymphocytes using flow cytometry. Furthermore, we analyzed the effect of PD98059 on CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA and protein expression levels using qRT-PCR analysis in brain tissues. Mechanistic investigations revealed that PD98059-treated in mice with EAE had reduced CD45R+CCR6+, CD45R+CD14+, CD45R+NF-κB p65+, CD45R+GM-CSF+, CD45R+iNOS+, CD45R+IL-6+, and CD45R+TNF-α+ cells and increased CD45R+IκBα+ cells compared with vehicle-treated control mice in the spleen. Moreover, downregulation of CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA expression level was observed in PD98059-treated mice with EAE compared with vehicle-treated control mice in the brain tissue. The results of this study demonstrate that PD98059 modulates inflammatory mediators through multiple cellular mechanisms. The results of this study suggest that PD98059 may be pursued as a therapeutic agent for the treatment of MS.
Collapse
Affiliation(s)
- Hatun A Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
5
|
Wang J, Song X, Tan G, Sun P, Guo L, Zhang N, Wang J, Li B. NAD+ improved experimental autoimmune encephalomyelitis by regulating SIRT1 to inhibit PI3K/Akt/mTOR signaling pathway. Aging (Albany NY) 2021; 13:25931-25943. [PMID: 34928817 PMCID: PMC8751589 DOI: 10.18632/aging.203781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/22/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To investigate the effect of NAD+ on thymus autophagy in experimental autoimmune encephalomyelitis (EAE) mice through SIRT1. METHODS Bioinformatic analysis was used to identify hub genes. Forty female C57BL/6 mice were randomly divided into 4 groups: control, EAE, NAD+, and NAD+ +SIRT1 inhibitor (SIRT-IN-3) groups and SIRT1 group. The NAD+ group and SIRT1 inhibitor group were treated with NAD+ drug and fed for 4 weeks. The neurological function scores were evaluated weekly. The thymus tissues of wild-type mice were removed, ground and filtered into single-cell suspension. MOG 35-55 (1 μg/mL) was given to primary thymic epithelial cells (TECs) to induce EAE model in vitro. The expression of LC-3A/B was observed by immunofluorescence. The expressions or the activation/phosphorylation of associated proteins were detected by Western blot. RESULTS Enrichment analysis showed PI3K-Akt-mTOR and autophagy pathway were main terms in EAE diseases, and the relationship between NAD+ and SIRT1. The activation of p-PI3K, p-Akt and p-mTOR were the highest in the EAE group consistent with decreased P62, Beclin1, LC-3A/B and SIRT1, and NAD+ reversed these results, furthermore SIRT1 inhibitor: SIRT-IN3 weakened the NAD+' effects in both in vivo and in vitro experiments. Immunofluorescence study in vivo and in vitro were accord with the results of western blot. CONCLUSIONS NAD+ exerted a protective effect on EAE mice by inhibiting PI3K/Akt/mTOR signaling pathway through SIRT1 in TECs, and prevented EAE mice from sustained damage.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Xueqin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Guojun Tan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Pengtao Sun
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Li Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Ning Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Jueqiong Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei, China
| | - Bin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang 050051, Hebei, China
| |
Collapse
|
6
|
Sun H, Wang J, Guo L, Wang Y, Zhang J, Wang J, Quan M, Li B. The combined treatment of NAD + and atorvastatin ameliorates the development of experimental autoimmune encephalomyelitis in C57BL/6 mice. J Neuroimmunol 2020; 350:577429. [PMID: 33176238 DOI: 10.1016/j.jneuroim.2020.577429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 10/08/2020] [Accepted: 10/22/2020] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is a demyelinating and degenerating disorder of the central nervous system impacting many patients worldwide. Due to the complex pathogenesis of MS, drugs to treat MS often show partial effectiveness. Earlier experiments have demonstrated that both atorvastatin and nicotinamide adenine dinucleotide (NAD+) may ameliorate experimental autoimmune encephalomyelitis (EAE), which is known as a classical model of MS, via different protective mechanisms. Since combination therapy can exhibit more beneficial effects than monotherapy, we observed the protective effects of combined treatment of atorvastatin and NAD+ in EAE mice. Our results showed that the combined treatment of these two drugs at half of their optimal dosages had synergistic effect to alleviate the severity of EAE in mice than treatment with each alone. The combined treatment of EAE mice with atorvastatin plus NAD+ relieved the clinical signs and histologic changes, increased the proportion of Treg cells, attenuated IL-17 production, reduced proinflammatory cytokine secretion of Th17 cells, and increased cytokine secretion of Treg cells. In addition, the combined treatment also diminished phosphorylation of both P38 MAPK and ERK, while elevated SIRT1 expression. Taken together, these data suggested that combined treatment with NAD+ and atorvastatin could attenuate the progression of EAE by synergistic immune regulation.
Collapse
Affiliation(s)
- Huanhuan Sun
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jueqiong Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Li Guo
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Ying Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jing Zhang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Jinli Wang
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Moyuan Quan
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China
| | - Bin Li
- Department of Neurology, Neurological Laboratory of Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China.
| |
Collapse
|
7
|
Song Z, Khaw YM, Pacheco LA, Tseng KY, Tan Z, Cai K, Ponnusamy E, Cheng J, Inoue M. Induction of a higher-ordered architecture in glatiramer acetate improves its biological efficiency in an animal model of multiple sclerosis. Biomater Sci 2020; 8:5271-5281. [PMID: 32744547 DOI: 10.1039/d0bm00957a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Glatiramer acetate (GA), a linear random copolypeptide, is a first-line treatment for multiple sclerosis (MS). A major concern, however, is that GA treatment is associated with adverse effects and poor patient adherence due to the need for frequent injections. Here we describe improved performance of the polymeric drug, even at low doses with less-frequent injections, through the modification of its architecture into a star-shaped GA (sGA). In a sGA, multiple GAs are covalently linked onto a core, which greatly changes their properties such as molecular weight, size, and shape. The spherical sGA is retained longer in the body after intraperitoneal injection, and is more readily internalized by RAW 264.7 macrophage cells and bone marrow-derived dendritic cells than GA. In C57BL/6 mice induced with experimental autoimmune encephalitis, a mouse model for MS, sGA treatment exerts disease amelioration effect that is significantly better than that of GA despite a lower dose and less frequent injection. Moreover, spinal cord pathologies of demyelination and leukocyte infiltration are dramatically less pronounced in the sGA treatment condition compared to the GA treatment condition. Thus, we propose that sGA with a higher-ordered architecture offers an attractive and potentially viable treatment option for MS patients.
Collapse
Affiliation(s)
- Ziyuan Song
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA. and Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Yee Ming Khaw
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA. and University of Illinois at Urbana-Champaign, Neuroscience Program, 405 North Matthews Avenue, Urbana, Illinois 61801, USA
| | - Lazaro A Pacheco
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Kuan-Ying Tseng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Zhengzhong Tan
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Kaimin Cai
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | | | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Makoto Inoue
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA. and University of Illinois at Urbana-Champaign, Neuroscience Program, 405 North Matthews Avenue, Urbana, Illinois 61801, USA
| |
Collapse
|
8
|
Aharoni R, Eilam R, Schottlender N, Radomir L, Leistner-Segal S, Feferman T, Hirsch D, Sela M, Arnon R. Glatiramer acetate increases T- and B -regulatory cells and decreases granulocyte-macrophage colony-stimulating factor (GM-CSF) in an animal model of multiple sclerosis. J Neuroimmunol 2020; 345:577281. [DOI: 10.1016/j.jneuroim.2020.577281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 01/21/2023]
|
9
|
Planchais C, Rayes J, Delignat S, Pashova S, Varthaman A, Pashov A, Bayry J, Kaveri SV, Dimitrov JD, Lacroix-Desmazes S. Stimulation with FITC-labeled antigens confers B cells with regulatory properties. Cell Immunol 2020; 355:104151. [PMID: 32615414 DOI: 10.1016/j.cellimm.2020.104151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 10/24/2022]
Abstract
B cells with regulatory properties (Bregs) were identified in human and in mice among different B-cell subsets. Their regulatory properties rely mainly on the production of anti-inflammatory cytokines, in particular IL10, IL-35 and TGFβ, and were extensively studied in mouse models of autoimmune and inflammatory diseases. However, the exact nature of the stimulatory signals conferring regulatory properties to B cells is still not clear. We serendipitously observed that fluorescein isothiocyanate (FITC) binds to a significant proportion of naïve mouse B cells. Binding of FITC to the B-cell surface implicated at least in part the B-cell receptor. It triggered IL-10 production and allowed the endocytosis of FITC-coupled antigens followed by their presentation to CD4+ T cells. In particular, B cells incubated with FITC-OVA polarized OTII T cells towards a Tr1/Th2 phenotype in vitro. Further, the adoptive transfer of B cells incubated with FITC-labeled myelin oligodendrocyte glycoprotein peptide protected mice from experimental autoimmune encephalomyelitis, a T-cell-dependent autoimmune model. Together, the data show that FITC-stimulated B cells polarize immune responses towards Tr1/Th2 and acquire immuno-modulatory properties.
