1
|
Laketa D, Lavrnja I. Extracellular Purine Metabolism-Potential Target in Multiple Sclerosis. Mol Neurobiol 2024; 61:8361-8386. [PMID: 38499905 DOI: 10.1007/s12035-024-04104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
The purinergic signaling system comprises a complex network of extracellular purines and purine-metabolizing ectoenzymes, nucleotide and nucleoside receptors, ATP release channels, and nucleoside transporters. Because of its immunomodulatory function, this system is critically involved in the pathogenesis of multiple sclerosis (MS) and its best-characterized animal model, experimental autoimmune encephalomyelitis (EAE). MS is a chronic neuroinflammatory demyelinating and neurodegenerative disease with autoimmune etiology and great heterogeneity, mostly affecting young adults and leading to permanent disability. In MS/EAE, alterations were detected in almost all components of the purinergic signaling system in both peripheral immune cells and central nervous system (CNS) glial cells, which play an important role in the pathogenesis of the disease. A decrease in extracellular ATP levels and an increase in its downstream metabolites, particularly adenosine and inosine, were frequently observed at MS, indicating a shift in metabolism toward an anti-inflammatory environment. Accordingly, upregulation of the major ectonucleotidase tandem CD39/CD73 was detected in the blood cells and CNS of relapsing-remitting MS patients. Based on the postulated role of A2A receptors in the transition from acute to chronic neuroinflammation, the association of variants of the adenosine deaminase gene with the severity of MS, and the beneficial effects of inosine treatment in EAE, the adenosinergic system emerged as a promising target in neuroinflammation. More recently, several publications have identified ADP-dependent P2Y12 receptors and the major extracellular ADP producing enzyme nucleoside triphosphate diphosphohydrolase 2 (NTPDase2) as novel potential targets in MS.
Collapse
Affiliation(s)
- Danijela Laketa
- Department of General Physiology and Biophysics, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Studentski Trg 3, Belgrade, Republic of Serbia.
| | - Irena Lavrnja
- Institute for Biological Research, Sinisa Stankovic" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade, Republic of Serbia
| |
Collapse
|
2
|
Ni D, Tan J, Robert R, Taitz J, Ge A, Potier-Villette C, Reyes JGA, Spiteri A, Wishart C, Mackay C, Piccio L, King NJC, Macia L. GPR109A expressed on medullary thymic epithelial cells affects thymic Treg development. Eur J Immunol 2023; 53:e2350521. [PMID: 37595951 DOI: 10.1002/eji.202350521] [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: 04/06/2023] [Revised: 07/31/2023] [Accepted: 08/17/2023] [Indexed: 08/20/2023]
Abstract
Regulatory T cells (Treg) maintain immune homeostasis due to their anti-inflammatory functions. They can be generated either centrally in the thymus or in peripheral organs. Metabolites such as short-chain fatty acids produced by intestinal microbiota can induce peripheral Treg differentiation, by activating G-protein-coupled-receptors like GPR109A. In this study, we identified a novel role for GPR109A in thymic Treg development. We found that Gpr109a-/- mice had increased Treg under basal conditions in multiple organs compared with WT mice. GPR109A was not expressed on T cells but on medullary thymic epithelial cells (mTECs), as revealed by single-cell RNA sequencing in both mice and humans and confirmed by flow cytometry in mice. mTECs isolated from Gpr109a-/- mice had higher expression of autoimmune regulator (AIRE), the key regulator of Treg development, while the subset of mTECs that did not express Gpr109a in the WT displayed increased Aire expression and also enhanced signaling related to mTEC functionality. Increased thymic Treg in Gpr109a-/- mice was associated with protection from experimental autoimmune encephalomyelitis, with ameliorated clinical signs and reduced inflammation. This work identifies a novel role for GPR109A and possibly the gut microbiota, on thymic Treg development via its regulation of mTECs.
