151
|
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: 30] [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
|
152
|
Abstract
Genetically engineered T cell immunotherapies have provided remarkable clinical success to treat B cell acute lymphoblastic leukaemia by harnessing a patient's own T cells to kill cancer, and these approaches have the potential to provide therapeutic benefit for numerous other cancers, infectious diseases and autoimmunity. By introduction of either a transgenic T cell receptor or a chimeric antigen receptor, T cells can be programmed to target cancer cells. However, initial studies have made it clear that the field will need to implement more complex levels of genetic regulation of engineered T cells to ensure both safety and efficacy. Here, we review the principles by which our knowledge of genetics and genome engineering will drive the next generation of adoptive T cell therapies.
Collapse
|
153
|
Dziedzic A, Saluk-Bijak J, Miller E, Niemcewicz M, Bijak M. The Impact of SARS-CoV-2 Infection on the Development of Neurodegeneration in Multiple Sclerosis. Int J Mol Sci 2021; 22:1804. [PMID: 33670394 PMCID: PMC7918534 DOI: 10.3390/ijms22041804] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a global challenge. Currently, there is some information on the consequences of COVID-19 infection in multiple sclerosis (MS) patients, as it is a newly discovered coronavirus, but its far-reaching effects on participation in neurodegenerative diseases seem to be significant. Recent cases reports showed that SARS-CoV-2 may be responsible for initiating the demyelination process in people who previously had no symptoms associated with any nervous system disorders. It is presently known that infection of SARS-CoV-2 evokes cytokine storm syndrome, which may be one of the factors leading to the acute cerebrovascular disease. One of the substantial problems is the coexistence of cerebrovascular disease and MS in an individual's life span. Epidemiological studies showed an enhanced risk of death rate from vascular disabilities in MS patients of approximately 30%. It has been demonstrated that patients with severe SARS-CoV-2 infection usually show increased levels of D-dimer, fibrinogen, C-reactive protein (CRP), and overactivation of blood platelets, which are essential elements of prothrombotic events. In this review, the latest knowledge gathered during an ongoing pandemic of SARS-CoV-2 infection on the neurodegeneration processes in MS is discussed.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Marcin Niemcewicz
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.N.); (M.B.)
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.N.); (M.B.)
| |
Collapse
|
154
|
Choi GM, Lee B, Hong R, Park SY, Cho DE, Yeom M, Park HJ, Bae H, Hahm DH. Bee venom phospholipase A2 alleviates collagen-induced polyarthritis by inducing Foxp3 + regulatory T cell polarization in mice. Sci Rep 2021; 11:3511. [PMID: 33568685 PMCID: PMC7876016 DOI: 10.1038/s41598-021-82298-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/19/2021] [Indexed: 11/18/2022] Open
Abstract
The mechanism underlying bee venom (BV) therapy is still controversial, with opinions ranging from constituent-based pharmacological action to homeopathic-like activity. The purpose of this study was to examine whether BV phospholipase A2 (bvPLA2), an enzymatic component of BV, is a novel anti-inflammatory and anti-arthritic mediator capable of stimulating CD25+ Foxp3+ regulatory T cell (Treg) polarization in a mouse model of human rheumatoid arthritis (RA). An experimental model of RA was established in male DBA/1 mouse by 2-week-interval injections of 100 μg type II collagen emulsified in complete (first injection) or incomplete Freund's adjuvant (second injection) at the base of the tail. During arthritis development, bvPLA2 (0.1, 0.5, 1.0 mg/kg) and/or Treg inhibitors such as anti-CD25 antibodies and peptide 60 (P60) were injected intraperitoneally for 5 weeks. Arthritic symptoms and the expansion of Tregs were then assessed by behavioral assessments, histological and micro-CT imaging, and flow cytometry. bvPLA2 injections significantly alleviated arthritic behaviors such as squeaking and joint swelling, consistent with changes seen on both histological and micro-CT images. The anti-arthritic effects of bvPLA2 were blocked by intraperitoneal injections of 0.25 mg/kg anti-CD25 antibody and 10 μg/kg P60, as determined by behavioral assessments. Flow cytometric analysis of dendritic cells, B cells, and major T cell subsets from spleens revealed a significant depletion of Tregs following anti-CD25 antibody, but not P60, treatment. bvPLA2 treatment exerted significant anti-inflammatory and anti-arthritic activities in a mouse model of RA via the induction of Tregs.