Collapse
Affiliation(s)
- Cyril Planchais
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France.
| | - Julie Rayes
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Sandrine Delignat
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Shina Pashova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Anastas Pashov
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Jagadeesh Bayry
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Srinivas V Kaveri
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Jordan D Dimitrov
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| | - Sebastien Lacroix-Desmazes
- Centre de recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France
| |
Collapse
|
10
|
Häusler D, Hajiyeva Z, Traub JW, Zamvil SS, Lalive PH, Brück W, Weber MS. Glatiramer acetate immune modulates B-cell antigen presentation in treatment of MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:e698. [PMID: 32184341 PMCID: PMC7136047 DOI: 10.1212/nxi.0000000000000698] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/31/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE We examined the effect of glatiramer acetate (GA) on B-cell maturation, differentiation, and antigen presentation in MS and experimental autoimmune encephalomyelitis (EAE). METHODS A cross-sectional study of blood samples from 20 GA-treated and 18 untreated patients with MS was performed by flow cytometry; 6 GA-treated patients with MS were analyzed longitudinally. GA-mediated effects on B-cell antigen-presenting function were investigated in EAE, or, alternatively, B cells were treated with GA in vitro using vehicle as a control. RESULTS In MS, GA diminished transitional B-cell and plasmablast frequency, downregulated CD69, CD25, and CD95 expression, and decreased TNF-α production, whereas IL-10 secretion and MHC Class II expression were increased. In EAE, we observed an equivalent dampening of proinflammatory B-cell properties and an enhanced expression of MHC Class II. When used as antigen-presenting cells for activation of naive T cells, GA-treated B cells promoted development of regulatory T cells, whereas proinflammatory T-cell differentiation was diminished. CONCLUSIONS GA immune modulates B-cell function in EAE and MS and efficiently interferes with pathogenic B cell-T cell interaction.
Collapse
Affiliation(s)
- Darius Häusler
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Zivar Hajiyeva
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Jan W Traub
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Scott S Zamvil
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Patrice H Lalive
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Wolfgang Brück
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland
| | - Martin S Weber
- From the Institute of Neuropathology (D.H., J.W.T., W.B., M.S.W.), University Medical Center; Department of Neurology (Z.H., J.W.T., M.S.W.), University Medical Center, Göttingen, Germany; Department of Neurology (S.S.Z.), University of California, San Francisco; Division of Neurology (P.H.L.), Department of Neurosciences, Hospital and University of Geneva; and Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, Geneva, Switzerland.
| |
Collapse
|
11
|
Nezamoleslami S, Sheibani M, Dehpour AR, Mobasheran P, Shafaroodi H. Glatiramer acetate attenuates renal ischemia reperfusion injury in rat model. Exp Mol Pathol 2019; 112:104329. [PMID: 31697931 DOI: 10.1016/j.yexmp.2019.104329] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/28/2019] [Accepted: 11/03/2019] [Indexed: 12/13/2022]
Abstract
Chronic renal failure can ultimately lead to kidney transplantation. Renal transplantation is associated with ischemia-reperfusion injury (I/R).2 The subsequent processes of kidney I/R can lead to irreversible damages to the kidney tissue. Glatiramer acetate is an immunomodulatory drug for the treatment of multiple sclerosis (MS) and the anti-inflammatory effects of this drug have already been proven in some inflammatory models. The purpose of this study was to evaluate the protective effects of Glatiramer on reducing the damages arising from kidney ischemia-reperfusion. In this study, 35 Wistar rats were used which divided into 5 groups: sham, control (I/R), I/R + Glatiramer 0.5 mg/kg, I/R + Glatiramer 1 mg/kg, I/R + Glatiramer 2 mg/kg. Renal arteries were clamped bilaterally for 45 min, then the clamps were removed and the reperfusion process continued to 24 h. In the following, serum and kidneys were separated for analysis. In the control group, serum levels of LDH, inflammatory factor TNF-α and renal functional markers such as BUN and Creatinine were remarkably increased, but in the treatment groups, especially in Glatiramer 2 mg/kg received group, a significant decrease in these factors was observed. Tissue concentration of MDA was reduced following Glatiramer treatment. Besides, Glatiramer attenuated the increased kidney level of NF-κB protein using immunohistochemical assay. NFkB migration to the nucleolus increases inflammatory cytokines production. The anti-inflammatory factor, IL-10, in serum was significantly increased in the treatment group of Glatiramer 2 mg/kg. Furthermore, Glatiramer decreased renal tissue injury score according to the histopathological study. These results demonstrate that Glatiramer may play protective effects in kidney ischemia-reperfusion injury by reducing inflammatory and oxidative damages.
Collapse
Affiliation(s)
- Sadaf Nezamoleslami
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran
| | - Parnia Mobasheran
- Islamic Azad University of Tehran Medical Unit, Faculty of pharmaceutical science, P.O. Box 1916893813, Iran
| | - Hamed Shafaroodi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran; Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, P.O. Box 13145-784, Iran.
| |
Collapse
|
12
|
Fan KR, Baskaran M, Nongpiur ME, Htoon HM, de Leon JMS, Perera SA, Belkin M, Aung T. Investigating the neuroprotective effect of Copolymer-1 in acute primary angle closure - Interim report of a randomized placebo-controlled double-masked clinical trial. Acta Ophthalmol 2019; 97:e827-e832. [PMID: 30916898 DOI: 10.1111/aos.14099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/04/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE To investigate the neuroprotective effect of Copolymer-1 (Cop-1) in patients with acute primary angle closure (APAC) in a randomized double-masked controlled trial. METHODS After initial medical management, APAC patients were randomized to receive either subcutaneous Cop-1 or placebo within 24 hr and at 1 week. After laser peripheral iridotomy (LPI), subjects underwent serial visual field (VF) tests and retinal nerve fibre layer (RNFL) thickness measurements with spectral-domain optical coherence tomography. The primary outcome measure was mean number of progressing points (significant slope of ≥ 1 dB per year sensitivity loss) over 16 weeks based on pointwise linear regression analysis, and the secondary outcome measure was the change in RNFL thickness. RESULTS Thirty-eight patients (19 in each group) completed the study. Twenty-five (65.8%) were female, the majority being Chinese (86.8%) with mean age 62.5 years (SD 8.1). Patients in the Cop-1 group were found to have mean of 0.32 (SD 0.95) progressing points compared to 2.74 (SD 5.31) in the placebo group (p = 0.09), while 3/19 (15.8%) of Cop-1 treated patients had 1 or more progressing points compared to 7/19 (36.8%) in the placebo group (p = 0.14). There was no difference in change of RNFL thickness between groups (p = 0.57). We found improvement of mean deviation (MD) at week 16 in the Cop-1 group (p = 0.01) compared to worsening of MD in the placebo group (p = 0.04). CONCLUSION After APAC, there was no difference in VF progression (or RNFL thickness change) between Cop-1 and placebo groups. However, there was improvement of MD in Cop-1 treated patients.
Collapse
Affiliation(s)
- Kenric Rui‐Pin Fan
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
| | - Mani Baskaran
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
- EYE‐ACP Duke‐NUS Medical School Singapore City Singapore
| | - Monisha E. Nongpiur
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
- EYE‐ACP Duke‐NUS Medical School Singapore City Singapore
| | - Hla Mynt Htoon
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
- EYE‐ACP Duke‐NUS Medical School Singapore City Singapore
| | - John Mark S. de Leon
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
| | - Shamira A. Perera
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
| | - Michael Belkin
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
- Tel Aviv University Tel Aviv Israel
| | - Tin Aung
- Singapore Eye Research Institute and Singapore National Eye Center Singapore City Singapore
- EYE‐ACP Duke‐NUS Medical School Singapore City Singapore
- Yong Loo Lin School of Medicine National University of Singapore City Singapore
| |
Collapse
|
13
|
Understanding regulatory B cells in autoimmune diseases: the case of multiple sclerosis. Curr Opin Immunol 2019; 61:26-32. [PMID: 31445312 DOI: 10.1016/j.coi.2019.07.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 01/04/2023]
Abstract
The suppressive function of B cells is mediated mostly through their provision of cytokines with anti-inflammatory properties, in particular interleukin-10. This B cell activity has been convincingly described in mice with autoimmune, infectious, as well as malignant diseases, and evidence is accumulating of its relevance in human. This review provides a personal view of this B cell function using multiple sclerosis and its animal model experimental autoimmune encephalomyelitis as representative examples, in an attempt to bridge observations obtained in mice and human, with the goal of providing a coherent transversal framework to further explore this field, and eventually manipulate this B cell function therapeutically.