Collapse
Affiliation(s)
- Duan Ni
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jian Tan
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Remy Robert
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jemma Taitz
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anjie Ge
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Camille Potier-Villette
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Julen Gabirel Araneta Reyes
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alanna Spiteri
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Claire Wishart
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Charles Mackay
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Laura Piccio
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas Jonathan Cole King
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, The University of Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Rossi B, Santos-Lima B, Terrabuio E, Zenaro E, Constantin G. Common Peripheral Immunity Mechanisms in Multiple Sclerosis and Alzheimer's Disease. Front Immunol 2021; 12:639369. [PMID: 33679799 PMCID: PMC7933037 DOI: 10.3389/fimmu.2021.639369] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are closely related to inflammatory and autoimmune events, suggesting that the dysregulation of the immune system is a key pathological factor. Both multiple sclerosis (MS) and Alzheimer's disease (AD) are characterized by infiltrating immune cells, activated microglia, astrocyte proliferation, and neuronal damage. Moreover, MS and AD share a common pro-inflammatory signature, characterized by peripheral leukocyte activation and transmigration to the central nervous system (CNS). MS and AD are both characterized by the accumulation of activated neutrophils in the blood, leading to progressive impairment of the blood–brain barrier. Having migrated to the CNS during the early phases of MS and AD, neutrophils promote local inflammation that contributes to pathogenesis and clinical progression. The role of circulating T cells in MS is well-established, whereas the contribution of adaptive immunity to AD pathogenesis and progression is a more recent discovery. Even so, blocking the transmigration of T cells to the CNS can benefit both MS and AD patients, suggesting that common adaptive immunity mechanisms play a detrimental role in each disease. There is also growing evidence that regulatory T cells are beneficial during the initial stages of MS and AD, supporting the link between the modulatory immune compartments and these neurodegenerative disorders. The number of resting regulatory T cells declines in both diseases, indicating a common pathogenic mechanism involving the dysregulation of these cells, although their precise role in the control of neuroinflammation remains unclear. The modulation of leukocyte functions can benefit MS patients, so more insight into the role of peripheral immune cells may reveal new targets for pharmacological intervention in other neuroinflammatory and neurodegenerative diseases, including AD.
Collapse
Affiliation(s)
- Barbara Rossi
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Bruno Santos-Lima
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Eleonora Terrabuio
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Elena Zenaro
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy.,The Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| |
Collapse
|
4
|
Moorman CD, Bastian AG, DeOca KB, Mannie MD. A GM-CSF-neuroantigen tolerogenic vaccine elicits inefficient antigen recognition events below the CD40L triggering threshold to expand CD4 + CD25 + FOXP3 + Tregs that inhibit experimental autoimmune encephalomyelitis (EAE). J Neuroinflammation 2020; 17:180. [PMID: 32522287 PMCID: PMC7285464 DOI: 10.1186/s12974-020-01856-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Background Tolerogenic vaccines represent antigen-specific interventions designed to re-establish self-tolerance and thereby alleviate autoimmune diseases, which collectively comprise over 100 chronic inflammatory diseases afflicting more than 20 million Americans. Tolerogenic vaccines comprised of single-chain GM-CSF-neuroantigen (GMCSF-NAg) fusion proteins were shown in previous studies to prevent and reverse disease in multiple rodent models of experimental autoimmune encephalomyelitis (EAE) by a mechanism contingent upon the function of CD4+ CD25+ FOXP3+ regulatory T cells (Tregs). GMCSF-NAg vaccines inhibited EAE in both quiescent and inflammatory environments in association with low-efficiency T cell receptor (TCR) signaling events that elicited clonal expansion of immunosuppressive Tregs. Methods This study focused on two vaccines, including GMCSF-MOG (myelin oligodendrocyte glycoprotein 35–55/MOG35–55) and GMCSF-NFM (neurofilament medium peptide 13–37/NFM13–37), that engaged the transgenic 2D2 TCR with either low or high efficiencies, respectively. 2D2 mice were crossed with FOXP3 IRES eGFP (FIG) mice to track Tregs and further crossed with Rag−/− mice to reduce pre-existing Treg populations. Results This study provided evidence that low and high efficiency TCR interactions were integrated via CD40L expression levels to control the Treg/Tcon balance. The high-efficiency GMCSF-NFM vaccine elicited memory Tcon responses in association with activation of the CD40L costimulatory system. Conversely, the low-efficiency GMCSF-MOG vaccine lacked adequate TCR signal strength to elicit CD40L expression and instead elicited Tregs by a mechanism that was impaired by a CD40 agonist. When combined, the low- and high-efficiency GMCSF-NAg vaccines resulted in a balanced outcome and elicited both Tregs and Tcon responses without the predominance of a dominant immunogenic Tcon response. Aside from Treg expansion in 2D2-FIG mice, GMCSF-MOG caused a sustained decrease in TCR-β, CD3, and CD62L expression and a sustained increase in CD44 expression in Tcon subsets. Subcutaneous administration of GMCSF-MOG without adjuvants inhibited EAE in wildtype mice, which had a replete Treg repertoire, but was pathogenic rather than tolerogenic in 2D2-FIG-Rag1−/− mice, which lacked pre-existing Tregs. Conclusions This study provided evidence that the GMCSF-MOG vaccine elicited antigenic responses beneath the CD40L triggering threshold, which defined an antigenic niche that drove dominant expansion of tolerogenic myelin-specific Tregs that inhibited EAE.