Collapse
Affiliation(s)
- Gwang-Muk Choi
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Bombi Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Riwon Hong
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seon-Young Park
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Da-Eun Cho
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Mijung Yeom
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyunsu Bae
- Department of Korean Medical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Dae-Hyun Hahm
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
- BioNanocomposite Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
155
|
Prado DS, Damasceno LEA, Sonego AB, Rosa MH, Martins TV, Fonseca MDM, Cunha TM, Cunha FQ, Alves-Filho JC. Pitavastatin ameliorates autoimmune neuroinflammation by regulating the Treg/Th17 cell balance through inhibition of mevalonate metabolism. Int Immunopharmacol 2021; 91:107278. [PMID: 33341737 DOI: 10.1016/j.intimp.2020.107278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 12/16/2022]
Abstract
While Treg cells are responsible for self-tolerance and immune homeostasis, pathogenic autoreactive Th17 cells produce pro-inflammatory cytokines that lead to tissue damage associated with autoimmunity, as observed in multiple sclerosis. Therefore, the immunological balance between Th17 and Treg cells may represent a promising option for immune therapy. Statin drugs are used to treat dyslipidemia; however, besides their effects on preventing cardiovascular diseases, statins also have anti-inflammatory effects. Here, we investigated the role of pitavastatin on experimental autoimmune encephalomyelitis (EAE) and the differentiation of Treg and Th17 cells. EAE was induced by immunizing C57BL/6 mice with MOG35-55. EAE severity was determined by analyzing the clinical score and inflammatory parameters in the spinal cord. Naive CD4 T cells were cultured under Treg and Th17-skewing conditions in vitro in the presence of pitavastatin. We found that pitavastatin decreased EAE development, which was accompanied by a reduction of all parameters investigated. Pitavastatin also reduced the expression of IBA1 and pSTAT3 (Y705 and S727) in the spinal cords of EAE mice. Interestingly, the reduction of Th17 cell frequency in the draining lymph nodes of EAE mice treated with pitavastatin was followed by an increase of Treg cells. Indeed, pitavastatin directly affects T cell differentiation in vitro by decreasing Th17 and increasing Treg cell differentiation. Mechanistically, pitavastatin effects are dependent on mevalonate synthesis. Thus, our data show the potential anti-inflammatory effect of pitavastatin on the pathogenesis of the experimental neuroinflammation by modulating the Th17/Treg axis.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Cell Differentiation/drug effects
- Cells, Cultured
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Inflammation Mediators/metabolism
- Lymph Nodes/drug effects
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Male
- Mevalonic Acid/metabolism
- Mice, Inbred C57BL
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments
- Quinolines/pharmacology
- Spinal Cord/drug effects
- Spinal Cord/immunology
- Spinal Cord/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Mice
Collapse
Affiliation(s)
- D S Prado
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - L E A Damasceno
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - A B Sonego
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - M H Rosa
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - T V Martins
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - M D M Fonseca
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - T M Cunha
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - F Q Cunha
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - J C Alves-Filho
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
156
|
Ramaglia V, Rojas O, Naouar I, Gommerman JL. The Ins and Outs of Central Nervous System Inflammation-Lessons Learned from Multiple Sclerosis. Annu Rev Immunol 2021; 39:199-226. [PMID: 33524273 DOI: 10.1146/annurev-immunol-093019-124155] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by the inappropriate invasion of lymphocytes and monocytes into the central nervous system (CNS), where they orchestrate the demyelination of axons, leading to physical and cognitive disability. There are many reasons immunologists should be interested in MS. Aside from the fact that there is still significant unmet need for patients living with the progressive form of the disease, MS is a case study for how immune cells cross CNS barriers and subsequently interact with specialized tissue parenchymal cells. In this review, we describe the types of immune cells that infiltrate the CNS and then describe interactions between immune cells and glial cells in different types of lesions. Lastly, we provide evidence for CNS-compartmentalized immune cells and speculate on how this impacts disease progression for MS patients.
Collapse
Affiliation(s)
- Valeria Ramaglia
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Olga Rojas
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | | |
Collapse
|
157
|
Correale J. Immunosuppressive Amino-Acid Catabolizing Enzymes in Multiple Sclerosis. Front Immunol 2021; 11:600428. [PMID: 33552055 PMCID: PMC7855700 DOI: 10.3389/fimmu.2020.600428] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/30/2020] [Indexed: 01/01/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease that affects the central nervous system. Although the pathogenesis of MS is not yet fully elucidated, several evidences suggest that autoimmune processes mediated by Th1, Th17, and B cells play an important role in the development of the disease. Similar to other cells, immune cells need continuous access to amino acids (AA) in order to maintain basal metabolism and maintain vitality. When immune cells are activated by inflammation or antigenic signals, their demand for AA increases rapidly. Although AA deprivation itself may weaken the immune response under certain conditions, cells also have AA sensitive pathways that can activate intense alterations in cell metabolism based on changes in AA levels. Several data indicate that cells expressing enzymes that can degrade AA can regulate the functions of antigen-presenting cells and lymphocytes, revealing that the AA pathways are essential for controlling the function, and survival of immune cells, as well as immune cell gene expression. Basal AA catabolism may contribute to immune homeostasis and prevent autoimmunity, while increased AA catalytic activity may enhance immune suppression. In addition, there is increasing evidence that some downstream AA metabolites are important biological mediators of autoimmune response regulation. Two of the most important AA that modulate the immune response are L-Tryptophan (Trp) and L-Arginine (Arg). Tryptophan is catabolized through 2,3-dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO) 1 and IDO2 enzymes, while three other enzymes catabolize Arg: inducible nitric oxide synthetase (iNOS), and two arginase isoforms (ARG1, ARG2). Genes encoding IDO, iNOS and ARG are induced by inflammatory cues such as cytokines, a key feature that distinguishes them from enzymes that catabolize other AA. Evidence suggests that AA catabolism is decreased in MS patients and that this decrease has functional consequences, increasing pro-inflammatory cytokines and decreasing Treg cell numbers. These effects are mediated by at least two distinct pathways involving serine/threonine kinases: the general control nonderepressible 2 kinase (GCN2K) pathway; and the mammalian target of rapamycin (mTOR) pathway. Similarly, IDO1-deficient mice showed exacerbation of experimental autoimmune encephalomyelitis (EAE), increased Th1 and Th17 cells, and decreased Treg cells. On the contrary, the administration of downstream Trp metabolite 3-HAA, inhibits Th1/Th17 effector cells and promotes Treg response by up-regulating TGF-β production by dendritic cells, thereby improving EAE. Collectively, these observations stand out the significance of AA catabolism in the regulation of the immune responses in MS patients. The molecules related to these pathways deserve further exploration as potential new therapeutic targets in MS.