Collapse
|
14
|
Valizadeh A, Sanaei R, Rezaei N, Azizi G, Fekrvand S, Aghamohammadi A, Yazdani R. Potential role of regulatory B cells in immunological diseases. Immunol Lett 2019; 215:48-59. [PMID: 31442542 DOI: 10.1016/j.imlet.2019.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/04/2019] [Accepted: 08/20/2019] [Indexed: 12/21/2022]
Abstract
Regulatory B cells (Bregs) are immune-modulating cells that affect the immune system by producing cytokines or cellular interactions. These cells have immunomodulatory effects on the immune system by cytokine production. The abnormalities in Bregs could be involved in various disorders such as autoimmunity, chronic infectious disease, malignancies, allergies, and primary immunodeficiencies are immune-related scenarios. Ongoing investigation could disclose the biology and the exact phenotype of these cells and also the assigned mechanisms of action of each subset, as a result, potential therapeutic strategies for treating immune-related anomalies. In this review, we collect the findings of human and mouse Bregs and the therapeutic efforts to change the pathogenicity of these cells in diverse disease.
Collapse
Affiliation(s)
- Amir Valizadeh
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Roozbeh Sanaei
- Immunology Research Center (IRC), Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
15
|
Tsareva EY, Favorova OO, Boyko AN, Kulakova OG. Genetic Markers for Personalized Therapy of Polygenic Diseases: Pharmacogenetics of Multiple Sclerosis. Mol Biol 2019. [DOI: 10.1134/s0026893319040149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Li R, Bar-Or A. The Multiple Roles of B Cells in Multiple Sclerosis and Their Implications in Multiple Sclerosis Therapies. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a029108. [PMID: 29661809 DOI: 10.1101/cshperspect.a029108] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Increasing evidence has suggested that both antibody-dependent and antibody-independent functions of B cells are involved in multiple sclerosis (MS). The contrasting results of distinct B-cell targeting therapies in MS patients underscores the importance of elucidating these multiple B-cell functions. In this review, we discuss the generation of autoreactive B cells, migration of B cells into the central nervous system (CNS), and how different functions of B cells may contribute to MS disease activity and potentially mitigation in both the periphery and CNS compartments. In addition, we propose several future therapeutic strategies that may better target/shape B-cell responses for long-term treatment of MS.
Collapse
Affiliation(s)
- Rui Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Amit Bar-Or
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
17
|
Van Kaer L, Postoak JL, Wang C, Yang G, Wu L. Innate, innate-like and adaptive lymphocytes in the pathogenesis of MS and EAE. Cell Mol Immunol 2019; 16:531-539. [PMID: 30874627 DOI: 10.1038/s41423-019-0221-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) in which the immune system damages the protective insulation surrounding the nerve fibers that project from neurons. A hallmark of MS and its animal model, experimental autoimmune encephalomyelitis (EAE), is autoimmunity against proteins of the myelin sheath. Most studies in this field have focused on the roles of CD4+ T lymphocytes, which form part of the adaptive immune system as both mediators and regulators in disease pathogenesis. Consequently, the treatments for MS often target the inflammatory CD4+ T-cell responses. However, many other lymphocyte subsets contribute to the pathophysiology of MS and EAE, and these subsets include CD8+ T cells and B cells of the adaptive immune system, lymphocytes of the innate immune system such as natural killer cells, and subsets of innate-like T and B lymphocytes such as γδ T cells, natural killer T cells, and mucosal-associated invariant T cells. Several of these lymphocyte subsets can act as mediators of CNS inflammation, whereas others exhibit immunoregulatory functions in disease. Importantly, the efficacy of some MS treatments might be mediated in part by effects on lymphocytes other than CD4+ T cells. Here we review the contributions of distinct subsets of lymphocytes on the pathogenesis of MS and EAE, with an emphasis on lymphocytes other than CD4+ T cells. A better understanding of the distinct lymphocyte subsets that contribute to the pathophysiology of MS and its experimental models will inform the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| | - Joshua L Postoak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Chuan Wang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Guan Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| |
Collapse
|
18
|
Amrouche K, Pers JO, Jamin C. Glatiramer Acetate Stimulates Regulatory B Cell Functions. THE JOURNAL OF IMMUNOLOGY 2019; 202:1970-1980. [DOI: 10.4049/jimmunol.1801235] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/07/2019] [Indexed: 01/14/2023]
|
19
|
Prod'homme T, Zamvil SS. The Evolving Mechanisms of Action of Glatiramer Acetate. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a029249. [PMID: 29440323 DOI: 10.1101/cshperspect.a029249] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Glatiramer acetate (GA) is a synthetic amino acid copolymer that is approved for treatment of relapsing remitting multiple sclerosis (RRMS) and clinically isolated syndrome (CIS). GA reduces multiple sclerosis (MS) disease activity and has shown comparable efficacy with high-dose interferon-β. The mechanism of action (MOA) of GA has long been an enigma. Originally, it was recognized that GA treatment promoted expansion of GA-reactive T-helper 2 and regulatory T cells, and induced the release of neurotrophic factors. However, GA treatment influences both innate and adaptive immune compartments, and it is now recognized that antigen-presenting cells (APCs) are the initial cellular targets for GA. The anti-inflammatory (M2) APCs induced following treatment with GA are responsible for the induction of anti-inflammatory T cells that contribute to its therapeutic benefit. Here, we review studies that have shaped our current understanding of the MOA of GA.
Collapse
Affiliation(s)
| | - Scott S Zamvil
- Department of Neurology and Program in Immunology, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
20
|
Abstract
B cells play a vital function in multiple sclerosis (MS) pathogenesis through an array of effector functions. All currently approved MS disease-modifying therapies alter the frequency, phenotype, or homing of B cells in one way or another. The importance of this mechanism of action has been reinforced with the successful development and clinical testing of B-cell-depleting monoclonal antibodies that target the CD20 surface antigen. Ocrelizumab, a humanized anti-CD20 monoclonal antibody, was approved by the Food and Drug Administration (FDA) in March 2017 after pivotal trials showed dramatic reductions in inflammatory disease activity in relapsing MS as well as lessening of disability progression in primary progressive MS. These and other clinical studies place B cells at the center of the inflammatory cascade in MS and provide a launching point for development of therapies that target selective pathogenic B-cell populations.
Collapse
Affiliation(s)
- Joseph J Sabatino
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| | - Scott S Zamvil
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| | - Stephen L Hauser
- Multiple Sclerosis Center, Department of Neurology, University of California, San Francisco, California 94158
| |
Collapse
|
21
|
Regulatory B and T lymphocytes in multiple sclerosis: friends or foes? AUTOIMMUNITY HIGHLIGHTS 2018; 9:9. [PMID: 30415321 PMCID: PMC6230324 DOI: 10.1007/s13317-018-0109-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Current clinical experience with immunomodulatory agents and monoclonal antibodies in principle has established the benefit of depleting lymphocytic populations in relapsing–remitting multiple sclerosis (RRMS). B and T cells may exert multiple pro-inflammatory actions, but also possess regulatory functions making their role in RRMS pathogenesis much more complex. There is no clear correlation of Tregs and Bregs with clinical features of the disease. Herein, we discuss the emerging data on regulatory T and B cell subset distributions in MS and their roles in the pathophysiology of MS and its murine model, experimental autoimmune encephalomyelitis (EAE). In addition, we summarize the immunomodulatory properties of certain MS therapeutic agents through their effect on such regulatory cell subsets and their relevance to clinical outcomes.
Collapse
|
22
|
Negron A, Robinson RR, Stüve O, Forsthuber TG. The role of B cells in multiple sclerosis: Current and future therapies. Cell Immunol 2018; 339:10-23. [PMID: 31130183 DOI: 10.1016/j.cellimm.2018.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
While it was long held that T cells were the primary mediators of multiple sclerosis (MS) pathogenesis, the beneficial effects observed in response to treatment with Rituximab (RTX), a monoclonal antibody (mAb) targeting CD20, shed light on a key contributor to MS that had been previously underappreciated: B cells. This has been reaffirmed by results from clinical trials testing the efficacy of subsequently developed B cell-depleting mAbs targeting CD20 as well as studies revisiting the effects of previous disease-modifying therapies (DMTs) on B cell subsets thought to modulate disease severity. In this review, we summarize current knowledge regarding the complex roles of B cells in MS pathogenesis and current and potential future B cell-directed therapies.
