1
|
Scherrer EC, Ramos KA, Valadares YM, Soares IGM, Carli AP, Sá Silva F, Silva JG, Alvarenga DG, Brugiolo ASS, Verly RM, Salvador MR, Denadai AML, Souza Alves CC, Castro SBR. Ursolic acid derivatives improved clinical signs of experimental autoimmune encephalomyelitis by modulating central nervous system inflammation. Metab Brain Dis 2025; 40:166. [PMID: 40167825 DOI: 10.1007/s11011-025-01591-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
The immunopathogenesis of multiple sclerosis (MS) involves the activation of T lymphocytes, leading to progressive axonal loss and brain atrophy. Ursolic acid (AU) has been widely used as an herbal medicine, with the ability to inhibit the production and secretion of cytokines and may influence the differentiation of CD4 + helper cells. In this study, we aimed to investigate the immunomodulatory effects of ursolic acid derivatives (methyl 3β-hydroxyurs-12-en-28-oate (AUD1) and methyl 3β-acetoxyurs-12-en-28-oate (AUD2)) in a model of experimental autoimmune encephalomyelitis (EAE). EAE was induced by subcutaneous immunization of myelin oligodendrocyte glycoprotein peptide (MOG35-55) in C57BL/6 mice. On the 15th day post-induction (dpi), the mice were treated with AU, AUD1, or AUD2 (50 mg/kg intraperitoneally per day) for six days. Clinical signs were monitored until 21 dpi, and parameters were assessed in the spinal cord, lymph nodes, and brain at 21 dpi. The results showed that both derivatives similarly attenuated the clinical signs of EAE and reduced inflammation and demyelination in the spinal cord. In addition, they reduced the number of pro-inflammatory cells in the brain, the level of IL-1β, TNF, and IFN-γ in the spinal cord, and, in the periphery, promoted the regulation of pro-inflammatory cells. In conclusion, regulating cells in the periphery and reducing the number of pro-inflammatory cells in the CNS, with AUD1 and AUD2, culminated in the efficacy of the clinical parameters presented in EAE, suggesting a therapeutic potential for treating MS.
Collapse
Affiliation(s)
- Elaine Carlos Scherrer
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Karla Antunes Ramos
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil
| | - Ydia Mariele Valadares
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Igor Gabriel Machado Soares
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil
| | - Alessandra Paula Carli
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil
| | - Fernando Sá Silva
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Jeferson Gomes Silva
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | - Daniel Gomes Alvarenga
- Life Sciences Institute, Federal University of Juiz de Fora, Governador Valadares, MG, Brazil
| | | | - Rodrigo Moreira Verly
- Department of Chemistry, Federal University of the Valleys of Jequitinhonha and Mucuri, Diamantina, MG, Brazil
| | | | | | - Caio César Souza Alves
- Faculty of Medicine, Federal University of the Valleys of Jequitinhonha and Mucuri, MG, Teófilo Otoni, Brazil.
- Faculdade de Medicina do Mucuri, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Rua do Cruzeiro, 01, Jardim São Paulo, Teófilo Otoni/MG, Brasil, 39803-371.
| | | |
Collapse
|
2
|
Mohammadi M, Yarmohammadi A, Salehi-Abargouei A, Ghasemirad H, Shirvani M, Ghoshouni H. Uric acid and glaucoma: a systematic review and meta-analysis. Front Med (Lausanne) 2023; 10:1159316. [PMID: 37575992 PMCID: PMC10422028 DOI: 10.3389/fmed.2023.1159316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
BACKGROUND Glaucoma, the leading cause of irreversible blindness, is a common disorder that contributes to gradual optic nerve degeneration. The beneficial impacts of uric acid (UA) have been reported in some neurodegenerative conditions such as Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. But the results of current studies about the association between serum UA level and glaucoma are conflicting. The present meta-analysis was conducted to provide a better understanding of the association between serum UA level and glaucoma. METHODS We searched the databases of PubMed, Scopus, Web of Science, and Google Scholar systematically until November 20, 2022 to identify case-control studies, comparing the serum UA concentrations of the patients with glaucoma and controls. The mean ± standard division difference was used to assess the difference in serum UA concentrations between the glaucoma patients and controls. RESULTS Six studies involving 1,221 glaucoma patients and 1,342 control group were included in the present meta-analysis. This meta-analysis using a random effect model indicated that the mean UA level in glaucoma patients was 0.13 (I2 = 91.92%, 95% CI = -0.42 to 0.68) higher than the controls; however, it was not statistically significant. CONCLUSIONS Our findings provide evidence that glaucoma patients have a higher serum UA level compared to the controls, but this difference is not statistically significant. Prospective studies are needed to determine the possible association between increased UA and glaucoma pathogenesis. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022364055, identifier: CRD42022364055.