Collapse
|
158
|
Morgan BP, Gommerman JL, Ramaglia V. An "Outside-In" and "Inside-Out" Consideration of Complement in the Multiple Sclerosis Brain: Lessons From Development and Neurodegenerative Diseases. Front Cell Neurosci 2021; 14:600656. [PMID: 33488361 PMCID: PMC7817777 DOI: 10.3389/fncel.2020.600656] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The last 15 years have seen an explosion of new findings on the role of complement, a major arm of the immune system, in the central nervous system (CNS) compartment including contributions to cell migration, elimination of synapse during development, aberrant synapse pruning in neurologic disorders, damage to nerve cells in autoimmune diseases, and traumatic injury. Activation of the complement system in multiple sclerosis (MS) is typically thought to occur as part of a primary (auto)immune response from the periphery (the outside) against CNS antigens (the inside). However, evidence of local complement production from CNS-resident cells, intracellular complement functions, and the more recently discovered role of early complement components in shaping synaptic circuits in the absence of inflammation opens up the possibility that complement-related sequelae may start and finish within the brain itself. In this review, the complement system will be introduced, followed by evidence that implicates complement in shaping the developing, adult, and normal aging CNS as well as its contribution to pathology in neurodegenerative conditions. Discussion of data supporting "outside-in" vs. "inside-out" roles of complement in MS will be presented, concluded by thoughts on potential approaches to therapies targeting specific elements of the complement system.
Collapse
Affiliation(s)
- B. Paul Morgan
- UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| | | | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
159
|
Cui C, Longinetti E, Larsson H, Andersson J, Pawitan Y, Piehl F, Fang F. Associations between autoimmune diseases and amyotrophic lateral sclerosis: a register-based study. Amyotroph Lateral Scler Frontotemporal Degener 2020; 22:211-219. [PMID: 33331190 DOI: 10.1080/21678421.2020.1861022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Objective: To assess the associations of 43 autoimmune diseases with the subsequent risk of ALS and further evaluate the contribution of familial confounding to these associations.Methods: We conducted a nationwide register-based nested case-control study including 3561 ALS patients diagnosed during 1990-2013 in Sweden and 35,610 controls that were randomly selected from the general population and individually matched to the cases on age, sex, and county of birth. To evaluate the contribution of familial factors on the studied association, we additionally studied the first-degree relatives (siblings and children) of ALS patients and their controls.Results: Patients with ALS had a 47% higher risk of being previously diagnosed with autoimmune disease (OR 1.47, 95% confidence interval [CI] 1.31-1.64), compared with controls. A positive association was noted for several autoimmune diseases, including myasthenia gravis, polymyositis or dermatomyositis, Guillain-Barre syndrome, type 1 diabetes diagnosed younger than 30 years, multiple sclerosis, and hypothyreosis. The increased risk of any autoimmune disease was greatest during the year before ALS diagnosis, likely due to misdiagnosis. A statistically significantly increased risk was also noted during 2-5 years, but not earlier, before ALS diagnosis. First-degree relatives of ALS patients had however no increased risk of autoimmune diseases compared with first-degree relatives of controls.Conclusions: Although it is difficult to completely remove the potential effects of misdiagnosis, there is likely a positive association between autoimmune disease (such as type 1 diabetes and multiple sclerosis) and ALS, which is not fully explained by shared familial confounding factors.
Collapse
Affiliation(s)
- Can Cui
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elisa Longinetti
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden, and.,School of Medical Sciences, Örebro University, Örebro, Sweden
| | - John Andersson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden, and
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
160
|
Rodero M, Cuéllar C. Modulation by Anisakis simplex antigen of inflammatory response generated in experimental autoimmune encephalomyelitis. Int Immunopharmacol 2020; 90:107241. [PMID: 33321294 DOI: 10.1016/j.intimp.2020.107241] [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: 11/04/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022]
Abstract
The impact of immunization with Anisakis simplex larval antigen on the occurrence and progression of experimental autoimmune encephalomyelitis (EAE) induced in mice was studied. C57BL/6J mice were immunized with the MOG35-55 peptide and one batch was treated with A. simplex total larval antigen on days 1, 8, 10 and 12 after EAE induction. Significantly higher values were obtained in the EAE clinical parameters of the antigen-treated group. Likewise, there was a significant decrease in the weights of the animals. Anisakis-treatment produced a significant decrease in anti-MOG35-55 specific IgG1 on day 21. On day 14 there was an increase in serum IL-2, IL-6, IL-10, IL-17A, and TGF-β in the treated group. On day 21, a decrease in IL-4, IL-6, TNF-α, TGF-β was observed. All brain determinations were made on day 21. The treatment decreased values of IL-6, IL-10, IL-17A and TNF-α. A. simplex antigen caused a significantly higher incidence of EAE and an advance in the appearance of the disease manifestations. However, treatment with the antigen was able to cause a decrease in proinflammatory cytokines (IL-6, IL-17A, and TNF-α) in nervous tissue that could establish a future preventive scenario for myelin damage.
Collapse
Affiliation(s)
- Marta Rodero
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Carmen Cuéllar
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain.
| |
Collapse
|
161
|
Tapia-Maltos MA, Treviño-Frenk I, García-González HB, Rosetti M, Barriga-Maldonado V, Morales-Ramírez F, López-Hernández DC, Rosetti F, Crispín JC. Identification of regulatory T cell molecules associated with severity of multiple sclerosis. Mult Scler 2020; 27:1695-1705. [PMID: 33300840 DOI: 10.1177/1352458520977045] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Regulatory CD4+ T cells (Tregs) exhibit functional alterations in patients with multiple sclerosis (MS). Transforming growth factor (TGF)-β is a key regulator of Treg development and function. OBJECTIVE The objective of this study is to determine whether the expression of functionally relevant TGF-β-regulated molecules is altered in Tregs from patients with MS. METHODS Expression of nine Treg markers was analyzed by multi-color flow cytometry in CD4+ T cells and Treg subpopulations of 31 untreated MS patients and age- and sex-matched healthy donors (HDs). Correlations between Treg marker expression and clinical variables were sought. RESULTS Expression of the transcription factor Helios, which defines thymic-derived Tregs, was decreased in this Treg subpopulation. The frequency of peripherally generated Tregs was increased in patients with MS, particularly in patients with progressive MS. Low frequencies of thymic-derived Tregs were associated with magnetic resonance imaging (MRI) lesion-burden and a high relapse rate. Four surface markers associated with TGF-β signaling (ABCA1, BTLA, DNAM-1, and GARP) were differentially expressed on Tregs from patients with MS and HDs. Expression levels of CD73, CD103, ABCA1, and PAR2 showed strong correlations with disease severity. CONCLUSION We have identified novel markers abnormally expressed on Tregs from patients with MS that could detect patients with severe disease.