Collapse
Affiliation(s)
- Austin Negron
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Rachel R Robinson
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA; Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| | | |
Collapse
|
23
|
Hartwell BL, Pickens CJ, Leon M, Northrup L, Christopher MA, Griffin JD, Martinez-Becerra F, Berkland C. Soluble antigen arrays disarm antigen-specific B cells to promote lasting immune tolerance in experimental autoimmune encephalomyelitis. J Autoimmun 2018; 93:76-88. [PMID: 30007842 PMCID: PMC6117839 DOI: 10.1016/j.jaut.2018.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022]
Abstract
Autoreactive lymphocytes that escape central immune tolerance may be silenced via an endogenous peripheral tolerance mechanism known as anergy. Antigen-specific therapies capable of inducing anergy may restore patients with autoimmune diseases to a healthy phenotype while avoiding deleterious side effects associated with global immunosuppression. Inducing anergy in B cells may be a particularly potent intervention, as B cells can contribute to autoimmune diseases through multiple mechanisms and offer the potential for direct antigen-specific targeting through the B cell receptor (BCR). Our previous results suggested autoreactive B cells may be silenced by multivalent 'soluble antigen arrays' (SAgAs), which are polymer conjugates displaying multiple copies of autoantigen with or without a secondary peptide that blocks intracellular cell-adhesion molecule-1 (ICAM-1). Here, key therapeutic molecular properties of SAgAs were identified and linked to the immunological mechanism through comprehensive cellular and in vivo analyses. We determined non-hydrolyzable 'cSAgAs' displaying multivalent 'click'-conjugated antigen more potently suppressed experimental autoimmune encephalomyelitis (EAE) compared to hydrolyzable SAgAs capable of releasing conjugated antigen. cSAgAs restored a healthy phenotype in disease-specific antigen presenting cells (APCs) by inducing an anergic response in B cells and a subset of B cells called autoimmune-associated B cells (ABCs) that act as potent APCs in autoimmune disease. Accompanied by a cytokine response skewed towards a Th2/regulatory phenotype, this generated an environment of autoantigenic tolerance. By identifying key therapeutic molecular properties and an immunological mechanism that drives SAgA efficacy, this work guides the design of antigen-specific immunotherapies capable of inducing anergy.
Collapse
MESH Headings
- Animals
- Autoantigens/genetics
- Autoantigens/immunology
- B-Lymphocyte Subsets/drug effects
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/pathology
- Click Chemistry
- Clonal Anergy/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Hydrolysis
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacology
- Immunotherapy/methods
- Injections, Subcutaneous
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/immunology
- Mice
- Myelin Proteolipid Protein/administration & dosage
- Peptide Fragments/administration & dosage
- Peptide Fragments/chemical synthesis
- Peptide Fragments/immunology
- Peptide Fragments/pharmacology
- Protein Array Analysis
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Spleen/immunology
- Spleen/pathology
- Th2 Cells/immunology
- Th2 Cells/pathology
Collapse
Affiliation(s)
- Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA
| | - Chad J Pickens
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - Martin Leon
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS 66045, USA
| | - Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - J Daniel Griffin
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA
| | - Francisco Martinez-Becerra
- Immunology Core Laboratory of the Kansas Vaccine Institute, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, USA
| | - Cory Berkland
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA; Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA; Department of Chemical and Petroleum Engineering, University of Kansas, 1530 West 15th Street, Lawrence, KS 66045, USA.
| |
Collapse
|
24
|
Abstract
Growing evidence indicates that B cells play a key role in the pathogenesis of multiple sclerosis (MS). B cells occupy distinct central nervous system (CNS) compartments in MS, including the cerebrospinal fluid and white matter lesions. Also, it is now known that, in addition to entering the CNS, B cells can circulate into the periphery via a functional lymphatic system. Data suggest that the role of B cells in MS mainly involves their in situ activation in demyelinating lesions, leading to altered pro- and anti-inflammatory cytokine secretion, and a highly effective antigen-presenting cell function, resulting in activation of memory or naïve T cells. Clinically, B cell-depleting agents show significant efficacy in MS. In addition, many disease-modifying therapies (DMTs) traditionally understood to target T cells are now known to influence B cell number and function. One of the earliest DMTs to be developed, glatiramer acetate (GA), has been shown to reduce the total frequency of B cells, plasmablasts, and memory B cells. It also appears to promote a shift toward reduced inflammation by increasing anti-inflammatory cytokine release and/or reducing pro-inflammatory cytokine release by B cells. In the authors' opinion, this may be mediated by cross-reactivity of B cell receptors for GA with antigen (possibly myelin basic protein) expressed in the MS lesion. More research is required to further characterize the role of B cells and their bidirectional trafficking in the pathogenesis of MS. This may uncover novel targets for MS treatments and facilitate the development of B cell biomarkers of drug response.
Collapse
|
25
|
Fillatreau S. B cells and their cytokine activities implications in human diseases. Clin Immunol 2018; 186:26-31. [PMID: 28736271 PMCID: PMC5844600 DOI: 10.1016/j.clim.2017.07.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/19/2022]
Abstract
B cells are the only cell type that can give rise to antibody-producing cells, and the only cell type whose selective depletion can, today, lead to an improvement of a wide range of immune-mediated inflammatory diseases, including disorders not primarily driven by autoantibodies. Here, I discuss this paradoxical observation, and propose that the capacity of B cells to act as cytokine-producing cells explains how they can control monocyte activity and subsequently disease pathogenesis. Together with current data on the effect of anti-CD20 B cell-depleting reagents in the clinic, this novel knowledge on B cell heterogeneity opens the way for novel safer and more efficient strategies to target B cells. The forthcoming identification of disease-relevant B cell subsets is awaited to permit their monitoring and specific targeting in a personalized medicine approach.
Collapse
Affiliation(s)
- Simon Fillatreau
- Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants Malades, Paris, France; Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Leibniz Institute, Berlin, Germany.
| |
Collapse
|
26
|
Amrouche K, Jamin C. Influence of drug molecules on regulatory B cells. Clin Immunol 2017; 184:1-10. [DOI: 10.1016/j.clim.2017.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/27/2017] [Indexed: 02/07/2023]
|
27
|
Staun-Ram E, Miller A. Effector and regulatory B cells in Multiple Sclerosis. Clin Immunol 2017; 184:11-25. [PMID: 28461106 DOI: 10.1016/j.clim.2017.04.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/27/2017] [Indexed: 12/21/2022]
Abstract
The role of B cells in the pathogenesis of Multiple Sclerosis (MS), an autoimmune neurodegenerative disease, is becoming eminent in recent years, but the specific contribution of the distinct B cell subsets remains to be elucidated. Several B cell subsets have shown regulatory, anti-inflammatory capacities in response to stimuli in vitro, as well as in the animal model of MS: Experimental Autoimmune Encephalomyelitis (EAE). However, the functional role of the B regulatory cells (Bregs) in vivo and specifically in the human disease is yet to be clarified. In the present review, we have summarized the updated information on the roles of effector and regulatory B cells in MS and the immune-modulatory effects of MS therapeutic agents on their phenotype and function.
Collapse
Affiliation(s)
- Elsebeth Staun-Ram
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ariel Miller
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Neuroimmunology Unit & Multiple Sclerosis Center, Carmel Medical Center, Haifa, Israel.
| |
Collapse
|
28
|
Yamamura T, Ashtamker N, Ladkani D, Fukazawa T, Houzen H, Tanaka M, Miura T, Knappertz V. Once-daily glatiramer acetate decreases magnetic resonance imaging disease activity in Japanese patients with relapsing-remitting multiple sclerosis. ACTA ACUST UNITED AC 2017; 8:129-137. [PMID: 28706565 PMCID: PMC5485168 DOI: 10.1111/cen3.12383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Multiple sclerosis (MS) prevalence, clinical patterns, and treatment responses vary between races and geographical latitudes. Glatiramer acetate (GA; Copaxone) has provided a safe, effective treatment option for relapsing-remitting MS patients in the USA, European nations, and other countries for decades. The objective of the present study was to assess the safety and efficacy of GA in reducing magnetic resonance imaging disease activity in Japanese patients with active relapsing-remitting MS. METHODS This phase 2, multicenter, open-label, single-arm, 52-week study measured the effect of GA 20 mg once-daily on magnetic resonance imaging disease activity. GA efficacy was evaluated through week 36, and safety through week 52. The primary end-point was change in the mean number of T1-weighted gadolinium-enhancing (GdE) lesions from pretreatment (weeks -8, -4 and baseline) to weeks 28, 32 and 36. Secondary end-points included a change in mean number of new T2-weighted lesions, GdE lesion and T2 lesion volumes, annualized relapse rate, and Expanded Disability Status Scale scores. RESULTS GA therapy reduced the number of new GdE lesions by 65.66% (95% CI 33.19-82.35%). The number of new T2 lesions and GdE lesion volume were also reduced from pretreatment. The annualized relapse rate was reduced by 42% compared with the 1 year before treatment. Changes in T2 lesion volume and Expanded Disability Status Scale scores were favorable, but less pronounced. Most common adverse events were injection-site reactions. CONCLUSIONS The present study confirmed the well-established safety, tolerability and efficacy profile of GA in Japanese MS patients.