Collapse
Affiliation(s)
- Mohammad Mohammadi
- Students' Research and Technology Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- NeuroTRACT Association, Student's Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amin Salehi-Abargouei
- Research Center for Food Hygiene and Safety, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamidreza Ghasemirad
- Students' Research and Technology Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Shirvani
- Geriatric Ophthalmology Research Center, Shahid Sadoughi University of Medical Science, Yazd, Iran
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Ghoshouni
- Students' Research and Technology Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
3
|
Brod SA. The genealogy, methodology, similarities and differences of immune reconstitution therapies for multiple sclerosis and neuromyelitis optica. Autoimmun Rev 2022; 21:103170. [PMID: 35963569 DOI: 10.1016/j.autrev.2022.103170] [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: 08/01/2022] [Accepted: 08/07/2022] [Indexed: 11/09/2022]
Abstract
Immune reconstitution therapies (IRTs) are a type of short course procedure or pharmaceutical agent within the MS pharmacopeia. They emanate from oncology and induce transient incomplete lympho-ablation with or without myelo-ablation, resulting in potential prolonged immunomodulation. Thus, they provide significant prophylaxis from disease activity without retreatment. Modern IRT for autoimmunity encompasses a heterogeneous group of pulsed lympho- and non-myelo-ablative treatments designed to re-boot the adaptive immune system in a quasi-permanent manner - a re-induction of ontogeny. IRT is the extensive debulking of an auto-aggressive immune system to attempt to reach the Holy Grail of immune tolerance. This incomplete yet significant lympho-ablation induces lymphoproliferation, reduces pathogenic clonal cells, causes thymopoiesis and results in the induction of immune tolerance. Lympho-ablation with immune reconstitution can result in minimal residual autoimmunity. There is a resetting of the immune thermostat - i.e., the immunostat. IRTs have the potential to provide prolonged periods of disease inactivity without retreatment in part through the immunological results of their pulsatile lymphocyte depletion. It is vital to increase our understanding of how IRTs alter a patient's immune response to the antigenic target of the disease so that we can devise newer, more durable and safer forms of such agents. What common features do extant IRTs (i.e., stem cell transplant, alemtuzumab and oral cladribine) have to produce the durable therapeutic response without long term treatment in neuroimmunological diseases such as MS (multiple sclerosis) and NMOSD (neuromyelitis optica spectrum disorders)? Can we learn from these critical features to predict what other maneuvers or agents might effect similar clinical results with equal or greater efficacy and safety?
Collapse
Affiliation(s)
- Staley A Brod
- Division of MS/Neuro-immunology, Department of Neurology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226, USA.
| |
Collapse
|
4
|
Kiapour N, Wu B, Wang Y, Seyedsadr M, Kapoor S, Zhang X, Elzoheiry M, Kasimoglu E, Wan Y, Markovic-Plese S. Therapeutic Effect of Anti-CD52 Monoclonal Antibody in Multiple Sclerosis and Its Animal Models Is Mediated via T Regulatory Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:49-56. [PMID: 35750335 PMCID: PMC9458467 DOI: 10.4049/jimmunol.2100176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
The objective of this study is to determine the mechanism of action of anti-CD52 mAb treatment in patients with relapsing-remitting multiple sclerosis (RRMS). Experimental autoimmune encephalomyelitis (EAE), an animal model of the disease, was used to address the role of T regulatory cells (Tregs) in the anti-CD52 mAb-induced suppression of the disease. In vitro studies on PBMCs from RRMS patients and matched healthy controls determined the effect of IL-7 on the expansion of CD4+CD25+CD127- Tregs and induction of their suppressive phenotype. This study using EAE animal models of MS has shown that mouse anti-CD52 mAb suppression of clinical disease was augmented by coadministration of IL-7 and partially reversed by anti-IL-7 mAb. In vitro human studies showed that IL-7 induced expansion of CD4+CD25+CD127- Tregs and increased their FOXP3, GITIR, CD46, CTLA-4, granzyme B, and perforin expression. Anti-CD52 mAb treatment of mice with relapsing-remitting EAE induced expansion of Foxp3+CD4+ Tregs and the suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in peripheral immune organs and CNS infiltrates. The effect was detected immediately after the treatment and maintained over long-term follow-up. Foxp3+CD4+ Treg-mediated suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in the spinal cord infiltrates was reversed after inducible Foxp3 depletion. Our results demonstrated that the therapeutic effect of U.S. Food and Drug Administration-approved anti-CD52 mAb is dependent on the presence of Tregs.
Collapse
Affiliation(s)
- Nazanin Kiapour
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Bing Wu
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Frontier Science Center for Immunology and Metabolism of Medical Research Institute, Wuhan University, Wuhan, China
| | - Yan Wang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | | | - Sahil Kapoor
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Xin Zhang
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Duke Molecular Physiology Institute, Department of Orthopedic Surgery, Duke University, Durham, NC
| | - Manal Elzoheiry
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | - Ezgi Kasimoglu
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | - Yisong Wan
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Silva Markovic-Plese
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC;
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| |
Collapse
|
5
|
Barbour M, Wood R, Harte T, Bushell TJ, Jiang HR. Anti-CD52 antibody treatment in murine experimental autoimmune encephalomyelitis induces dynamic and differential modulation of innate immune cells in peripheral immune and central nervous systems. Immunology 2021; 165:312-327. [PMID: 34826154 PMCID: PMC9426620 DOI: 10.1111/imm.13437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/31/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022] Open
Abstract
Anti‐CD52 antibody (anti‐CD52‐Ab) leads to a rapid depletion of T and B cells, followed by reconstitution of immune cells with tolerogenic characteristics. However, very little is known about its effect on innate immune cells. In this study, experimental autoimmune encephalomyelitis mice were administered murine anti‐CD52‐Ab to investigate its effect on dendritic cells and monocytes/macrophages in the periphery lymphoid organs and the central nervous system (CNS). Our data show that blood and splenic innate immune cells exhibited significantly increased expression of MHC‐II and costimulatory molecules, which was associated with increased capacity of activating antigen‐specific T cells, at first day but not three weeks after five daily treatment with anti‐CD52‐Ab in comparison with controls. In contrast to the periphery, microglia and infiltrating macrophages in the CNS exhibited reduced expression levels of MHC‐II and costimulatory molecules after antibody treatment at both time‐points investigated when compared to controls. Furthermore, the transit response of peripheral innate immune cells to anti‐CD52‐Ab treatment was also observed in the lymphocyte‐deficient SCID mice, suggesting the changes are not a direct consequence of the mass depletion of lymphocytes in the periphery. Our study demonstrates a dynamic and tissue‐specific modulation of the innate immune cells in their phenotype and function following the antibody treatment. The findings of differential modulation of the microglia and infiltrating macrophages in the CNS in comparison with the innate immune cells in the peripheral organs support the CNS‐specific beneficial effect of alemtuzumab treatment on inhibiting neuroinflammation in multiple sclerosis patients.