Collapse
Affiliation(s)
- Cody D Moorman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Alexander G Bastian
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Kayla B DeOca
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
5
|
Yeola AP, Ignatius Arokia Doss PM, Baillargeon J, Akbar I, Mailhot B, Balood M, Talbot S, Anderson AC, Lacroix S, Rangachari M. Endogenous T Cell Receptor Rearrangement Represses Aggressive Central Nervous System Autoimmunity in a TcR-Transgenic Model on the Non-Obese Diabetic Background. Front Immunol 2020; 10:3115. [PMID: 32010149 PMCID: PMC6974510 DOI: 10.3389/fimmu.2019.03115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 12/20/2019] [Indexed: 12/16/2022] Open
Abstract
The T cell response to central nervous system (CNS) antigen in experimental autoimmune encephalomyelitis (EAE) permits one to model the immune aspects of multiple sclerosis. 1C6 transgenic mice on the non-obese diabetic (NOD) background possess a class II-restricted T cell receptor (TcR; Vα5-Vβ7) specific for the encephalitogenic peptide myelin oligodendrocyte glycoprotein (MOG)[35−55]. It remains to be determined what role is played by allelic inclusion in shaping the TcR repertoire of these mice. Here, we show that 1C6 T cells display substantial promiscuity in their expression of non-transgenically derived Vα chains. Further, enforced expression of the transgenic TcR in 1C6 × Rag1−/− mice profoundly disrupted thymic negative selection and led to a sharp decrease in the number of mature peripheral T cells. 1C6 × Rag1−/− mice developed spontaneous EAE at a significant frequency and rapidly developed fatal EAE upon immunization with myelin oligodendrocyte glycoprotein (MOG)[35−55]. Passive transfer of 1C6 × Rag1+/+ CD4+ T cells, but not CD8+ T cells or B cells, partially rescued 1C6 × Rag1−/− mice from severe EAE. FoxP3+ CD4+ Treg cells were present in the CNS of immunized 1C6 mice, as well as immunized 1C6 × Rag1−/− that had been supplemented with 1C6 CD4+ T cells. However, they were not observed in 1C6 × Rag1−/− that did not receive Rag1-sufficient 1C6 CD4+. Further, in vivo blockade of Treg accelerated the onset of symptoms in 1C6 mice immunized with MOG[35−55], indicating the pertinence of Treg-mediated control of autoimmune inflammation in this model. Thus, TcR allelic inclusion is crucial to the generation of FoxP3+ CD4+ T cells necessary for the suppression of severe CNS autoimmunity.
Collapse
Affiliation(s)
- Asmita Pradeep Yeola
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | | | - Joanie Baillargeon
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Irshad Akbar
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Benoit Mailhot
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Mohammad Balood
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Physiology and Pharmacology, Université de Montréal, Montréal, QC, Canada
| | - Sébastien Talbot
- Department of Physiology and Pharmacology, Université de Montréal, Montréal, QC, Canada
| | - Ana Carrizosa Anderson
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, United States
| | - Steve Lacroix
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Manu Rangachari
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
6
|
Lee G, Kang GH, Bae H. Bee venom phospholipase A2 suppression of experimental autoimmune encephalomyelitis is dependent on its enzymatic activity. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0034-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
7
|
Zhong SS, Xiang YJ, Liu PJ, He Y, Yang TT, Wang YY, Rong A, Zhang J, Liu GZ. Effect of Cordyceps sinensis on the Treatment of Experimental Autoimmune Encephalomyelitis: A Pilot Study on Mice Model. Chin Med J (Engl) 2017; 130:2296-2301. [PMID: 28937034 PMCID: PMC5634078 DOI: 10.4103/0366-6999.215335] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: As a traditional Chinese medicine, Cordyceps sinensis (CS) possesses a variety of immunoregulatory properties. This study aimed to explore the therapeutic potential of CS in a mice model of multiple sclerosis (MS)-experimental autoimmune encephalomyelitis (EAE). Methods: Female C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein35–55 to induce EAE, followed by an instant intragastric feeding with a low dosage of CS (low-CS group, n = 5), high dosage of CS (high-CS group, n = 5), or the same volume of normal saline (control group, n = 5). All the mice were observed for clinical assessment. Over the 30 days of CS treatment, flow cytometry was used to detect the frequency of helper T-cell (Th) subsets, Th1 and Th17, and CD4+ CD25+ regulatory T cells in the spleen and lymph nodes. Meanwhile, pathological changes in brain were determined using both hematoxylin-eosin and luxol fast blue staining. Data were analyzed using the one-way analysis of variance (ANOVA). Results: Over the 15 and 30 days of CS treatment, the clinical assessment for EAE demonstrated that both high-CS group (2.51 ± 0.31 and 2.26 ± 0.39 scores, respectively) and low-CS group (2.99 ± 0.40 and 2.69 ± 0.46, respectively) had lower disease severity scores than those of control group (3.57 ± 0.53 and 3.29 ± 0.53, all P < 0.01, respectively). Meanwhile, after 15 and 30 days, the high-CS group (19.18 ± 1.34 g and 20.41 ± 1.56 g, respectively) and low-CS group (18.07 ± 1.18 g and 19.48 ± 1.69 g, respectively) had a lower body weight, as compared with control group (16.85 ± 1.15 g and 18.22 ± 1.63 g, all P < 0.01, respectively). At 30 days post-CS treatment, there was a lower Th1 frequency in the lymph nodes (2.85 ± 1.54% and 2.77 ± 1.07% vs. 5.35 ± 1.34%, respectively; P < 0.05) and spleens (3.96 ± 1.09% and 3.09 ± 0.84% vs. 5.07 ± 1.50%, respectively; P < 0.05) and less inflammatory infiltration and demyelination in the brain of CS-treated mice than that of control group. Conclusions: Our preliminary study demonstrated that CS efficiently alleviated EAE severity and EAE-related pathology damage and decreased the number of Th1s in the periphery, indicating its effectiveness in the treatment of murine EAE. Thus, our findings strongly support the therapeutic potential of this agent as a new traditional Chinese medicine approach in MS treatment.