Collapse
Affiliation(s)
- Takashi Yamamura
- Department of Immunology National Center of Neurology and Psychiatry National Institute of Neuroscience, and Multiple Sclerosis Center Tokyo Japan
| | - Natalia Ashtamker
- Research and Development Teva Pharmaceutical Industries Netanya Israel
| | - David Ladkani
- Research and Development Teva Pharmaceutical Industries Netanya Israel
| | | | - Hideki Houzen
- Department of Neurology Obihiro Kosei General Hospital Hokkaido Japan
| | - Masami Tanaka
- Multiple Sclerosis Center Kyoto Min-iren Chuo Hospital Kyoto Japan.,Department of Neurology Kaikoukai Josai Hospital Nagoya Japan.,Department of Neurology School of Medicine Fujita Health University Aichi Japan
| | - Toshiro Miura
- Research and Development Teva Pharmaceutical K.K. Tokyo Japan
| | - Volker Knappertz
- Research and Development Teva Pharmaceutical Industries Frazer PA USA.,Department of Neurology Heinrich Heine University Düsseldorf Germany
| |
Collapse
|
29
|
Ghaebi M, Nouri M, Ghasemzadeh A, Farzadi L, Jadidi-Niaragh F, Ahmadi M, Yousefi M. Immune regulatory network in successful pregnancy and reproductive failures. Biomed Pharmacother 2017; 88:61-73. [PMID: 28095355 DOI: 10.1016/j.biopha.2017.01.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/27/2016] [Accepted: 01/02/2017] [Indexed: 12/23/2022] Open
Abstract
Maternal immune system must tolerate semiallogenic fetus to establish and maintain a successful pregnancy. Despite the existence of several strategies of trophoblast to avoid recognition by maternal leukocytes, maternal immune system may react against paternal alloantigenes. Leukocytes are important components in decidua. Not only T helper (Th)1/Th2 balance, but also regulatory T (Treg) cells play an important role in pregnancy. Although the frequency of Tregs is elevated during normal pregnancies, their frequency and function are reduced in reproductive defects such as recurrent miscarriage and preeclampsia. Tregs are not the sole population of suppressive cells in the decidua. It has recently been shown that regulatory B10 (Breg) cells participate in pregnancy through secretion of IL-10 cytokine. Myeloid derived suppressor cells (MDSCs) are immature developing precursors of innate myeloid cells that are increased in pregnant women, implying their possible function in pregnancy. Natural killer T (NKT) cells are also detected in mouse and human decidua. They can also affect the fetomaternal tolerance. In this review, we will discuss on the role of different immune regulatory cells including Treg, γd T cell, Breg, MDSC, and NKT cells in pregnancy outcome.
Collapse
Affiliation(s)
- Mahnaz Ghaebi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliyeh Ghasemzadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laya Farzadi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Wang J, Zhao C, Kong P, Bian G, Sun Z, Sun Y, Guo L, Li B. Methylene blue alleviates experimental autoimmune encephalomyelitis by modulating AMPK/SIRT1 signaling pathway and Th17/Treg immune response. J Neuroimmunol 2016; 299:45-52. [DOI: 10.1016/j.jneuroim.2016.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 08/05/2016] [Accepted: 08/15/2016] [Indexed: 12/17/2022]
|
31
|
Wang J, Zhao C, Kong P, Sun H, Sun Z, Bian G, Sun Y, Guo L. Treatment with NAD(+) inhibited experimental autoimmune encephalomyelitis by activating AMPK/SIRT1 signaling pathway and modulating Th1/Th17 immune responses in mice. Int Immunopharmacol 2016; 39:287-294. [PMID: 27500459 DOI: 10.1016/j.intimp.2016.07.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/13/2016] [Accepted: 07/31/2016] [Indexed: 10/21/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) plays vital roles in mitochondrial functions, cellular energy metabolism and calcium homeostasis. In this study, we investigated the effect of NAD(+) administration for the treatment of experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice. EAE, a classical animal model of multiple sclerosis (MS), was induced by subcutaneous injection of myelin oligodendrocyteglycoprotein (MOG). The mice were treated with 250mg/kg (body weight) NAD(+) in PBS administered intraperitoneally once daily. We observed that NAD(+) treatment could lessen the severity of EAE. Additionally, NAD(+) treatment attenuated pathological injuries of EAE mice. We also found that the AMP-activated protein kinase (AMPK)/silent mating-type information regulation 2 homolog 1(SIRT1) pathway was activated in the NAD(+)-treated mice and NAD(+) treatment suppressed pro-inflammatory T cell responses. Our findings demonstrated that NAD(+) could be an effective and promising agent to treat multiple sclerosis and its effects on other autoimmune diseases should be explored.
Collapse
Affiliation(s)
- Jueqiong Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang, Hebei 050000, China
| | - Congying Zhao
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang, Hebei 050000, China
| | - Peng Kong
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang, Hebei 050000, China
| | - Huanhuan Sun
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Heping West Road 215, Shijiazhuang, Hebei 050000, China
| | - Zhe Sun
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang, Hebei 050000, China
| | - Guanyun Bian
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang, Hebei 050000, China
| | - Yafei Sun
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang, Hebei 050000, China
| | - Li Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, Key Laboratory of Hebei Neurology, Shijiazhuang, Hebei 050000, China.
| |
Collapse
|
32
|
Korniotis S, Gras C, Letscher H, Montandon R, Mégret J, Siegert S, Ezine S, Fallon PG, Luther SA, Fillatreau S, Zavala F. Treatment of ongoing autoimmune encephalomyelitis with activated B-cell progenitors maturing into regulatory B cells. Nat Commun 2016; 7:12134. [PMID: 27396388 PMCID: PMC4942579 DOI: 10.1038/ncomms12134] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 06/02/2016] [Indexed: 12/24/2022] Open
Abstract
The influence of signals perceived by immature B cells during their development in bone marrow on their subsequent functions as mature cells are poorly defined. Here, we show that bone marrow cells transiently stimulated in vivo or in vitro through the Toll-like receptor 9 generate proB cells (CpG-proBs) that interrupt experimental autoimmune encephalomyelitis (EAE) when transferred at the onset of clinical symptoms. Protection requires differentiation of CpG-proBs into mature B cells that home to reactive lymph nodes, where they trap T cells by releasing the CCR7 ligand, CCL19, and to inflamed central nervous system, where they locally limit immunopathogenesis through interleukin-10 production, thereby cooperatively inhibiting ongoing EAE. These data demonstrate that a transient inflammation at the environment, where proB cells develop, is sufficient to confer regulatory functions onto their mature B-cell progeny. In addition, these properties of CpG-proBs open interesting perspectives for cell therapy of autoimmune diseases. Evidence of how functional Bregs develop in vivo has been lacking. Here the authors show that proB cells exposed in vivo to CpG differentiate into distinct Breg subsets that inhibit autoimmunity by arresting T cells in the lymph nodes via CCL19 and by producing IL-10 at the site of immunopathology.
Collapse
Affiliation(s)
- Sarantis Korniotis
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Christophe Gras
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Hélène Letscher
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Ruddy Montandon
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Jérôme Mégret
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Structure Fédérative de Recherche Necker, INSERM US 24, CNRS UMS 3633, Paris 75014, France
| | - Stefanie Siegert
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Sophie Ezine
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Padraic G Fallon
- Department of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Simon Fillatreau
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Assistance Publique-Hopitaux de Paris (AP-HP), Hopital Necker Enfants Malades, Paris 75015, France.,Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Chariteplatz 1, Berlin 10117, Germany
| | - Flora Zavala
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| |
Collapse
|
33
|
Bhise V, Dhib-Jalbut S. Further understanding of the immunopathology of multiple sclerosis: impact on future treatments. Expert Rev Clin Immunol 2016; 12:1069-89. [PMID: 27191526 DOI: 10.1080/1744666x.2016.1191351] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The understanding of the immunopathogenesis of multiple sclerosis (MS) has expanded with more research into T-cell subtypes, cytokine contributors, B-cell participation, mitochondrial dysfunction, and more. Treatment options have rapidly expanded with three relatively recent oral therapy alternatives entering the arena. AREAS COVERED In the following review, we discuss current mechanisms of immune dysregulation in MS, how they relate to current treatments, and the impact these findings will have on the future of therapy. Expert commentary: The efficacy of these medications and understanding their mechanisms of actions validates the immunopathogenic mechanisms thought to underlie MS. Further research has exposed new targets, while new promising therapies have shed light on new aspects into the pathophysiology of MS.
Collapse
Affiliation(s)
- Vikram Bhise
- a Rutgers Biomedical and Health Sciences - Departments of Pediatrics , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| | - Suhayl Dhib-Jalbut
- b Rutgers Biomedical and Health Sciences - Departments of Neurology , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| |
Collapse
|
34
|
Kok LF, Marsh-Wakefield F, Marshall JE, Gillis C, Halliday GM, Byrne SN. B cells are required for sunlight protection of mice from a CNS-targeted autoimmune attack. J Autoimmun 2016; 73:10-23. [PMID: 27289166 DOI: 10.1016/j.jaut.2016.05.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/09/2023]
Abstract
The ultraviolet (UV) radiation contained in sunlight is a powerful immune suppressant. While exposure to UV is associated with protection from the development of autoimmune diseases, particularly multiple sclerosis, the precise mechanism by which UV achieves this protection is not currently well understood. Regulatory B cells play an important role in preventing autoimmunity and activation of B cells is a major way in which UV suppresses adaptive immune responses. Whether UV-protection from autoimmunity is mediated by the activation of regulatory B cells has never been considered before. When C57BL/6 mice were exposed to low, physiologically relevant doses of UV, a unique population of B cells was activated in the skin draining lymph nodes. As determined by flow cytometry, CD1d(low)CD5(-)MHC-II(hi)B220(hi) UV-activated B cells expressed significantly higher levels of CD19, CD21/35, CD25, CD210 and CD268 as well as the co-stimulatory molecules CD80, CD86, CD274 and CD275. Experimental autoimmune encephalomyelitis (EAE) in mice immunized with MOG/CFA was reduced by exposure to UV. UV significantly inhibited demyelination and infiltration of inflammatory cells into the spinal cord. Consequently, UV-exposed groups showed elevated IL-10 levels in secondary lymphoid organs, delayed EAE onset, reduced peak EAE score and significantly suppressed overall disease incidence and burden. Importantly, protection from EAE could be adoptively transferred using B cells isolated from UV-exposed, but not unirradiated hosts. Indeed, UV-protection from EAE was dependent on UV activation of lymph node B cells because UV could not protect mice from EAE who were pharmacologically depleted of B cells using antibodies. Thus, UV maintenance of a pool of unique regulatory B cells in peripheral lymph nodes appears to be essential to prevent an autoimmune attack on the central nervous system.