Collapse
Affiliation(s)
- Mark Barbour
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Rachel Wood
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Tanith Harte
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Trevor J Bushell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Hui-Rong Jiang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
6
|
Dziedziak J, Kasarełło K, Cudnoch-Jędrzejewska A. Dietary Antioxidants in Age-Related Macular Degeneration and Glaucoma. Antioxidants (Basel) 2021; 10:antiox10111743. [PMID: 34829613 PMCID: PMC8614766 DOI: 10.3390/antiox10111743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Age-related macular degeneration (AMD) and glaucoma are ophthalmic neurodegenerative diseases responsible for irreversible vision loss in the world population. Only a few therapies can be used to slow down the progression of these diseases and there are no available treatment strategies for reversing the degeneration of the neural retina. In AMD, the pathological process causes the malfunction and damage of the retinal pigmented epithelium and photoreceptors in the macula. In glaucoma, damage of the retinal ganglion cells and their axons is observed and treatment strategies are limited to intraocular pressure lowering. Therefore, other prophylactic and/or therapeutic methods are needed. Oxidative stress is involved in the neurodegenerative process accompanying both AMD and glaucoma; therefore, the use of antioxidant agents would clearly be beneficial, which is supported by the decreased prevalence and progression of AMD in patients adherent to a diet naturally rich in antioxidants. Dietary antioxidants are easily available and their use is based on the natural route of administration. Many preclinical studies both in vitro and using animal models of retinal degeneration showed the efficacy of dietary antioxidants, which was further proved in clinical trials. Resveratrol is beneficial both in AMD and glaucoma animal models, but confirmed only among AMD patients. For AMD, carotenoids and omega-3 fatty acids were also proved to be sufficient in preventing neurodegeneration. For glaucoma, coenzyme Q10 and alpha-lipoic acid showed efficacy for decreasing retinal ganglion cell loss and inhibiting the accompanying destructive processes. Interestingly, the benefits of vitamins, especially vitamin E was not confirmed, neither in preclinical nor in clinical studies.
Collapse
|
7
|
Kasarello K, Mirowska-Guzel D. Anti-CD52 Therapy for Multiple Sclerosis: An Update in the COVID Era. Immunotargets Ther 2021; 10:237-246. [PMID: 34268256 PMCID: PMC8273745 DOI: 10.2147/itt.s240890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/22/2021] [Indexed: 01/01/2023] Open
Abstract
CD52 is a small surface glycoprotein composed of 12 amino acids. CD52 is found mostly on the surface of mature immune cells, such as lymphocytes, monocytes, eosinophils, and dendritic cells, as well as the male genital tract: within the epididymis and on the surface of mature sperm. Low CD52 expression is also found in neutrophils. CD52 function is not fully understood, although experiments with anti-CD52 antibodies have shown that CD52 is essential for lymphocyte transendothelial migration and may contribute to costimulation of CD4+ T cells and T-cell activation and proliferation. Although knowledge about exact CD52 function is still poor, CD52 presence on the surface of a broad spectrum of immune cells makes it a therapeutic target, especially in immunomediated diseases, such as multiple sclerosis. In multiple sclerosis, alemtuzumab is registered for adult patients with the relapsing-remitting form of the disease defined by clinical and imaging features. Despite the high efficacy of the drug, the main issue is its safety. The main adverse effects of alemtuzumab are associated with drug infusion due to cytokine release and cytotoxic effects of antibodies associated with lymphocyte depletion, which leads to immunosuppression, and secondary autoimmunity that may be the effect of excessive B-cell repopulation and cancer. This review presents current knowledge on the drug's mechanism of action, efficacy and safety data from clinical trials, and real-world observations, including available though scarce data on using alemtuzumab in the COVID era.