Collapse
Affiliation(s)
- Shan-Shan Zhong
- Department of Neurology, Peking University People's Hospital, Beijing 100044, China
| | - Ya-Juan Xiang
- Department of Neurology, The First Hospital of Tsinghua University, Beijing 100016, China
| | - Pen-Ju Liu
- Deptartment of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yang He
- Department of Neurology, Peking University People's Hospital, Beijing 100044, China
| | - Ting-Ting Yang
- Deptartment of Neurology, Tsinghua Changgung Hospital, Beijing 102218, China
| | - Yang-Yang Wang
- Department of Neurology, Peking University People's Hospital, Beijing 100044, China
| | - A Rong
- Department of Neurology, Peking University People's Hospital, Beijing 100044, China
| | - Jun Zhang
- Department of Neurology, Peking University People's Hospital, Beijing 100044, China
| | - Guang-Zhi Liu
- Department of Neurology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
8
|
Duffy SS, Keating BA, Perera CJ, Moalem-Taylor G. The role of regulatory T cells in nervous system pathologies. J Neurosci Res 2017; 96:951-968. [DOI: 10.1002/jnr.24073] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/28/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Samuel S. Duffy
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| | - Brooke A. Keating
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| | - Chamini J. Perera
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| | - Gila Moalem-Taylor
- School of Medical Sciences; University of New South Wales UNSW; Sydney Australia
| |
Collapse
|
9
|
Jain RW, Dang AK, Kerfoot SM. Simple and Efficient Production and Purification of Mouse Myelin Oligodendrocyte Glycoprotein for Experimental Autoimmune Encephalomyelitis Studies. J Vis Exp 2016. [PMID: 27842340 DOI: 10.3791/54727] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS), thought to occur as a result of autoimmune responses targeting myelin. Experimental autoimmune encephalomyelitis (EAE) is the most common animal model of CNS autoimmune disease, and is typically induced via immunization with short peptides representing immunodominant CD4+ T cell epitopes of myelin proteins. However, B cells recognize unprocessed protein directly, and immunization with short peptide does not activate B cells that recognize the native protein. As recent clinical trials of B cell-depleting therapies in MS have suggested a role for B cells in driving disease in humans, there is an urgent need for animal models that incorporate B cell-recognition of autoantigen. To this end, we have generated a new fusion protein containing the extracellular domain of the mouse version of myelin oligodendrocyte glycoprotein (MOG) as well as N-terminal fusions of a His-tag for purification purposes and the thioredoxin protein to improve solubility (MOGtag). A tobacco etch virus (TEV) protease cleavage site was incorporated to allow the removal of all tag sequences, leaving only the pure MOG1-125 extracellular domain. Here, we describe a simple protocol using only standard laboratory equipment to produce large quantities of pure MOGtag or MOG1-125. This protocol consistently generates over 200 mg of MOGtag protein. Immunization with either MOGtag or MOG1-125 generates an autoimmune response that includes pathogenic B cells that recognize the native mouse MOG.