Collapse
Affiliation(s)
- M A Tapia-Maltos
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico/Plan de Estudios Combinados en Medicina (PECEM), Facultad de Medicina, UNAM, Mexico City, Mexico
| | - I Treviño-Frenk
- Departamento de Neurología y Psiquiatría, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - H B García-González
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - M Rosetti
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - V Barriga-Maldonado
- Departamento de Neurología y Psiquiatría, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - F Morales-Ramírez
- Departamento de Neurología y Psiquiatría, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - D C López-Hernández
- Departamento de Neurología y Psiquiatría, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - F Rosetti
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - J C Crispín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico/Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Monterrey, Mexico
| |
Collapse
|
162
|
Killick J, Hay J, Morandi E, Vermeren S, Kari S, Angles T, Williams A, Damoiseaux J, Astier AL. Vitamin D/CD46 Crosstalk in Human T Cells in Multiple Sclerosis. Front Immunol 2020; 11:598727. [PMID: 33329593 PMCID: PMC7732696 DOI: 10.3389/fimmu.2020.598727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/27/2020] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS), in which T-cell migration into the CNS is key for pathogenesis. Patients with MS exhibit impaired regulatory T cell populations, and both Foxp3+ Tregs and type I regulatory T cells (Tr1) are dysfunctional. MS is a multifactorial disease and vitamin D deficiency is associated with disease. Herein, we examined the impact of 1,25(OH)2D3 on CD4+ T cells coactivated by either CD28 to induce polyclonal activation or by the complement regulator CD46 to promote Tr1 differentiation. Addition of 1,25(OH)2D3 led to a differential expression of adhesion molecules on CD28- and CD46-costimulated T cells isolated from both healthy donors or from patients with MS. 1,25(OH)2D3 favored Tr1 motility though a Vitamin D-CD46 crosstalk highlighted by increased VDR expression as well as increased CYP24A1 and miR-9 in CD46-costimulated T cells. Furthermore, analysis of CD46 expression on T cells from a cohort of patients with MS supplemented by vitamin D showed a negative correlation with the levels of circulating vitamin D. Moreover, t-Distributed Stochastic Neighbor Embedding (t-SNE) analysis allowed the visualization and identification of clusters increased by vitamin D supplementation, but not by placebo, that exhibited similar adhesion phenotype to what was observed in vitro. Overall, our data show a crosstalk between vitamin D and CD46 that allows a preferential effect of Vitamin D on Tr1 cells, providing novel key insights into the role of an important modifiable environmental factor in MS.
Collapse
Affiliation(s)
- Justin Killick
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Joanne Hay
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Elena Morandi
- Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - Sonja Vermeren
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Saniya Kari
- Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - Thibault Angles
- Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - Anna Williams
- Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, Netherlands
| | - Anne L Astier
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| |
Collapse
|
163
|
Yang S, Tan L, Chen Y, Liu A, Hong M, Peng Z. DIM mitigates the development of experimental autoimmune encephalomyelitis by maintaining the stability and suppressive function of regulatory T cells. Cell Immunol 2020; 358:104238. [PMID: 33137648 DOI: 10.1016/j.cellimm.2020.104238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/28/2020] [Accepted: 10/08/2020] [Indexed: 12/21/2022]
Abstract
Recent studies have revealed that indoles, dietary ligands of the aryl hydrocarbon receptor (AhR), have immunomodulatory characteristics of balancing the differentiation of regulatory T cells (Tregs) and Th17 cells in multiple autoimmune diseases. In this study, we aimed to investigate the potency of the indole, 3,3'-diindolylmethane (DIM), on the stability and suppressive function of Tregs in experimental autoimmune encephalomyelitis (EAE). Furthermore, we used the AhR antagonist CH223191 to verify that DIM exerts its effects on Tregs through the activation of AhR. We found that DIM treatment significantly alleviated the severity of EAE by maintaining the stability and suppressive function of Tregs instead of facilitating the differentiation of Tregs. Thus, these DIM-treated Tregs might indirectly inhibit the generation of Th17 cells and the production of proinflammatory cytokines. And we confirmed the critical role of AhR in the EAE model. Our study further investigated the mechanisms by which dietary indoles promote Treg activity in the EAE model. DIM may act as a novel therapeutic to restrain autoimmune inflammation in multiple sclerosis.
Collapse
Affiliation(s)
- Sujuan Yang
- Department of Neurology, First Affiliated Hospital of Guangdong Pharmaceutical College, Guangzhou 510000, China
| | - Lixi Tan
- Department of Neurology, First Affiliated Hospital of Guangdong Pharmaceutical College, Guangzhou 510000, China
| | - Yingying Chen
- Department of Neurology, The People's Hospital of Linying County, Luohe 462600, China
| | - Aiqun Liu
- Department of Neurology, First Affiliated Hospital of Guangdong Pharmaceutical College, Guangzhou 510000, China
| | - Mingfan Hong
- Department of Neurology, First Affiliated Hospital of Guangdong Pharmaceutical College, Guangzhou 510000, China
| | - Zhongxing Peng
- Department of Neurology, First Affiliated Hospital of Guangdong Pharmaceutical College, Guangzhou 510000, China.