Collapse
Affiliation(s)
- Kaja Kasarello
- Department of Experimental and Clinical Physiology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| |
Collapse
|
8
|
B Cells and Microbiota in Autoimmunity. Int J Mol Sci 2021; 22:ijms22094846. [PMID: 34063669 PMCID: PMC8125537 DOI: 10.3390/ijms22094846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Trillions of microorganisms inhabit the mucosal membranes maintaining a symbiotic relationship with the host's immune system. B cells are key players in this relationship because activated and differentiated B cells produce secretory immunoglobulin A (sIgA), which binds commensals to preserve a healthy microbial ecosystem. Mounting evidence shows that changes in the function and composition of the gut microbiota are associated with several autoimmune diseases suggesting that an imbalanced or dysbiotic microbiota contributes to autoimmune inflammation. Bacteria within the gut mucosa may modulate autoimmune inflammation through different mechanisms from commensals ability to induce B-cell clones that cross-react with host antigens or through regulation of B-cell subsets' capacity to produce cytokines. Commensal signals in the gut instigate the differentiation of IL-10 producing B cells and IL-10 producing IgA+ plasma cells that recirculate and exert regulatory functions. While the origin of the dysbiosis in autoimmunity is unclear, compelling evidence shows that specific species have a remarkable influence in shaping the inflammatory immune response. Further insight is necessary to dissect the complex interaction between microorganisms, genes, and the immune system. In this review, we will discuss the bidirectional interaction between commensals and B-cell responses in the context of autoimmune inflammation.
Collapse
|
9
|
Remez L, Ganelin-Cohen E, Safina D, Hellmann MA, Lotan I, Bosak N, Buxbaum C, Vaknin A, Shifrin A, Rozenberg A. Alemtuzumab mediates the CD39 + T-regulatory cell response via CD23 + macrophages. Immunol Cell Biol 2021; 99:521-531. [PMID: 33306219 DOI: 10.1111/imcb.12431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/22/2020] [Accepted: 12/08/2020] [Indexed: 11/24/2022]
Abstract
Alemtuzumab (ALM) effectively prevents relapses of multiple sclerosis (MS). It causes lymphocyte depletion with subsequent enhancement of the T-regulatory cell population. Direct administration of ALM to T cells causes cytolysis. However, the T cells may be indirectly affected by monocyte-derived cells, which are resistant to ALM cytotoxicity. We aimed to examine whether ALM modulates monocytes and whether the crosstalk between monocytes and lymphocytes previously exposed to ALM would result in anti-inflammatory effects. The CD14+ monocytes of 10 healthy controls and 10 MS (treatment naive) patients were isolated from peripheral blood mononuclear cells (PBMCs), exposed to ALM and reintroduced to PBMCs depleted of CD14+ cells. The macrophage profile was assessed and T-cell markers were measured. ALM promoted M2 anti-inflammatory phenotype as noted by an increased percentage in the populations of CD23+ , CD83+ and CD163+ cells. The CD23+ cells were the most upregulated (7-fold, P = 0.0002), and the observed effect was higher in patients with MS than in healthy subjects. ALM-exposed macrophages increased the proportion of T-regulatory cells, without affecting the proportion of T-effector cells. Neutralizing the CD23+ monocytes with antibodies reversed the effect specifically on the CD4+ CD39+ T-regulatory cell subpopulation but not on the CD4+ CD25hi CD127lo FOXP3+ subpopulation. ALM induces the conversion of monocytes into anti-inflammatory macrophages, which in turn promotes T-regulatory cell enhancement, in a CD23-dependent manner. These findings suggest that the mechanism of action of ALM is relevant to aspects of MS pathogenesis.
Collapse
Affiliation(s)
- Lital Remez
- Neuroimmunology Laboratory, Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Esther Ganelin-Cohen
- Neuroimmunological Clinic, Institute of Pediatric Neurology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dina Safina
- Neuroimmunology Laboratory, Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Mark A Hellmann
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neurology, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Itay Lotan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Neurology, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Noam Bosak
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Chen Buxbaum
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Adi Vaknin
- Unit for Neuro-Immunology, Multiple Sclerosis & Cell Therapy, Department of Neurology, Hadassah Medical Center, Jerusalem, Israel
| | - Alla Shifrin
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Ayal Rozenberg
- Neuroimmunology Laboratory, Department of Neurology, Rambam Health Care Campus, Haifa, Israel.,Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
10
|
Bogie JF, Grajchen E, Wouters E, Broux B, Stinissen P, Van Wijmeersch B, Hendriks JJ. CNS delivery of anti-CD52 antibodies modestly reduces disease severity in an animal model for multiple sclerosis. Ther Adv Chronic Dis 2020; 11:2040622320947378. [PMID: 32913622 PMCID: PMC7443992 DOI: 10.1177/2040622320947378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 07/08/2020] [Indexed: 11/15/2022] Open
Abstract
Background and aims Alemtuzumab is a humanized monoclonal antibody that depletes CD52-bearing B and T lymphocytes. Clinical trials defined that systemic administration of alemtuzumab reduces disease severity in the relapsing-remitting phase of multiple sclerosis (MS). However, its efficacy in progressive MS patients is limited, which may reflect the inability of alemtuzumab to cross the reconstituted BBB in these patients. Objective: to study whether central nervous system (CNS) delivery of anti-CD52 antibodies reduces disease severity and the neuroinflammatory burden in the experimental autoimmune encephalomyelitis (EAE) model. Methods Anti-CD52 antibodies were administered intrathecally during the acute and chronic phases of EAE. Flow cytometry and immunohistochemistry were utilized to define immunological and pathological parameters. Results We show that subcutaneously administrated anti-CD52 antibodies completely abolish EAE disease severity. CNS delivery of anti-CD52 antibodies during both the acute and chronic phases of EAE moderately reduces disease severity and the neuroinflammatory burden. Our findings further suggest that CNS delivery of anti-CD52 antibodies impacts both the peripheral and CNS immune cell compartments in the EAE model but not in healthy mice. Conclusion Collectively, our findings highlight the therapeutic potential of CNS delivery of alemtuzumab for the treatment of progressive as well as early MS.