Collapse
Affiliation(s)
- Rajiv W Jain
- Department of Microbiology and Immunology, Western University
| | - Amy K Dang
- Department of Microbiology and Immunology, Western University
| | | |
Collapse
|
10
|
Haghmorad D, Salehipour Z, Nosratabadi R, Rastin M, Kokhaei P, Mahmoudi MB, Amini AA, Mahmoudi M. Medium-dose estrogen ameliorates experimental autoimmune encephalomyelitis in ovariectomized mice. J Immunotoxicol 2016; 13:885-896. [PMID: 27602995 DOI: 10.1080/1547691x.2016.1223768] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Estrogen is a neuro-protective hormone in various central nervous system (CNS) disorders. The present study evaluated the role of estrogen during experimental autoimmune encephalomyelitis (EAE) at doses selected to mimic any suppressive potential from the hormone during pregnancy. Here, mice were ovariectomized and then 2 weeks later treated with MOG antigen to induce EAE. Concurrently, mice then received (subcutaneously) an implanted pellet to deliver varying estrogen amounts over a 21-day period. Clinical scores and other parameters were monitored daily for the 21 days. At the end of the period, brain/spinal cord histology was performed to measure lymphocyte infiltration; T-cell profiles were determined through ELISA, flow cytometry, and real-time PCR. Transcription factor expression levels in the CNS were assessed using real-time PCR; T-cell differentiation was evaluated via flow cytometry. The results demonstrated that estrogen inhibited development of EAE. Histological studies revealed limited leukocyte infiltration into the CNS. High and medium dose of estrogen increased TH2 and Treg cell production of interleukin (IL)-4, IL-10, and transforming growth factor (TGF)-β, but concurrently resulted in a significant reduction in production of interferon (IFN)-γ, IL-17, and IL-6. Flow cytometry revealed there were also significant decreases in the percentages of TH1 and TH17 cells, as well as significant increase in percentages of Treg and TH2 cells in the spleen and lymph nodes. Real-time PCR results indicated that high- and medium-dose estrogen treatments reduced T-bet and ROR-γt factor expression, but enhanced Foxp3 and GATA3 expression. Collectively, these results demonstrated that a medium dose of estrogen - similar to a pregnancy level of estrogen - could potentially reduce the incidence and severity of autoimmune EAE and possibly other autoimmune pathologies.
Collapse
Affiliation(s)
- Dariush Haghmorad
- a Department of Immunology, School of Medicine , Semnan University of Medical Sciences , Semnan , Iran
| | - Zohreh Salehipour
- b Department of Immunology and Allergy, School of Medicine, Immunology Research Center, BuAli Research Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Reza Nosratabadi
- c Immunology of Infectious Diseases Research Center , Rafsanjan University of Medical Sciences , Rafsanjan , Iran
| | - Maryam Rastin
- b Department of Immunology and Allergy, School of Medicine, Immunology Research Center, BuAli Research Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Parviz Kokhaei
- a Department of Immunology, School of Medicine , Semnan University of Medical Sciences , Semnan , Iran
| | | | - Abbas Ali Amini
- e Department of Immunology, Faculty of Medicine , Kurdistan University of Medical Sciences , Sanandaj , Iran
| | - Mahmoud Mahmoudi
- b Department of Immunology and Allergy, School of Medicine, Immunology Research Center, BuAli Research Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
11
|
Lee SW, Park HJ, Jeon SH, Lee C, Seong RH, Park SH, Hong S. Ubiquitous Over-Expression of Chromatin Remodeling Factor SRG3 Ameliorates the T Cell-Mediated Exacerbation of EAE by Modulating the Phenotypes of both Dendritic Cells and Macrophages. PLoS One 2015; 10:e0132329. [PMID: 26147219 PMCID: PMC4492541 DOI: 10.1371/journal.pone.0132329] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/14/2015] [Indexed: 12/18/2022] Open
Abstract
Although SWI3-related gene (SRG3), a chromatin remodeling factor, is critical for various biological processes including early embryogenesis and thymocyte development, it is unclear whether SRG3 is involved in the differentiation of CD4+ T cells, the key mediator of adaptive immune responses. Because it is known that experimental autoimmune encephalomyelitis (EAE) development is determined by the activation of CD4+ T helper cells, here, we investigated the role of SRG3 in EAE development using SRG3 transgenic mouse models exhibiting two distinct SRG3 expression patterns: SRG3 expression driven by either the CD2 or β-actin promoter. We found that the outcome of EAE development was completely different depending on the expression pattern of SRG3. The specific over-expression of SRG3 using the CD2 promoter facilitated EAE via the induction of Th1 and Th17 cells, whereas the ubiquitous over-expression of SRG3 using the β-actin promoter inhibited EAE by promoting Th2 differentiation and suppressing Th1 and Th17 differentiation. In addition, the ubiquitous over-expression of SRG3 polarized CD4+ T cell differentiation towards the Th2 phenotype by converting dendritic cells (DCs) or macrophages to Th2 types. SRG3 over-expression not only reduced pro-inflammatory cytokine production by DCs but also shifted macrophages from the inducible nitric oxide synthase (iNOS)-expressing M1 phenotype to the arginase-1-expressing M2 phenotype during EAE. In addition, Th2 differentiation in β-actin-SRG3 Tg mice during EAE was associated with an increase in the basophil and mast cell populations and in IL4 production. Furthermore, the increased frequency of Treg cells in the spinal cord of β-actin-SRG3 Tg mice might induce the suppression of and accelerate the recovery from EAE symptoms. Taken together, our results provide the first evidence supporting the development of a new therapeutic strategy for EAE involving the modulation of SRG3 expression to induce M2 and Th2 polarization, thereby inhibiting inflammatory immune responses.