Collapse
Affiliation(s)
- Lai Fong Kok
- Cellular Photoimmunology Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School at the Charles Perkins Centre, University of Sydney, Australia
| | - Felix Marsh-Wakefield
- Cellular Photoimmunology Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School at the Charles Perkins Centre, University of Sydney, Australia
| | - Jacqueline E Marshall
- Cellular Photoimmunology Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School at the Charles Perkins Centre, University of Sydney, Australia
| | - Caitlin Gillis
- Discipline of Dermatology at the Bosch Institute, Sydney Medical School, University of Sydney and Royal Prince Alfred Hospital, Australia
| | - Gary M Halliday
- Cellular Photoimmunology Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School at the Charles Perkins Centre, University of Sydney, Australia; Discipline of Dermatology at the Bosch Institute, Sydney Medical School, University of Sydney and Royal Prince Alfred Hospital, Australia
| | - Scott N Byrne
- Cellular Photoimmunology Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School at the Charles Perkins Centre, University of Sydney, Australia; Discipline of Dermatology at the Bosch Institute, Sydney Medical School, University of Sydney and Royal Prince Alfred Hospital, Australia.
| |
Collapse
|
35
|
Huarte E, Jun S, Rynda-Apple A, Golden S, Jackiw L, Hoffman C, Maddaloni M, Pascual DW. Regulatory T Cell Dysfunction Acquiesces to BTLA+ Regulatory B Cells Subsequent to Oral Intervention in Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2016; 196:5036-46. [PMID: 27194787 DOI: 10.4049/jimmunol.1501973] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 04/14/2016] [Indexed: 12/21/2022]
Abstract
Regulatory T cells (Tregs) induced during autoimmunity often become quiescent and unable to resolve disease, suggesting inadequate activation. Resolution of established experimental autoimmune encephalomyelitis (EAE) can be achieved with myelin oligodendrocyte glycoprotein (MOG) fused to reovirus protein σ1 (MOG-pσ1), which activates Tregs, restoring protection, but requiring other regulatory cells to revitalize them. B cells have a dichotomous role in both the pathogenesis and recovery from EAE. Although inflammatory B cells contribute to EAE's pathogenesis, treatment of EAE mice with MOG-pσ1, but not OVA-pσ1, resulted in an influx of IL-10-producing B220(+)CD5(+) B regulatory cells (Bregs) enabling Tregs to recover their inhibitory activity, and in turn, leading to the rapid amelioration of EAE. These findings implicate direct interactions between Bregs and Tregs to facilitate this recovery. Adoptive transfer of B220(+)CD5(-) B cells from MOG-pσ1-treated EAE or Bregs from PBS-treated EAE mice did not resolve disease, whereas the adoptive transfer of MOG-pσ1-induced B220(+)CD5(+) Bregs greatly ameliorated EAE. MOG-pσ1-, but not OVA-pσ1-induced IL-10-producing Bregs, expressed elevated levels of B and T lymphocyte attenuator (BTLA) relative to CD5(-) B cells, as opposed to Tregs or effector T (Teff) cells, whose BTLA expression was not affected. These induced Bregs restored EAE Treg function in a BTLA-dependent manner. BTLA(-/-) mice showed more pronounced EAE with fewer Tregs, but upon adoptive transfer of MOG-pσ1-induced BTLA(+) Bregs, BTLA(-/-) mice were protected against EAE. Hence, this evidence shows the importance of BTLA in activating Tregs to facilitate recovery from EAE.
Collapse
Affiliation(s)
- Eduardo Huarte
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611; and
| | - SangMu Jun
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611; and
| | - Agnieszka Rynda-Apple
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59718
| | - Sara Golden
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59718
| | - Larissa Jackiw
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59718
| | - Carol Hoffman
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611; and
| | - Massimo Maddaloni
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611; and
| | - David W Pascual
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611; and
| |
Collapse
|
36
|
Abstract
A rapidly changing set of drugs for treatment of multiple sclerosis (MS) leads to the necessity of searching for predictors of their efficacy. Understanding of pathogenetic processes in MS and mechanisms of action of different drugs play an important role in the search for markers of potential responders. The author analyses the presently accumulated information on the original drug copaxone (glatiramer acetate) including current concepts on the mechanism of action, long-term safety and efficacy. Data on the frequency and significance of adverse effects during treatment with glatiramer acetate as well as on the influence of the drug on pregnancy, postpartum course of MS and development of the infant who received glatiramer acetate prenatally compared to other disease-modifying drugs are presented.
Collapse
Affiliation(s)
- D S Kasatkin
- Department of Nervous Diseases with Medical Genetics and Neurosurgery 'Yaroslavl state medical University', Yaroslavl, Russia
| |
Collapse
|
37
|
Sonar S, Lal G. Role of Tumor Necrosis Factor Superfamily in Neuroinflammation and Autoimmunity. Front Immunol 2015; 6:364. [PMID: 26257732 PMCID: PMC4507150 DOI: 10.3389/fimmu.2015.00364] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/05/2015] [Indexed: 12/18/2022] Open
Abstract
Tumor necrosis factor superfamily (TNFSF) molecules play an important role in the activation, proliferation, differentiation, and migration of immune cells into the central nervous system (CNS). Several TNF superfamily molecules are known to control alloimmunity, autoimmunity, and immunity. Development of transgenic and gene knockout animals, and monoclonal antibodies against TNFSF molecules have increased our understanding of individual receptor-ligand interactions, and their intracellular signaling during homeostasis and neuroinflammation. A strong clinical association has been observed between TNFSF members and CNS autoimmunity such as multiple sclerosis and also in its animal model experimental autoimmune encephalomyelitis. Therefore, they are promising targets for alternative therapeutic options to control autoimmunity. Although, TNFSF ligands are widely distributed and have diverse functions, we have restricted the discussions in this review to TNFSF receptor-ligand interactions and their role in the pathogenesis of neuroinflammation and CNS autoimmunity.
Collapse
|
38
|
Ireland SJ, Monson NL, Davis LS. Seeking balance: Potentiation and inhibition of multiple sclerosis autoimmune responses by IL-6 and IL-10. Cytokine 2015; 73:236-44. [PMID: 25794663 PMCID: PMC4437890 DOI: 10.1016/j.cyto.2015.01.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/12/2015] [Accepted: 01/22/2015] [Indexed: 01/07/2023]
Abstract
The cytokines IL-6 and IL-10 are produced by cells of the adaptive and innate arms of the immune system and they appear to play key roles in genetically diverse autoimmune diseases such as relapsing remitting multiple sclerosis (MS), rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). Whereas previous intense investigations focused on the generation of autoantibodies and their contribution to immune-mediated pathogenesis in these diseases; more recent attention has focused on the roles of cytokines such as IL-6 and IL-10. In response to pathogens, antigen presenting cells (APC), including B cells, produce IL-6 and IL-10 in order to up-or down-regulate immune cell activation and effector responses. Evidence of elevated levels of the proinflammatory cytokine IL-6 has been routinely observed during inflammatory responses and in a number of autoimmune diseases. Our recent studies suggest that MS peripheral blood B cells secrete higher quantities of IL-6 and less IL-10 than B cells from healthy controls. Persistent production of IL-6, in turn, contributes to T cell expansion and the functional hyperactivity of APC such as MS B cells. Altered B cell activity can have a profound impact on resultant T cell effector functions. Enhanced signaling through the IL-6 receptor can effectively inhibit cytolytic activity, induce T cell resistance to IL-10-mediated immunosuppression and increase skewing of autoreactive T cells to a pathogenic Th17 phenotype. Our recent findings and studies by others support a role for the indirect attenuation of B cell responses by Glatiramer acetate (GA) therapy. Our studies suggest that GA therapy temporarily permits homeostatic regulatory mechanisms to be reinstated. Future studies of mechanisms underlying dysregulated B cell cytokine production could lead to the identification of novel targets for improved immunoregulatory therapies for autoimmune diseases.