Collapse
Affiliation(s)
- Jeroen Fj Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Elien Grajchen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Elien Wouters
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Bieke Broux
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Piet Stinissen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Bart Van Wijmeersch
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jerome Ja Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Agoralaan Building C, Diepenbeek, 3590, Belgium
| |
Collapse
|
11
|
Repopulation of T, B, and NK cells following alemtuzumab treatment in relapsing-remitting multiple sclerosis. J Neuroinflammation 2020; 17:189. [PMID: 32539719 PMCID: PMC7296935 DOI: 10.1186/s12974-020-01847-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 05/19/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To characterize long-term repopulation of peripheral immune cells following alemtuzumab-induced lymphopenia in relapsing-remitting MS (RRMS), with a focus on regulatory cell types, and to explore associations with clinical outcome measures. METHODS The project was designed as a multicenter add-on longitudinal mechanistic study for RRMS patients enrolled in CARE-MS II, CARE-MS II extension at the University of Southern California and Stanford University, and an investigator-initiated study conducted at the Universities of British Columbia and Chicago. Methods involved collection of blood at baseline, prior to alemtuzumab administration, and at months 5, 11, 17, 23, 36, and 48 post-treatment. T cell, B cell, and natural killer (NK) cell subsets, chemokine receptor expression in T cells, in vitro cytokine secretion patterns, and regulatory T cell (Treg) function were assessed. Clinical outcomes, including expanded disability status score (EDSS), relapses, conventional magnetic resonance imaging (MRI) measures, and incidents of secondary autoimmunity were tracked. RESULTS Variable shifts in lymphocyte populations occurred over time in favor of CD4+ T cells, B cells, and NK cells with surface phenotypes characteristic of regulatory subsets, accompanied by reduced ratios of effector to regulatory cell types. Evidence of increased Treg competence was observed after each treatment course. CD4+ and CD8+ T cells that express CXCR3 and CCR5 and CD8+ T cells that express CDR3 and CCR4 were also enriched after treatment, indicating heightened trafficking potential in activated T cells. Patterns of repopulation were not associated with measures of clinical efficacy or secondary autoimmunity, but exploratory analyses using a random generalized estimating equation (GEE) Poisson model provide preliminary evidence of associations between pro-inflammatory cell types and increased risk for gadolinium (Gd+) enhancing lesions, while regulatory subsets were associated with reduced risk. In addition, the risk for T2 lesions correlated with increases in CD3+CD8+CXCR3+ cells. CONCLUSIONS Lymphocyte repopulation after alemtuzumab treatment favors regulatory subsets in the T cell, B cell, and NK cell compartments. Clinical efficacy may reflect the sum of interactions among them, leading to control of potentially pathogenic effector cell types. Several immune measures were identified as possible biomarkers of lesion activity. Future studies are necessary to more precisely define regulatory and effector subsets and their contributions to clinical efficacy and risk for secondary autoimmunity in alemtuzumab-treated patients, and to reveal new insights into mechanisms of immunopathogenesis in MS. TRIAL REGISTRATION Parent trials for this study are registered with ClinicalTrials.gov: CARE-MS II: NCT00548405, CARE-MS II extension: NCT00930553 and ISS: NCT01307332.
Collapse
|
12
|
Pitarokoili K, Bachir H, Sgodzai M, Grüter T, Haupeltshofer S, Duscha A, Pedreiturria X, Motte J, Gold R. Induction of Regulatory Properties in the Intestinal Immune System by Dimethyl Fumarate in Lewis Rat Experimental Autoimmune Neuritis. Front Immunol 2019; 10:2132. [PMID: 31552056 PMCID: PMC6746892 DOI: 10.3389/fimmu.2019.02132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 08/23/2019] [Indexed: 12/22/2022] Open
Abstract
Objective: Dimethyl fumarate (DMF) exerts immunomodulatory and neuroprotective effects in the animal model of experimental autoimmune neuritis (EAN) in the Lewis rat. DMF has been shown to modulate gut microbiota in veterinary medicine, however the effects of oral DMF on the gut-associated lymphoid tissue (GALT) remain unknown. Methods: Lewis rats were treated orally twice daily with DMF up to day 10 after immunization with immunogenic P2 peptide. Histological, flow cytometric and RT-PCR analyses of the GALT (intraepithelial layer, lamina propria, and Peyer patches) in duodenum, jejunum, and ileum were performed ex vivo. Moreover, cell transfer experiments were used to examine the protective effects of GALT regulatory T cells of the Peyer patches. Results: In the upper layers of duodenum, DMF induced a reduction of the toll-like receptor 4 (TLR4) mRNA expression. This was combined by a decrease of the pro-inflammatory lamina propria IFN-γ mRNA expression. In the ileum, we detected an immunoregulatory phenotype characterized by an increase of FoxP3 mRNA expression and of the nuclear factor (erythroid-derived-2)- like 2 (Nrf2) downstream molecule heme oxygenase-1 (HO-1) mRNA. Finally, CD4+ CD25+ regulatory T cells were increased in the Peyer patches. In vivo, the protective effect of these regulatory cells was verified by cell transfer into recipient EAN rats. Conclusions: Our results identified a novel immunomodulatory effect of DMF through the different regions and layers of the small intestine, which led to an increase of regulatory T cells, exerting a protective role in experimental neuritis.