Collapse
Affiliation(s)
- Sung Won Lee
- Dept. of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul 143–747, Korea
- School of Life Sciences and Biotechnology, Korea University, Seoul 136–701, Korea
| | - Hyun Jung Park
- Dept. of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul 143–747, Korea
| | - Sung Ho Jeon
- Dept. of Life Science, Hallym University, Chuncheon 200–702, Korea
| | - Changjin Lee
- Dept. of Biological Sciences, Institute of Molecular Biology and Genetics, Research Center for Functional Cellulomics, Seoul National University, Seoul 151–742, Korea
| | - Rho Hyun Seong
- Dept. of Biological Sciences, Institute of Molecular Biology and Genetics, Research Center for Functional Cellulomics, Seoul National University, Seoul 151–742, Korea
| | - Se-Ho Park
- School of Life Sciences and Biotechnology, Korea University, Seoul 136–701, Korea
| | - Seokmann Hong
- Dept. of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul 143–747, Korea
- * E-mail:
| |
Collapse
|
12
|
Dang AK, Jain RW, Craig HC, Kerfoot SM. B cell recognition of myelin oligodendrocyte glycoprotein autoantigen depends on immunization with protein rather than short peptide, while B cell invasion of the CNS in autoimmunity does not. J Neuroimmunol 2015; 278:73-84. [DOI: 10.1016/j.jneuroim.2014.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/12/2014] [Accepted: 12/08/2014] [Indexed: 10/24/2022]
|
13
|
Dai H, Ciric B, Zhang GX, Rostami A. Interleukin-10 plays a crucial role in suppression of experimental autoimmune encephalomyelitis by Bowman-Birk inhibitor. J Neuroimmunol 2012; 245:1-7. [PMID: 22365083 DOI: 10.1016/j.jneuroim.2012.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/29/2011] [Accepted: 01/12/2012] [Indexed: 11/26/2022]
Abstract
The Bowman-Birk inhibitor (BBI) is a soybean-derived serine protease inhibitor with anti-inflammatory properties. Experimental autoimmune encephalomyelitis (EAE) serves as an animal model of the central nervous system (CNS) inflammatory disorder multiple sclerosis (MS). EAE is mediated by Th1 and Th17 cells which migrate into the CNS and initiate inflammation directed against myelin components, resulting in CNS pathology and neurological clinical deficit. We have shown previously that oral treatment with BBI delays onset of EAE and reduces its severity. These beneficial effects were associated with an increase in IL-10 secretion by immune cells of BBI-treated mice. It is not known, however, whether this was a causal relationship or simply an epiphenomenon. In the present study we provide evidence that BBI regulates CD4+ T cell immune responses in EAE. BBI administration delayed the onset of EAE and reduced its severity in an IL-10-dependent manner, as BBI-mediated suppression of EAE was abrogated in IL-10 knockout mice. The beneficial effects were accompanied by reduced IFN-γ, IL-17 and increased IL-10 production, as well as increased Foxp3 expression. CD4+ T cells were the major source of IL-10 in the periphery and in the CNS during BBI treatment. Furthermore, BBI-treated mice had reduced numbers of infiltrated cells in the CNS, including Th17 cells, as compared with PBS-treated control animals. In conclusion, our data provide clear evidence for the essential role of IL-10 in BBI-mediated suppression in EAE, and indicate that BBI may be a promising candidate for the development of a novel MS therapy.