| |
Collapse
|
164
|
CCR6 blockade on regulatory T cells ameliorates experimental model of multiple sclerosis. Cent Eur J Immunol 2020; 45:256-266. [PMID: 33437177 PMCID: PMC7790011 DOI: 10.5114/ceji.2020.101241] [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: 11/21/2018] [Accepted: 04/30/2019] [Indexed: 01/28/2023] Open
Abstract
Regulatory T cells (Tregs) play a significant role in limiting damage of tissue affected by autoimmune process, which has been demonstrated in various experimental models for multiple sclerosis (MS) (mostly experimental autoimmune encephalomyelitis – EAE), rheumatoid arthritis, and type 1 diabetes. In this study, we demonstrated that Tregs increasingly migrate to central nervous system (CNS) during subsequent phases of EAE (preclinical, initial attack, and remission). In contrast, in peripheral tissues (blood, lymph nodes, and spleen), a significant accumulation of Tregs is mostly present during EAE remission. Moreover, an increased expression of CCR6 on Tregs in the CNS, blood, lymph nodes, and spleen in all phases of EAE was observed. The highest expression of CCR6 on Tregs from the CNS, lymph nodes, and spleen was noted during the initial attack of EAE, whereas in the blood, the peak expression of CCR6 was detected during the preclinical phase. The presence of Tregs in the CNS during EAE was confirmed by immunohistochemistry. To analyze additional functional significance of CCR6 expression on Tregs for EAE pathology, we modulated the clinical course of this MS model using Tregs with blocked CCR6. EAE mice, which received CCR6-deficient Tregs showed significant amelioration of disease severity. This observation suggests that CCR6 on Tregs may be a potential target for future therapeutic interventions in MS.
Collapse
|
165
|
Primaquine elicits Foxp3+ regulatory T cells with a superior ability to limit CNS autoimmune inflammation. J Autoimmun 2020; 114:102505. [DOI: 10.1016/j.jaut.2020.102505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022]
|
166
|
[Genetically modified regulatory T cells: therapeutic concepts and regulatory aspects]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2020; 63:1403-1411. [PMID: 33067665 PMCID: PMC7647973 DOI: 10.1007/s00103-020-03230-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/22/2020] [Indexed: 11/24/2022]
Abstract
Adoptive T‑Zelltherapien sind neuartige Konzepte zur Behandlung verschiedener Krankheiten. CAR-T-Zellen sind dabei als Letztlinientherapie für fortgeschrittene B‑Zelllymphome und die B‑Zellleukämie etabliert und zugelassen. TCR-basierte T‑Zellen als Behandlungsoption verschiedener hämatologischer und solider Tumoren befinden sich in der klinischen Entwicklung. Genetisch modifizierte regulatorische T‑Zellen stehen dagegen noch am Anfang ihrer klinischen Entwicklung zur Induktion von Immuntoleranz in einer Vielzahl von Anwendungsgebieten. In diesem Artikel wird zunächst ein Überblick über die Funktion der regulatorischen T‑Zellen für die Induktion der Immuntoleranz sowie über ihre Rolle im Pathomechanismus bestimmter Immunerkrankungen gegeben und der aktuelle Stand der klinischen Entwicklungen von therapeutischen Ansätzen auf Basis genetisch modifizierter regulatorischer T‑Zellen zusammengefasst. Im Weiteren werden die regulatorisch-wissenschaftlichen Anforderungen und Herausforderungen hinsichtlich Herstellung und Qualitätskontrolle sowie nichtklinischer und klinischer Testung genetisch modifizierter regulatorischer T‑Zellen als Arzneimittel für neuartige Therapien diskutiert.
Collapse
|
167
|
Sotiropoulos MG, Chitnis T. Opposing and potentially antagonistic effects of BMP and TGF-β in multiple sclerosis: The "Yin and Yang" of neuro-immune Signaling. J Neuroimmunol 2020; 347:577358. [PMID: 32795734 DOI: 10.1016/j.jneuroim.2020.577358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Bone Morphogenetic Proteins (BMP) and Transforming Growth Factor-beta (TGF-β) are cytokines with similar receptors and messengers. They are important for immune cell function, with BMPs exerting mainly proinflammatory but also anti-inflammatory effects, and TGF-β suppressing inflammation. Patients with Multiple Sclerosis exhibit BMP overactivity and suppressed TGF-β signaling. This dysregulated signaling participates in the crosstalk between infiltrating immune cells and glia, where BMP inhibits remyelination. Reciprocal antagonism between the two pathways takes place via a variety of mechanisms. Although this antagonism has not been studied in the setting of Multiple Sclerosis, it could inform further research and treatment discovery.
Collapse
Affiliation(s)
- Marinos G Sotiropoulos
- Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA.
| | - Tanuja Chitnis
- Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA.
| |
Collapse
|
168
|
Janssens I, Cools N. Regulating the regulators: Is introduction of an antigen-specific approach in regulatory T cells the next step to treat autoimmunity? Cell Immunol 2020; 358:104236. [PMID: 33137651 DOI: 10.1016/j.cellimm.2020.104236] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/22/2022]
Abstract
In autoimmunity, the important and fragile balance between immunity and tolerance is disturbed, resulting in abnormal immune responses to the body's own tissues and cells. CD4+CD25hiFoxP3+ regulatory T cells (Tregs) induce peripheral tolerance in vivo by means of direct cell-cell contact and release of soluble factors, or indirectly through antigen-presenting cells (APC), thereby controlling auto-reactive effector T cells. Based on these unique capacities of Tregs, preclinical studies delivered proof-of-principle for the clinical use of Tregs for the treatment of autoimmune diseases. To date, the first clinical trials using ex vivo expanded polyclonal Tregs have been completed. These pioneering studies demonstrate the feasibility of generating large numbers of polyclonal Tregs in a good manufacturing practices (GMP)-compliant manner, and that infusion of Tregs is well tolerated by patients with no evidence of general immunosuppression. Nonetheless, only modest clinical results were observed, arguing that a more antigen-specific approach might be needed to foster a durable patient-specific clinical cell therapy without the risk for general immunosuppression. In this review, we discuss current knowledge, applications and future goals of adoptive immune-modulatory Treg therapy for the treatment of autoimmune disease and transplant rejection. We describe the key advances and prospects of the potential use of T cell receptor (TCR)- and chimeric antigen receptor (CAR)-engineered Tregs in future clinical applications. These approaches could deliver the long-awaited breakthrough in stopping undesired autoimmune responses and transplant rejections.