Collapse
Affiliation(s)
- Kalliopi Pitarokoili
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Hussein Bachir
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Melissa Sgodzai
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Grüter
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Steffen Haupeltshofer
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Duscha
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Xiomara Pedreiturria
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jeremias Motte
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
13
|
On the immunoregulatory role of statins in multiple sclerosis: the effects on Th17 cells. Immunol Res 2019; 67:310-324. [DOI: 10.1007/s12026-019-09089-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
14
|
Mestre L, Carrillo-Salinas FJ, Mecha M, Feliú A, Espejo C, Álvarez-Cermeño JC, Villar LM, Guaza C. Manipulation of Gut Microbiota Influences Immune Responses, Axon Preservation, and Motor Disability in a Model of Progressive Multiple Sclerosis. Front Immunol 2019; 10:1374. [PMID: 31258540 PMCID: PMC6587398 DOI: 10.3389/fimmu.2019.01374] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/30/2019] [Indexed: 12/15/2022] Open
Abstract
Gut microbiota dysbiosis has been implicated in MS and other immune diseases, although it remains unclear how manipulating the gut microbiota may affect the disease course. Using a well-established model of progressive MS triggered by intracranial infection with Theiler's murine encephalomyelitis virus (TMEV), we sought to determine whether dysbiosis induced by oral antibiotics (ABX) administered on pre-symptomatic and symptomatic phases of the disease influences its course. We also addressed the effects of microbiota recolonization after ABX withdrawn in the presence or absence of probiotics. Central and peripheral immunity, plasma acetate and butyrate levels, axon damage and motor disability were evaluated. The cocktail of ABX prevented motor dysfunction and limited axon damage in mice, which had fewer CD4+ and CD8+ T cells in the CNS, while gut microbiota recolonization worsened motor function and axonal integrity. The underlying mechanisms of ABX protective effects seem to involve CD4+CD39+ T cells and CD5+CD1d+ B cells into the CNS. In addition, microglia adopted a round amoeboid morphology associated to an anti-inflammatory gene profile in the spinal cord of TMEV mice administered ABX. The immune changes in the spleen and mesenteric lymph nodes were modest, yet ABX treatment of mice limited IL-17 production ex vivo. Collectively, our results provide evidence of the functional relevance of gut microbiota manipulation on the neurodegenerative state and disease severity in a model of progressive MS and reinforce the role of gut microbiota as target for MS treatment.
Collapse
Affiliation(s)
- Leyre Mestre
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, Madrid, Spain.,Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| | | | - Miriam Mecha
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, Madrid, Spain.,Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| | - Ana Feliú
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, Madrid, Spain.,Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| | - Carmen Espejo
- Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain.,Servei de Neurología-Neuroimmunología, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain.,Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - José Carlos Álvarez-Cermeño
- Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain.,Immunology Department, Hospital Universitario Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Luisa María Villar
- Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain.,Immunology Department, Hospital Universitario Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carmen Guaza
- Neuroimmunology Group, Functional and Systems Neurobiology Department, Instituto Cajal, CSIC, Madrid, Spain.,Red Española de Esclerosis Múltiple (REEM), Barcelona, Spain
| |
Collapse
|
15
|
Álvarez-Sánchez N, Cruz-Chamorro I, Díaz-Sánchez M, Lardone PJ, Guerrero JM, Carrillo-Vico A. Peripheral CD39-expressing T regulatory cells are increased and associated with relapsing-remitting multiple sclerosis in relapsing patients. Sci Rep 2019; 9:2302. [PMID: 30783191 PMCID: PMC6381140 DOI: 10.1038/s41598-019-38897-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/09/2019] [Indexed: 12/02/2022] Open
Abstract
CD39, an ectonucleotidase that hydrolyses pro-inflammatory ATP, is a marker of highly active and suppressive T regulatory cells (Tregs). Although CD39 has a role in Treg suppression and might be important in the control of neuroinflammation in relapsing-remitting multiple sclerosis (RR-MS), to date, there are contradictory reports concerning the Tregs expression of CD39 in RR-MS patients. Thus, our objectives were to assess the activity and expression of CD39, especially in Tregs from peripheral blood mononuclear cells (PBMCs) of relapsing RR-MS patients compared with control subjects and to evaluate the association of CD39+ Tregs with disability and the odds of RR-MS. The activity and expression of CD39 and the CD39+ Treg frequency were measured in PBMCs from 55 relapsing RR-MS patients (19 untreated and 36 receiving immunomodulatory treatment) and 55 age- and sex-paired controls. Moreover, the association between CD39+ Tregs and RR-MS was assessed by multivariate logistic regression. CD39 activity and the frequency of CD39-expressing Tregs were elevated in relapsing RR-MS patients. Moreover, CD39+ Tregs were significantly correlated with the EDSS score and were independently associated with the odds of RR-MS. Our results highlight the relevance of CD39+ Treg subset in the clinical outcomes of RR-MS.