Collapse
Affiliation(s)
- Hong Dai
- Department of Neurology, Thomas Jefferson University, Jefferson Hospital for Neuroscience, 900 Walnut Street, Suite 200, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
14
|
Engler JB, Undeutsch R, Kloke L, Rosenberger S, Backhaus M, Schneider U, Egerer K, Dragun D, Hofmann J, Huscher D, Burmester GR, Humrich JY, Enghard P, Riemekasten G. Unmasking of autoreactive CD4 T cells by depletion of CD25 regulatory T cells in systemic lupus erythematosus. Ann Rheum Dis 2011; 70:2176-83. [PMID: 21926188 DOI: 10.1136/ard.2011.153619] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Autoreactive CD4 T cells specific for nuclear peptide antigens play an important role in tolerance breakdown during the course of systemic lupus erythematosus (SLE). However, reliable detection of these cells is limited due to their low frequency in peripheral blood. The authors assess autoreactive CD4 T cells in a representative SLE collective (n=38) by flow cytometry and study the influence of regulatory T cells (Treg) on their antigenic challenge. METHODS CD4 T-cell responses were determined according to intracellular CD154 expression induced after 6-h short-term in-vitro stimulation with the SLE-associated autoantigen SmD1(83-119). To clarify the influence of Treg on the activation of autoreactive CD4 T cells, CD25 Treg were depleted by magnetic activated cell sorting before antigen-specific stimulation in selected experiments. RESULTS In the presence of Treg, autoreactive CD4 T-cell responses to SmD1(83-119) were hardly observable. However, Treg removal significantly increased the frequency of detectable SmD1(83-119)-specific CD4 T cells in SLE patients but not in healthy individuals. Consequently, by depleting Treg the percentage of SmD1(83-119)-reactive SLE patients increased from 18.2% to 63.6%. This unmasked autoreactivity of CD4 T cells correlated with the disease activity as determined by the SLE disease activity index (p=0.005*, r=0.779). CONCLUSIONS These data highlight the pivotal role of the balance between autoreactive CD4 T cells and CD25 Treg in the dynamic course of human SLE. Analysing CD154 expression in combination with a depletion of CD25 Treg, as shown here, may be of further use in approaching autoantigen-specific CD4 T cells in SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Jan Broder Engler
- Department of Rheumatology and Clinical Immunology, University Hospital Charité, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Chen XT, Chan ST, Hosseini H, Layton D, Boyd R, Alderuccio F, Toh BH, Chan J. Transplantation of retrovirally transduced bone marrow prevents autoimmune disease in aged mice by peripheral tolerance mechanisms. Autoimmunity 2011; 44:384-93. [DOI: 10.3109/08916934.2010.541173] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
16
|
Subramanian S, Yates M, Vandenbark AA, Offner H. Oestrogen-mediated protection of experimental autoimmune encephalomyelitis in the absence of Foxp3+ regulatory T cells implicates compensatory pathways including regulatory B cells. Immunology 2010; 132:340-7. [PMID: 21091909 DOI: 10.1111/j.1365-2567.2010.03380.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oestrogen (17β-oestradiol, E₂) is a highly effective treatment for experimental autoimmune encephalomyelitis (EAE) that may potentiate Foxp3+ regulatory T (Treg) cells, which in turn limit the expansion of encephalitogenic T-cell specificities. To determine if Treg cells constitute the major non-redundant protective pathway for E₂, we evaluated E₂ protection of EAE after targeted deletion of Foxp3 expression in Foxp3-DTR mice. Unexpectedly, E₂-treated Foxp3-deficient mice were completely protected against clinical and histological myelin oligodendrocyte glycoprotein (MOG)-35-55 peptide-induced EAE before succumbing to diphtheria toxin-induced mortality. This finding indicated the presence of alternative E₂-dependent EAE-protective pathways that could compensate for the lack of Treg cells. Further investigation revealed that E₂ treatment inhibited proliferation and expression of CCL2 and CXCL2, but enhanced secretion of interleukin-10 (IL-10) and IL-13 by MOG-35-55-specific spleen cells. These changes occurred concomitantly with increased expression of several chemokines and receptors, including CXCL13 and CXCR5, and the negative co-activation molecules, PD-L1 and B7.2, by B cells and dendritic cells. Furthermore, E₂ treatment resulted in higher percentages of spleen and lymph node T cells expressing IL-17, interferon-γ and tumour necrosis factor-α, but with lower expression of CCR6, suggesting sequestration of MOG-35-55 peptide-specific T cells in peripheral immune organs. Taken together, these data suggest that E₂-induced mechanisms that provide protection against EAE in the absence of Foxp3+ Treg cells include induction of regulatory B cells and peripheral sequestration of encephalitogenic T cells.
Collapse
Affiliation(s)
- Sandhya Subramanian
- Neuroimmunology Research, Veterans' Affairs Medical Center, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
17
|
Larocque D, Sanderson NSR, Bergeron J, Curtin JF, Girton J, Wibowo M, Bondale N, Kroeger KM, Yang J, Lacayo LM, Reyes KC, Farrokhi C, Pechnick RN, Castro MG, Lowenstein PR. Exogenous fms-like tyrosine kinase 3 ligand overrides brain immune privilege and facilitates recognition of a neo-antigen without causing autoimmune neuropathology. Proc Natl Acad Sci U S A 2010; 107:14443-8. [PMID: 20660723 PMCID: PMC2922551 DOI: 10.1073/pnas.0913496107] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Soluble antigens diffuse out of the brain and can thus stimulate a systemic immune response, whereas particulate antigens (from infectious agents or tumor cells) remain within brain tissue, thus failing to stimulate a systemic immune response. Immune privilege describes how the immune system responds to particulate antigens localized selectively within the brain parenchyma. We believe this immune privilege is caused by the absence of antigen presenting dendritic cells from the brain. We tested the prediction that expression of fms-like tyrosine kinase ligand 3 (Flt3L) in the brain will recruit dendritic cells and induce a systemic immune response against exogenous influenza hemagglutinin in BALB/c mice. Coexpression of Flt3L with HA in the brain parenchyma induced a robust systemic anti-HA immune response, and a small response against myelin basic protein and proteolipid protein epitopes. Depletion of CD4(+)CD25+ regulatory T cells (Tregs) enhanced both responses. To investigate the autoimmune impact of these immune responses, we characterized the neuropathological and behavioral consequences of intraparenchymal injections of Flt3L and HA in BALB/c and C57BL/6 mice. T cell infiltration in the forebrain was time and strain dependent, and increased in animals treated with Flt3L and depleted of Tregs; however, we failed to detect widespread defects in myelination throughout the forebrain or spinal cord. Results of behavioral tests were all normal. These results demonstrate that Flt3L overcomes the brain's immune privilege, and supports the clinical development of Flt3L as an adjuvant to stimulate clinically effective immune responses against brain neo-antigens, for example, those associated with brain tumors.