Collapse
Affiliation(s)
- Ibo Janssens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
169
|
Nakatsuka N, Patterson N, Patsopoulos NA, Altemose N, Tandon A, Beecham AH, McCauley JL, Isobe N, Hauser S, De Jager PL, Hafler DA, Oksenberg JR, Reich D. Two genetic variants explain the association of European ancestry with multiple sclerosis risk in African-Americans. Sci Rep 2020; 10:16902. [PMID: 33037294 PMCID: PMC7547691 DOI: 10.1038/s41598-020-74035-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/24/2020] [Indexed: 12/31/2022] Open
Abstract
Epidemiological studies have suggested differences in the rate of multiple sclerosis (MS) in individuals of European ancestry compared to African ancestry, motivating genetic scans to identify variants that could contribute to such patterns. In a whole-genome scan in 899 African-American cases and 1155 African-American controls, we confirm that African-Americans who inherit segments of the genome of European ancestry at a chromosome 1 locus are at increased risk for MS [logarithm of odds (LOD) = 9.8], although the signal weakens when adding an additional 406 cases, reflecting heterogeneity in the two sets of cases [logarithm of odds (LOD) = 2.7]. The association in the 899 individuals can be fully explained by two variants previously associated with MS in European ancestry individuals. These variants tag a MS susceptibility haplotype associated with decreased CD58 gene expression (odds ratio of 1.37; frequency of 84% in Europeans and 22% in West Africans for the tagging variant) as well as another haplotype near the FCRL3 gene (odds ratio of 1.07; frequency of 49% in Europeans and 8% in West Africans). Controlling for all other genetic and environmental factors, the two variants predict a 1.44-fold higher rate of MS in European-Americans compared to African-Americans.
Collapse
Affiliation(s)
- Nathan Nakatsuka
- Department of Genetics, Harvard Medical School, New Research Building, Boston, MA, 02115, USA. .,Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Nick Patterson
- Department of Human Evolutionary Biology, Harvard University, 16 Divinity Ave., Cambridge, MA, 02138, USA.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02141, USA
| | - Nikolaos A Patsopoulos
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02141, USA.,Systems Biology and Computer Science Program, Department of Neurology, Ann Romney Center for Neurological Diseases, Brigham & Women's Hospital, Boston, MA, 02115, USA.,Division of Genetics, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nicolas Altemose
- Department of Bioengineering, University of California Berkeley, San Francisco, Berkeley, CA, 94720, USA
| | - Arti Tandon
- Department of Genetics, Harvard Medical School, New Research Building, Boston, MA, 02115, USA.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02141, USA
| | - Ashley H Beecham
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Jacob L McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Noriko Isobe
- Department of Neurology, University of California San Francisco School of Medicine, San Francisco, CA, 94158, USA.,Department of Neurology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka, 812-8582, Japan
| | - Stephen Hauser
- Department of Neurology, University of California San Francisco School of Medicine, San Francisco, CA, 94158, USA
| | - Philip L De Jager
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02141, USA.,Department of Neurology, Center for Translational & Computational Neuroimmunology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - David A Hafler
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02141, USA.,Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Jorge R Oksenberg
- Department of Neurology, University of California San Francisco School of Medicine, San Francisco, CA, 94158, USA
| | - David Reich
- Department of Genetics, Harvard Medical School, New Research Building, Boston, MA, 02115, USA. .,Department of Human Evolutionary Biology, Harvard University, 16 Divinity Ave., Cambridge, MA, 02138, USA. .,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, 02141, USA. .,Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
170
|
Alissafi T, Kalafati L, Lazari M, Filia A, Kloukina I, Manifava M, Lim JH, Alexaki VI, Ktistakis NT, Doskas T, Garinis GA, Chavakis T, Boumpas DT, Verginis P. Mitochondrial Oxidative Damage Underlies Regulatory T Cell Defects in Autoimmunity. Cell Metab 2020; 32:591-604.e7. [PMID: 32738205 PMCID: PMC7611060 DOI: 10.1016/j.cmet.2020.07.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 05/14/2020] [Accepted: 07/07/2020] [Indexed: 11/22/2022]
Abstract
Regulatory T cells (Tregs) are vital for the maintenance of immune homeostasis, while their dysfunction constitutes a cardinal feature of autoimmunity. Under steady-state conditions, mitochondrial metabolism is critical for Treg function; however, the metabolic adaptations of Tregs during autoimmunity are ill-defined. Herein, we report that elevated mitochondrial oxidative stress and a robust DNA damage response (DDR) associated with cell death occur in Tregs in individuals with autoimmunity. In an experimental autoimmune encephalitis (EAE) mouse model of autoimmunity, we found a Treg dysfunction recapitulating the features of autoimmune Tregs with a prominent mtROS signature. Scavenging of mtROS in Tregs of EAE mice reversed the DDR and prevented Treg death, while attenuating the Th1 and Th17 autoimmune responses. These findings highlight an unrecognized role of mitochondrial oxidative stress in defining Treg fate during autoimmunity, which may facilitate the design of novel immunotherapies for diseases with disturbed immune tolerance.