Collapse
Affiliation(s)
- Nuria Álvarez-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), Seville, Spain
| | - Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), Seville, Spain.,Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Seville, Spain
| | - María Díaz-Sánchez
- Unidad de Gestión Clínica de Neurociencias, Servicio de Neurología del Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Patricia Judith Lardone
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), Seville, Spain.,Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Seville, Spain
| | - Juan Miguel Guerrero
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), Seville, Spain.,Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Seville, Spain.,Department of Clinical Biochemistry, Virgen del Rocío University Hospital, Seville, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla, IBiS (Universidad de Sevilla, HUVR, Junta de Andalucía, CSIC), Seville, Spain. .,Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
16
|
Simon M, Ipek R, Homola GA, Rovituso DM, Schampel A, Kleinschnitz C, Kuerten S. Anti-CD52 antibody treatment depletes B cell aggregates in the central nervous system in a mouse model of multiple sclerosis. J Neuroinflammation 2018; 15:225. [PMID: 30098594 PMCID: PMC6086993 DOI: 10.1186/s12974-018-1263-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023] Open
Abstract
Background Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) for which several new treatment options were recently introduced. Among them is the monoclonal anti-CD52 antibody alemtuzumab that depletes mainly B cells and T cells in the immune periphery. Considering the ongoing controversy about the involvement of B cells and in particular the formation of B cell aggregates in the brains of progressive MS patients, an in-depth understanding of the effects of anti-CD52 antibody treatment on the B cell compartment in the CNS itself is desirable. Methods We used myelin basic protein (MBP)-proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) in C57BL/6 (B6) mice as B cell-dependent model of MS. Mice were treated intraperitoneally either at the peak of EAE or at 60 days after onset with 200 μg murine anti-CD52 vs. IgG2a isotype control antibody for five consecutive days. Disease was subsequently monitored for 10 days. The antigen-specific B cell/antibody response was measured by ELISPOT and ELISA. Effects on CNS infiltration and B cell aggregation were determined by immunohistochemistry. Neurodegeneration was evaluated by Luxol Fast Blue, SMI-32, and Olig2/APC staining as well as by electron microscopy and phosphorylated heavy neurofilament serum ELISA. Results Treatment with anti-CD52 antibody attenuated EAE only when administered at the peak of disease. While there was no effect on the production of MP4-specific IgG, the treatment almost completely depleted CNS infiltrates and B cell aggregates even when given as late as 60 days after onset. On the ultrastructural level, we observed significantly less axonal damage in the spinal cord and cerebellum in chronic EAE after anti-CD52 treatment. Conclusion Anti-CD52 treatment abrogated B cell infiltration and disrupted existing B cell aggregates in the CNS. Electronic supplementary material The online version of this article (10.1186/s12974-018-1263-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Micha Simon
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Rojda Ipek
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - György A Homola
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Würzburg, Würzburg, Germany
| | - Damiano M Rovituso
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Andrea Schampel
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.,Department of Neurology, University Hospital Essen, Essen, Germany
| | - Stefanie Kuerten
- Department of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany. .,Institute of Anatomy and Cell Biology, Friedrich Alexander University Erlangen-Nürnberg (FAU), Krankenhausstr. 9, 91054, Erlangen, Bavaria, Germany.
| |
Collapse
|
17
|
Massey JC, Sutton IJ, Ma DDF, Moore JJ. Regenerating Immunotolerance in Multiple Sclerosis with Autologous Hematopoietic Stem Cell Transplant. Front Immunol 2018; 9:410. [PMID: 29593711 PMCID: PMC5857574 DOI: 10.3389/fimmu.2018.00410] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/14/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disorder of the central nervous system where evidence implicates an aberrant adaptive immune response in the accrual of neurological disability. The inflammatory phase of the disease responds to immunomodulation to varying degrees of efficacy; however, no therapy has been proven to arrest progression of disability. Recently, more intensive therapies, including immunoablation with autologous hematopoietic stem cell transplantation (AHSCT), have been offered as a treatment option to retard inflammatory disease, prior to patients becoming irreversibly disabled. Empirical clinical observations support the notion that the immune reconstitution (IR) that occurs following AHSCT is associated with a sustained therapeutic benefit; however, neither the pathogenesis of MS nor the mechanism by which AHSCT results in a therapeutic benefit has been clearly delineated. Although the antigenic target of the aberrant immune response in MS is not defined, accumulated data suggest that IR following AHSCT results in an immunotolerant state through deletion of pathogenic clones by a combination of direct ablation and induction of a lymphopenic state driving replicative senescence and clonal attrition. Restoration of immunoregulation is evidenced by changes in regulatory T cell populations following AHSCT and normalization of genetic signatures of immune homeostasis. Furthermore, some evidence exists that AHSCT may induce a rebooting of thymic function and regeneration of a diversified naïve T cell repertoire equipped to appropriately modulate the immune system in response to future antigenic challenge. In this review, we discuss the immunological mechanisms of IR therapies, focusing on AHSCT, as a means of recalibrating the dysfunctional immune response observed in MS.