Collapse
Affiliation(s)
- Sara J Ireland
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, United States.
| | - Nancy L Monson
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, United States.
| | - Laurie S Davis
- Rheumatic Diseases Division, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, United States.
| |
Collapse
|
39
|
Telesford K, Ochoa-Repáraz J, Kasper LH. Gut commensalism, cytokines, and central nervous system demyelination. J Interferon Cytokine Res 2015; 34:605-14. [PMID: 25084177 DOI: 10.1089/jir.2013.0134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is increasing support for the importance of risk factors such as genetic makeup, obesity, smoking, vitamin D insufficiency, and antibiotic exposure contributing to the development of autoimmune diseases, including human multiple sclerosis (MS). Perhaps the greatest environmental risk factor associated with the development of immune-mediated conditions is the gut microbiome. Microbial and helminthic agents are active participants in shaping the immune systems of their hosts. This concept is continually reinforced by studies in the burgeoning area of commensal-mediated immunomodulation. The clinical importance of these findings for MS is suggested by both their participation in disease and, perhaps of greater clinical importance, attenuation of disease severity. Observations made in murine models of central nervous system demyelinating disease and a limited number of small studies in human MS suggest that immune homeostasis within the gut microbiome may be of paramount importance in maintaining a disease-free state. This review describes three immunological factors associated with the gut microbiome that are central to cytokine network activities in MS pathogenesis: T helper cell polarization, T regulatory cell function, and B cell activity. Comparisons are drawn between the regulatory mechanisms attributed to first-line therapies and those described in commensal-mediated amelioration of central nervous system demyelination.
Collapse
Affiliation(s)
- Kiel Telesford
- 1 Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire
| | | | | |
Collapse
|
40
|
Abstract
The role of CD8+ T cells in the process of autoimmune pathology has been both understudied and controversial. Multiple sclerosis (MS) is an inflammatory, demyelinating disorder of the central nervous system (CNS) with underlying T cell-mediated immunopathology. CD8+ T cells are the predominant T cells in human MS lesions, showing oligoclonal expansion at the site of pathology. It is still unclear whether these cells represent pathogenic immune responses or disease-regulating elements. Through studies in human MS and its animal model, experimental autoimmune encephalomyelitis (EAE), we have discovered two novel CD8+ T cell populations that play an essential immunoregulatory role in disease: (1) MHC class Ia-restricted neuroantigen-specific "autoregulatory" CD8+ T cells and (2) glatiramer acetate (GA/Copaxone(®)) therapy-induced Qa-1/HLA-E-restricted GA-specific CD8+ T cells. These CD8+ Tregs suppress proliferation of pathogenic CD4+ CD25- T cells when stimulated by their cognate antigens. Similarly, CD8+ Tregs significantly suppress EAE when transferred either pre-disease induction or during peak disease. The mechanism of disease inhibition depends, at least in part, on an antigen-specific, contact-dependent process and works through modulation of CD4+ T cell responses as well as antigen-presenting cells through a combination of cytotoxicity and cytokine-mediated modulation. This review provides an overview of our understanding of CD8+ T cells in immune-mediated disease, focusing particularly on our findings regarding regulatory CD8+ T cells both in MS and in EAE. Clinical relevance of these novel CD8-regulatory populations is discussed, providing insights into a potentially intriguing, novel therapeutic strategy for these diseases.
Collapse
|
41
|
Ireland SJ, Guzman AA, O'Brien DE, Hughes S, Greenberg B, Flores A, Graves D, Remington G, Frohman EM, Davis LS, Monson NL. The effect of glatiramer acetate therapy on functional properties of B cells from patients with relapsing-remitting multiple sclerosis. JAMA Neurol 2015; 71:1421-8. [PMID: 25264704 DOI: 10.1001/jamaneurol.2014.1472] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
IMPORTANCE This study describes what is, to our knowledge, the previously unknown effect of glatiramer acetate therapy on B cells in patients with relapsing-remitting multiple sclerosis (MS). OBJECTIVE To determine whether glatiramer acetate therapy normalizes dysregulated B-cell proliferation and cytokine production in patients with MS. DESIGN, SETTING, AND PARTICIPANTS Twenty-two patients with MS who were receiving glatiramer acetate therapy and 22 treatment-naive patients with MS were recruited at The University of Texas Southwestern Medical Center MS clinic. Cell samples from healthy donors were obtained from HemaCare (Van Nuys, California) or Carter Blood Bank (Dallas, Texas). Treatment-naive patients with MS had not received any disease-modifying therapies for at least 3 months before the study. EXPOSURES Glatiramer acetate therapy for at least 3 months at the time of the study. MAIN OUTCOMES AND MEASURES B-cell phenotype and proliferation and immunoglobulin and cytokine secretion. RESULTS A restoration of interleukin 10 production by peripheral B cells was observed in patients undergoing glatiramer acetate therapy as well as a significant reduction of interleukin 6 production in a subset of patients who received therapy for less than 32 months. Furthermore, proliferation in response to high-dose CD40L was altered and immunoglobulin production was elevated in in vitro-activated B cells obtained from patients who received glatiramer acetate. CONCLUSIONS AND RELEVANCE Glatiramer acetate therapy remodels the composition of the B-cell compartment and influences cytokine secretion and immunoglobulin production. These data suggest that glatiramer acetate therapy affects several aspects of dysregulated B-cell function in MS that may contribute to the therapeutic mechanisms of glatiramer acetate.
Collapse
Affiliation(s)
- Sara J Ireland
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Alyssa A Guzman
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Dina E O'Brien
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Samuel Hughes
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Benjamin Greenberg
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Angela Flores
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Donna Graves
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Gina Remington
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Elliot M Frohman
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas
| | - Laurie S Davis
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas
| | - Nancy L Monson
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas3Department of Immunology, The University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
42
|
|
43
|
Jones TB. Lymphocytes and autoimmunity after spinal cord injury. Exp Neurol 2014; 258:78-90. [PMID: 25017889 DOI: 10.1016/j.expneurol.2014.03.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 12/23/2022]
Abstract
Over the past 15 years an immense amount of data has accumulated regarding the infiltration and activation of lymphocytes in the traumatized spinal cord. Although the impact of the intraspinal accumulation of lymphocytes is still unclear, modulation of the adaptive immune response via active and passive vaccination is being evaluated for its preclinical efficacy in improving the outcome for spinal-injured individuals. The complexity of the interaction between the nervous and the immune systems is highlighted in the contradictions that appear in response to these modulations. Current evidence regarding augmentation and inhibition of the adaptive immune response to spinal cord injury is reviewed with an aim toward reconciling conflicting data and providing consensus issues that may be exploited in future therapies. Opportunities such an approach may provide are highlighted as well as the obstacles that must be overcome before such approaches can be translated into clinical trials.
Collapse
Affiliation(s)
- T Bucky Jones
- Department of Anatomy, Arizona College of Medicine, Midwestern University, Glendale, AZ, USA.
| |
Collapse
|
44
|
Ben-Nun A, Kaushansky N, Kawakami N, Krishnamoorthy G, Berer K, Liblau R, Hohlfeld R, Wekerle H. From classic to spontaneous and humanized models of multiple sclerosis: impact on understanding pathogenesis and drug development. J Autoimmun 2014; 54:33-50. [PMID: 25175979 DOI: 10.1016/j.jaut.2014.06.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/04/2014] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS), a demyelinating disease of the central nervous system (CNS), presents as a complex disease with variable clinical and pathological manifestations, involving different pathogenic pathways. Animal models, particularly experimental autoimmune encephalomyelitis (EAE), have been key to deciphering the pathophysiology of MS, although no single model can recapitulate the complexity and diversity of MS, or can, to date, integrate the diverse pathogenic pathways. Since the first EAE model was introduced decades ago, multiple classic (induced), spontaneous, and humanized EAE models have been developed, each recapitulating particular aspects of MS pathogenesis. The advances in technologies of genetic ablation and transgenesis in mice of C57BL/6J background and the development of myelin-oligodendrocyte glycoprotein (MOG)-induced EAE in C57BL/6J mice yielded several spontaneous and humanized EAE models, and resulted in a plethora of EAE models in which the role of specific genes or cell populations could be precisely interrogated, towards modeling specific pathways of MS pathogenesis/regulation in MS. Collectively, the numerous studies on the different EAE models contributed immensely to our basic understanding of cellular and molecular pathways in MS pathogenesis as well as to the development of therapeutic agents: several drugs available today as disease modifying treatments were developed from direct studies on EAE models, and many others were tested or validated in EAE. In this review, we discuss the contribution of major classic, spontaneous, and humanized EAE models to our understanding of MS pathophysiology and to insights leading to devising current and future therapies for this disease.