Collapse
Affiliation(s)
- Daniel Larocque
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - Nicholas S. R. Sanderson
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - Josée Bergeron
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - James F. Curtin
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - Joe Girton
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - Mia Wibowo
- Gene Therapeutics Research Institute
- Cedars-Sinai Biomedical Sciences Graduate Program, and
| | - Niyati Bondale
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - Kurt M. Kroeger
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - Jieping Yang
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
| | - Liliana M. Lacayo
- Department of Psychiatry and Behavioral Neurosciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Kevin C. Reyes
- Department of Psychiatry and Behavioral Neurosciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Catherine Farrokhi
- Department of Psychiatry and Behavioral Neurosciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
| | - Robert N. Pechnick
- Department of Psychiatry and Behavioral Neurosciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048; and
- Brain Research Institute, and
| | - Maria G. Castro
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
- Brain Research Institute, and
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| | - Pedro R. Lowenstein
- Gene Therapeutics Research Institute
- Department of Medicine and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine
- Brain Research Institute, and
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| |
Collapse
|
18
|
Crucial role of interleukin-7 in T helper type 17 survival and expansion in autoimmune disease. Nat Med 2010; 16:191-7. [PMID: 20062065 DOI: 10.1038/nm.2077] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 11/18/2009] [Indexed: 01/01/2023]
Abstract
Interleukin-7 receptor (IL-7R) is genetically associated with susceptibility to multiple sclerosis. Here we describe that IL-7 is essential for survival and expansion of pathogenic T helper type 17 (T(H)17) cells in experimental autoimmune encephalomyelitis (EAE). IL-7 directly expanded effector T(H)17 cells in EAE and human T(H)17 cells from subjects with multiple sclerosis, whereas it was not required for T(H)17 differentiation. IL-7R antagonism rendered differentiated T(H)17 cells susceptible to apoptosis through the inhibition of Janus kinase-signal transducer and activator of transcription-5 (JAK-STAT5) pathway and altered expression of the prosurvival protein Bcl-2 and the proapoptotic protein Bax, leading to decreased severity of EAE. In contrast, T(H)1 and regulatory T (T(reg)) cells were less susceptible to or not affected by IL-7R antagonism in vivo. The selectivity was attributable to minimal expression of IL-7Ralpha in T(reg) cells and correlated with a high level of Socs1 (encoding suppressor of cytokine signaling-1) expression in T(H)1 cells. The study reveals a unique, previously undescribed role of IL-7-IL-7R in T(H)17 cell survival and expansion and has implications in the treatment of autoimmune disease.
Collapse
|
19
|
Riordan NH, Ichim TE, Min WP, Wang H, Solano F, Lara F, Alfaro M, Rodriguez JP, Harman RJ, Patel AN, Murphy MP, Lee RR, Minev B. Non-expanded adipose stromal vascular fraction cell therapy for multiple sclerosis. J Transl Med 2009; 7:29. [PMID: 19393041 PMCID: PMC2679713 DOI: 10.1186/1479-5876-7-29] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Accepted: 04/24/2009] [Indexed: 12/18/2022] Open
Abstract
The stromal vascular fraction (SVF) of adipose tissue is known to contain mesenchymal stem cells (MSC), T regulatory cells, endothelial precursor cells, preadipocytes, as well as anti-inflammatory M2 macrophages. Safety of autologous adipose tissue implantation is supported by extensive use of this procedure in cosmetic surgery, as well as by ongoing studies using in vitro expanded adipose derived MSC. Equine and canine studies demonstrating anti-inflammatory and regenerative effects of non-expanded SVF cells have yielded promising results. Although non-expanded SVF cells have been used successfully in accelerating healing of Crohn's fistulas, to our knowledge clinical use of these cells for systemic immune modulation has not been reported. In this communication we discuss the rationale for use of autologous SVF in treatment of multiple sclerosis and describe our experiences with three patients. Based on this rationale and initial experiences, we propose controlled trials of autologous SVF in various inflammatory conditions.
Collapse
|