Collapse
Affiliation(s)
- Jennifer C Massey
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Neurology, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Ian J Sutton
- Neurology, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - David D F Ma
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - John J Moore
- Haematology and Bone Marrow Transplantation, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,Centre for Applied Medical Research, St Vincent's Hospital Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
18
|
Barbour M, Wood R, Hridi SU, Wilson C, McKay G, Bushell TJ, Jiang HR. The therapeutic effect of anti-CD52 treatment in murine experimental autoimmune encephalomyelitis is associated with altered IL-33 and ST2 expression levels. J Neuroimmunol 2018. [PMID: 29526407 DOI: 10.1016/j.jneuroim.2018.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) mice were administered with murine anti-CD52 antibody to investigate its therapeutic effect and whether the treatment modulates IL-33 and ST2 expression. EAE severity and central nervous system (CNS) inflammation were reduced following the treatment, which was accompanied by peripheral T and B lymphocyte depletion and reduced production of various cytokines including IL-33, while sST2 was increased. In spinal cords of EAE mice, while the number of IL-33+ cells remained unchanged, the extracellular level of IL-33 protein was significantly reduced in anti-CD52 antibody treated mice compared with controls. Furthermore the number of ST2+ cells in the spinal cord of treated EAE mice was downregulated due to decreased inflammation and immune cell infiltration in the CNS. These results suggest that treatment with anti-CD52 antibody differentially alters expression of IL-33 and ST2, both systemically and within the CNS, which may indicate IL-33/ST2 axis is involved in the action of the antibody in inhibiting EAE.
Collapse
Affiliation(s)
- Mark Barbour
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Rachel Wood
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Shehla U Hridi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Chelsey Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Grant McKay
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Trevor J Bushell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Hui-Rong Jiang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
19
|
Monoclonal Antibodies in Preclinical EAE Models of Multiple Sclerosis: A Systematic Review. Int J Mol Sci 2017; 18:ijms18091992. [PMID: 28926943 PMCID: PMC5618641 DOI: 10.3390/ijms18091992] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 02/08/2023] Open
Abstract
Monoclonal antibodies (mAb) are promising therapeutics in multiple sclerosis and multiple new candidates have been developed, hence increasing the need for some agreement for preclinical mAb studies. We systematically analyzed publications of experimental autoimmune encephalomyelitis (EAE) studies showing effects of monoclonal antibodies. A PubMed search retrieved 570 records, out of which 122 studies with 253 experiments were eligible based on experimental design, number of animals and presentation of time courses of EAE scores. Analysis of EAE models, treatment schedules, single and total doses, routes of administration, and onset of treatment from pre-immunization up to 35 days after immunization revealed high heterogeneity. Total doses ranged from 0.1 to 360 mg/kg for observation times of up to 35 days after immunization. About half of experiments (142/253) used total doses of 10-70 mg/kg. Employing this range, we tested anti-Itga4 as a reference mAb at varying schedules and got no, mild or substantial EAE-score reductions, depending on the mouse strain and onset of the treatment. The result agrees with the range of outcomes achieved in 10 reported anti-Itga4 experiments. Studies comparing low and high doses of various mAbs or early vs. late onset of treatment did not reveal dose-effect or timing-effect associations, with a tendency towards better outcomes with preventive treatments starting within the first week after immunization. The systematic comparison allows for extraction of some "common" design characteristics, which may be helpful to further assess the efficacy of mAbs and role of specific targets in preclinical models of multiple sclerosis.
Collapse
|
20
|
Zhao Y, Su H, Shen X, Du J, Zhang X, Zhao Y. The immunological function of CD52 and its targeting in organ transplantation. Inflamm Res 2017; 66:571-578. [PMID: 28283679 DOI: 10.1007/s00011-017-1032-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/18/2017] [Accepted: 02/22/2017] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION CD52 (Campath-1 antigen), a glycoprotein of 12 amino acids anchored to glycosylphosphatidylinositol, is widely expressed on the cell surface of immune cells, such as mature lymphocytes, natural killer cells (NK), eosinophils, neutrophils, monocytes/macrophages, and dendritic cells (DCs). The anti-CD52 mAb, alemtuzumab, was used widely in clinics for the treatment of patients such as organ transplantation. In the present manuscript, we will briefly summarize the immunological function of CD52 and discuss the application of anti-CD52 mAb in transplantation settings. FINDINGS We reviewed studies published until July 2016 to explore the role of CD52 in immune cell function and its implication in organ transplantation. We showed that ligation of cell surface CD52 molecules may offer costimulatory signals for T-cell activation and proliferation. However, soluble CD52 molecules will interact with the inhibitory sialic acid-binding immunoglobulin-like lectin 10 (Siglec10) to significantly inhibit T cell proliferation and activation. Although the physiological and pathological significances of CD52 molecules are still poorly understood, the anti-CD52 mAb, alemtuzumab, was used widely for the treatment of patients with chronic lymphocytic leukemia, autoimmune diseases as well as cell and organ transplantation in clinics. CONCLUSION Studies clearly showed that CD52 can modulate T-cell activation either by its intracellular signal pathways or by the interaction of soluble CD52 and Siglec-10 expressing on T cells. However, the regulatory functions of CD52 on other immune cell subpopulations in organ transplantation require to be studied in the near future.
Collapse
Affiliation(s)
- Yang Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huiting Su
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaofei Shen
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
- Department of General Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Junfeng Du
- Department of General Surgery, PLA Army General Hospital, Dongsishitiao Namencang 5, Dongcheng District, Beijing, 100007, China.
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, 8 Gong Ti Nan Road, Chaoyang District, Beijing, 100020, China.
| | - Yong Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|