Collapse
Affiliation(s)
- Avraham Ben-Nun
- Department of Immunology, The Weizmann Institute of Science, 234 Herzl St. Rehovot, 7610001, Israel.
| | - Nathali Kaushansky
- Department of Immunology, The Weizmann Institute of Science, 234 Herzl St. Rehovot, 7610001, Israel.
| | - Naoto Kawakami
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried 82152, Germany; Institute of Clinical Neuroimmunology, Ludwig-Maximilians-University, 81377 Munich, Germany.
| | | | - Kerstin Berer
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried 82152, Germany.
| | | | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians-University, 81377 Munich, Germany.
| | - Hartmut Wekerle
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried 82152, Germany.
| |
Collapse
|
45
|
Kashi VP, Ortega SB, Karandikar NJ. Neuroantigen-specific autoregulatory CD8+ T cells inhibit autoimmune demyelination through modulation of dendritic cell function. PLoS One 2014; 9:e105763. [PMID: 25144738 PMCID: PMC4140828 DOI: 10.1371/journal.pone.0105763] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/24/2014] [Indexed: 01/29/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a well-established murine model of multiple sclerosis, an immune-mediated demyelinating disorder of the central nervous system (CNS). We have previously shown that CNS-specific CD8+ T cells (CNS-CD8+) ameliorate EAE, at least in part through modulation of CNS-specific CD4+ T cell responses. In this study, we show that CNS-CD8+ also modulate the function of CD11c+ dendritic cells (DC), but not other APCs such as CD11b+ monocytes or B220+ B cells. DC from mice receiving either myelin oligodendrocyte glycoprotein-specific CD8+ (MOG-CD8+) or proteolipid protein-specific CD8+ (PLP-CD8+) T cells were rendered inefficient in priming T cell responses from naïve CD4+ T cells (OT-II) or supporting recall responses from CNS-specific CD4+ T cells. CNS-CD8+ did not alter DC subset distribution or MHC class II and CD86 expression, suggesting that DC maturation was not affected. However, the cytokine profile of DC from CNS-CD8+ recipients showed lower IL-12 and higher IL-10 production. These functions were not modulated in the absence of immunization with CD8-cognate antigen, suggesting an antigen-specific mechanism likely requiring CNS-CD8-DC interaction. Interestingly, blockade of IL-10 in vitro rescued CD4+ proliferation and in vivo expression of IL-10 was necessary for the suppression of EAE by MOG-CD8+. These studies demonstrate a complex interplay between CNS-specific CD8+ T cells, DC and pathogenic CD4+ T cells, with important implications for therapeutic interventions in this disease.
Collapse
Affiliation(s)
- Venkatesh P. Kashi
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Sterling B. Ortega
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Nitin J. Karandikar
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
46
|
Starossom SC, Veremeyko T, Dukhinova M, Yung AWY, Ponomarev ED. Glatiramer acetate (copaxone) modulates platelet activation and inhibits thrombin-induced calcium influx: possible role of copaxone in targeting platelets during autoimmune neuroinflammation. PLoS One 2014; 9:e96256. [PMID: 24788965 PMCID: PMC4008572 DOI: 10.1371/journal.pone.0096256] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 04/07/2014] [Indexed: 11/18/2022] Open
Abstract
Background Glatiramer acetate (GA, Copaxone, Copolymer-1) is an FDA approved drug for the treatment of MS and it is very effective in suppressing neuroinflammation in experimental autoimmune encephalitis (EAE), an animal model of MS. Although this drug was designed to inhibit pathogenic T cells, the exact mechanism of EAE/MS suppression by GA is still not well understood. Previously we presented evidence that platelets become activated and promote neuroinflammation in EAE, suggesting a possible pathogenic role of platelets in MS and EAE. We hypothesized that GA could inhibit neuroinflammation by affecting not only immune cells but also platelets. Methodology/Principal Findings We investigated the effect of GA on the activation of human platelets in vitro: calcium influx, platelet aggregation and expression of activation markers. Our results in human platelets were confirmed by in-vitro and in-vivo studies of modulation of functions of platelets in mouse model. We found that GA inhibited thrombin-induced calcium influx in human and mouse platelets. GA also decreased thrombin-induced CD31, CD62P, CD63, and active form of αIIbβ3 integrin surface expression and formation of platelet aggregates for both mouse and human platelets, and prolonged the bleeding time in mice by 2.7-fold. In addition, we found that GA decreased the extent of macrophage activation induced by co-culture of macrophages with platelets. Conclusions GA inhibited the activation of platelets, which suggests a new mechanism of GA action in suppression of EAE/MS by targeting platelets and possibly preventing their interaction with immune cells such as macrophages. Furthermore, the reduction in platelet activation by GA may have additional cardiovascular benefits to prevent thrombosis.
Collapse
Affiliation(s)
- Sarah C. Starossom
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Institute for Medical Immunology and NeuroCure, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tatyana Veremeyko
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Marina Dukhinova
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Amanda W. Y. Yung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Eugene D. Ponomarev
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
- * E-mail:
| |
Collapse
|
47
|
Johnson KP. Glatiramer acetate for treatment of relapsing–remitting multiple sclerosis. Expert Rev Neurother 2014; 12:371-84. [DOI: 10.1586/ern.12.25] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
48
|
van der Vlugt LEPM, Haeberlein S, de Graaf W, Martha TED, Smits HH. Toll-like receptor ligation for the induction of regulatory B cells. Methods Mol Biol 2014; 1190:127-141. [PMID: 25015278 DOI: 10.1007/978-1-4939-1161-5_10] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Toll-like receptors (TLRs) are key components for the recognition of microorganisms, for the initiation of innate immunity, and for promoting adaptive immune responses. TLR signaling in B cells, in addition to B cell receptor or CD40 ligation, plays an important role in B cell differentiation and activation. In contrast, various infectious agents and/or TLR ligands can also prime B cells to induce tolerance and downregulate inflammatory reactions; those B cells are called regulatory B (Breg) cells and are characterized by a dominant IL-10 production. Several studies have suggested that Breg cells are impaired in patients with autoimmune diseases and allergic asthma. However, the role for TLR ligands in the induction of Breg cells as a potential therapy for some of these inflammatory diseases has not yet been investigated. Here, we provide detailed instructions on how to analyze and validate cytokine production in human and mouse B cells in response to various TLR ligands. Furthermore, we describe an assay to investigate the suppressive properties of TLR-induced B cells to confirm their regulatory B cell status.
Collapse
Affiliation(s)
- Luciën E P M van der Vlugt
- Department of Parasitology, Cellular Immunology of Helminths, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
49
|
Muzzio D, Zygmunt M, Jensen F. The role of pregnancy-associated hormones in the development and function of regulatory B cells. Front Endocrinol (Lausanne) 2014; 5:39. [PMID: 24744750 PMCID: PMC3978254 DOI: 10.3389/fendo.2014.00039] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/14/2014] [Indexed: 12/14/2022] Open
Abstract
During mammalian pregnancy, highly specialized mechanisms of immune tolerance are triggered in order to allow the semi-allogeneic fetus to grow within the maternal uterus in harmony with the maternal immune system. Among other mechanisms, changes in the endocrine status have been proposed to be at least part of the machinery responsible for the induction of immune tolerance during pregnancy. Indeed, pregnancy-associated hormones, estradiol, progesterone, and human chorionic gonadotropin are known to confer immune suppressive capacity to innate as well as adaptive immune cells. Regulatory B cells, a subpopulation of B lymphocytes with strong immunosuppressive functions, were shown to expand during pregnancy. Furthermore, it is well-known that some women suffering from multiple sclerosis, significantly improve their symptoms during pregnancy and this was attributed to the effect of female sex hormones. Accordingly, estradiol protects mice from developing experimental autoimmune encephalomyelitis by triggering the expansion and activation of regulatory B cells. In this review, we discuss different mechanisms associated with the development, activation, and function of regulatory B cells with a special focus on those involving pregnancy-associated hormones.
Collapse
Affiliation(s)
- Damián Muzzio
- Research Laboratory, Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Marek Zygmunt
- Research Laboratory, Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Federico Jensen
- Research Laboratory, Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
- *Correspondence: Federico Jensen, Research Laboratory, Department of Obstetrics and Gynecology, University of Greifswald, Sauerbruchstr., Greifswald 17475, Germany e-mail:
| |
Collapse
|
50
|
Abstract
B cells are thought to play a pathogenic role in multiple sclerosis (MS), an autoimmune disease affecting the central nervous system (CNS). This idea is supported by the reduction of disease in MS patients undergoing antibody-mediated B cell depletion therapy. In contrast, in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, B cells have been shown to play a regulatory role. This is suggestive of a dual role for B cells in CNS autoimmunity. It is possible that a critical balance between the pathogenic and regulatory populations of B cells might be involved in the manifestation of the disease. Although in mice, different B cell subsets have been shown to exert immunoregulation through varied mechanisms, the phenotype of regulatory B cells in humans and factors affecting their function are not well known. Also, the origin and development of regulatory B cells is not known. It is important to thoroughly identify the different populations of B cells that might be involved in suppressing CNS autoimmunity, their mode of function and factors that regulate their immunosuppressive properties for using regulatory B cells as a therapy for MS. Here we present methods to study the phenotype and mechanisms of immune suppression by B cells in different mouse models of EAE.
Collapse
Affiliation(s)
- Avijit Ray
- BloodCenter of Wisconsin, Blood Research Institute, 2178, Milwaukee, WI, 53201-2178, USA,
| | | |
Collapse
|