Collapse
Affiliation(s)
- Themis Alissafi
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece.
| | - Lydia Kalafati
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece; Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany, and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Maria Lazari
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Anastasia Filia
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Ismini Kloukina
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Maria Manifava
- Babraham Institute, Signaling Programme, Cambridge CB22 3AT, UK
| | - Jong-Hyung Lim
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany
| | | | | | - George A Garinis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas, Crete, Heraklion 70013, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Dimitrios T Boumpas
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece; Joint Rheumatology Program, 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens 12462, Greece
| | - Panayotis Verginis
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece; Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden 01307, Germany.
| |
Collapse
|
171
|
Sadeghmousavi S, Rezaei N. COVID-19 and Multiple Sclerosis: Predisposition and Precautions in Treatment. ACTA ACUST UNITED AC 2020; 2:1802-1807. [PMID: 32895640 PMCID: PMC7467844 DOI: 10.1007/s42399-020-00504-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), leading to Coronavirus disease 2019 (COVID-19), is not always confined to the respiratory tract, while patients with can develop neurological manifestations. The patients with multiple sclerosis (MS) pose challenges in this pandemic situation, because of the immunosuppressive medications they get and the fact that viral infections may contribute to MS exacerbation and relapses as an environmental factor in genetically predisposed individuals. Herein, possible consequences of COVID-19 which may carry for the MS patients and the underlying mechanisms of its impact are discussed.
Collapse
Affiliation(s)
- Shaghayegh Sadeghmousavi
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran, 14194 Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
172
|
Eggers C, Kopatz V, Dörr W, Lassmann H, Geinitz H. Localised multiple sclerosis-like disease after irradiation of the cervicothoracic spinal cord. Mult Scler Relat Disord 2020; 45:102443. [PMID: 32827971 DOI: 10.1016/j.msard.2020.102443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Radiation has been discussed as a potential causative factor for Multiple Sclerosis (MS). However, it is unresolved whether radiation increases the aggressiveness of the immune system or whether it alters the nervous tissue to become vulnerable to a pre-existing autoimmune attackdisposition. We report a patient with an MS-like disease confined to the irradiated part of his central nervous system (CNS). CASE REPORT within the course of a year, a 29 year-old man developed three relapses that were distinguishable regarding their neurological signs and symptoms. Clinically and on MRI, all relapses were localised to the cervico-thoracic spinal cord (sensory level Th6, monoparesis right leg, sign of Lhermitte). Four years before, he had been diagnosed with supradiaphragmatic Hodgkin´s lymphoma stage IIa. Four courses of chemotherapy with the ABVD-protocol and irradiation with 29,5 Gray led to complete tumour remission. Consecutive MR-imaging of the brain and spinal cord revealed fluctuating and partially contrast-enhancing lesions exclusively in those sections of the spinal cord that were localised in the field irradiated four years before. Treatment with pulsed i.v. steroids led to improvement. CSF analysis showed mild pleocytosis and isolated oligoclonal bands. Extensive work-up for differential diagnoses was negative. Genetic sequencing for DNA repair enzymes and in-vitro assays of the patients peripheral blood mononuclear cells for increased sensitivity to irradiation was unrevealing. CONCLUSION The fact that this patients MS-like disease was strictly confined to the irradiated parts of the body suggests that the co-occurrence of Hodgkins and MS-like disease was not simple coincidence but that they are pathogenetically linked. An increased aggressiveness of the immune system caused by the radiation is an unlikely explanation as the autoimmune attack would not be expected to spare the non-radiated parts of the CNS. We propose that in our patient the nervous tissue in the radiation clinical target volume was altered by radiation. This alteration of antigenic make-up, in turn, may have enabled an MS-specific autoimmune attack by a pre-existent immunological mechanism. This hypothesis is supported by experimental studies of induction of experimental autoimmune encephalitis (EAE) in irradiated rats.
Collapse
Affiliation(s)
| | - Verena Kopatz
- Applied and Translational Radiology, Dept. Radiation Oncology, Medical University, Vienna, Austria
| | - Wolfgang Dörr
- Applied and Translational Radiology, Dept. Radiation Oncology, Medical University, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Medical University, Vienna, Austria
| | - Hans Geinitz
- Dept. of Radiation Oncology, Ordensklinikum Linz, Linz, Austria
| |
Collapse
|
173
|
IL-12 p40 monomer is different from other IL-12 family members to selectively inhibit IL-12Rβ1 internalization and suppress EAE. Proc Natl Acad Sci U S A 2020; 117:21557-21567. [PMID: 32817415 DOI: 10.1073/pnas.2000653117] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is the most common human demyelinating disease of the central nervous system. The IL-12 family of cytokines has four members, which are IL-12 (p40:p35), IL-23 (p40:p19), the p40 monomer (p40), and the p40 homodimer (p402). Since all four members contain p40 in different forms, it is important to use a specific monoclonal antibody (mAb) to characterize these molecules. Here, by using such mAbs, we describe selective loss of p40 in serum of MS patients as compared to healthy controls. Similarly, we also observed decrease in p40 and increase in IL-12, IL-23, and p402 in serum of mice with experimental autoimmune encephalomyelitis (EAE), an animal model of MS, as compared to control mice. Interestingly, weekly supplementation of mouse and human recombinant p40 ameliorated clinical symptoms and disease progression of EAE. On the other hand, IL-12, IL-23, and p402 did not exhibit such inhibitory effect. In addition to EAE, p40 also suppressed collagen-induced arthritis in mice. Using IL-12Rβ1-/-, IL-12Rβ2-/-, and IL-12Rβ1+/-/IL-12Rβ2-/- mice, we observed that p40 required IL-12Rβ1, but not IL-12Rβ2, to suppress EAE. Interestingly, p40 arrested IL-12-, IL-23-, or p402-mediated internalization of IL-12Rβ1, but neither IL-12Rβ2 nor IL-23R, protected regulatory T cells, and suppressed Th1 and Th17 biasness. These studies identify p40 as an anti-autoimmune cytokine with a biological role different from IL-12, IL-23, and p402 in which it attenuates autoimmune signaling via suppression of IL-12Rβ1 internalization, which may be beneficial in patients with MS and other autoimmune disorders.
Collapse
|
174
|
Rana J, Biswas M. Regulatory T cell therapy: Current and future design perspectives. Cell Immunol 2020; 356:104193. [PMID: 32823038 DOI: 10.1016/j.cellimm.2020.104193] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
Regulatory T cells (Tregs) maintain immune equilibrium by suppressing immune responses through various multistep contact dependent and independent mechanisms. Cellular therapy using polyclonal Tregs in transplantation and autoimmune diseases has shown promise in preclinical models and clinical trials. Although novel approaches have been developed to improve specificity and efficacy of antigen specific Treg based therapies, widespread application is currently restricted. To date, design-based approaches to improve the potency and persistence of engineered chimeric antigen receptor (CAR) Tregs are limited. Here, we describe currently available Treg based therapies, their advantages and limitations for implementation in clinical studies. We also examine various strategies for improving CAR T cell design that can potentially be applied to CAR Tregs, such as identifying co-stimulatory signalling domains that enhance suppressive ability, determining optimal scFv affinity/avidity, and co-expression of accessory molecules. Finally, we discuss the importance of tailoring CAR Treg design to suit the individual disease.
Collapse
Affiliation(s)
- Jyoti Rana
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
175
|
Arevalo-Villalobos JI, Govea-Alonso DO, Bañuelos-Hernández B, González-Ortega O, Zarazúa S, Rosales-Mendoza S. Inducible expression of antigens in plants: a study focused on peptides related to multiple sclerosis immunotherapy. J Biotechnol 2020; 318:51-56. [PMID: 32387449 DOI: 10.1016/j.jbiotec.2020.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 11/26/2022]
Abstract
Multiple sclerosis (MS) affects 2.3 million patients worldwide with no effective treatments available thus far. Depletion of autoreactive T-cells is considered the basis for immunotherapeutic approaches. For this purpose the peptides BV5S2, BV6S5, and BV13S1 have been identified as candidates for the development of a MS vaccine. Herein, the plant-based simultaneous production of these peptides is described as an effort to generate a new model of MS immunotherapy. A polyprotein comprising the sequence of the target peptides was designed having the picornaviral 2A sequence in between to mediate the release of the individual peptides upon translation. A codon optimized gene was cloned in vectors mediating constitutive (CaMV35S promoter) or inducible (AlcA promoter) expression. No transgenic tobacco plants were recovered from the constitutive vector suggesting toxicity of the target peptides. In contrast, several transformed lines were obtained with the inducible vector. The individual BV5S2, BV6S5, and BV13S1 peptides were detected in transformed lines upon ethanol-mediated induction and a quantitative analysis based on a OVA conjugate carrying the three peptides revealed accumulation levels up to 0.5 μg g-1 FW leaves. The plant-made peptides were able to induce humoral responses in orally immunized mice. This platform will be useful in the development of alternative immunotherapies against MS having low cost and safety as main attributes. Moreover the platform represents an attractive alternative for the expression of antigens having detrimental effects in plants.
Collapse
Affiliation(s)
- Jaime I Arevalo-Villalobos
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, 78210, Mexico; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª Sección, San Luis Potosí, 78210, Mexico
| | - Dania O Govea-Alonso
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, 78210, Mexico; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª Sección, San Luis Potosí, 78210, Mexico
| | - Bernardo Bañuelos-Hernández
- Escuela de Veterinaria, Universidad De La Salle Bajío, Avenida Universidad 602, Lomas del Campestre, 37150, León, Guanajuato, Mexico
| | - Omar González-Ortega
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, 78210, Mexico
| | - Sergio Zarazúa
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, 78210, Mexico
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, 78210, Mexico; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª Sección, San Luis Potosí, 78210, Mexico.
| |
Collapse
|
176
|
Obesity-associated T-cell and macrophage activation improve partly after a lifestyle intervention. Int J Obes (Lond) 2020; 44:1838-1850. [PMID: 32678324 DOI: 10.1038/s41366-020-0615-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 04/30/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND The relation between low-grade inflammation and metabolic dysfunction in obesity is not fully explored. OBJECTIVE To evaluate immune parameters in the obese state and after a lifestyle intervention program. METHODS Patients with obesity (n = 87) from an academic obesity clinic were compared with controls with regard to macrophage and T-cell activation (reflected by serum levels of soluble CD163 (sCD163) and soluble IL-2 receptor (sIL-2R), respectively), and an array of cytokines, chemokines, and growth factors. In addition, these parameters and regulatory T-cells (Treg), were studied in 27 patients who followed a 75-week lifestyle intervention (dietary advice, exercise, and psychoeducation). RESULTS Mean sIL-2R and sCD163 levels were higher in patients than controls (sIL-2R:2884 ± 936 pg/ml vs. 2207 ± 813 pg/ml, p = 0.001; sCD163:1279 ± 580 pg/ml vs. 661 ± 271 pg/ml, p < 0.0001 respectively). Patients with metabolic syndrome (MetS) had higher sCD163 than those without (1467 ± 656 pg/ml vs. 1103 ± 438 pg/ml). Patients had higher IL-1β, IL-1RA, IL-2, IL-4, IL-5, IL-6, IL-8, IL-9, IL-10, IL-15, IL-17A, MCP-1/CCL2, MIP-1α/CCL3, MIP-1β/CCL4, G-CSF, GM-CSF, FGF, IFN-γ, and TNF-α than controls, whereas VEGF-A, PDGF-BB, and eotaxin were lower. Upon intervention, sIL-2R decreased while peripheral Treg frequencies increased within the reference range (p = 0.042 and p = 0.005 respectively). The sIL-2R decrease correlated to a decrease in waist circumference (rho = 0.388, p = 0.045) and in trend to a decrease in MetS components (rho = 0.345, p = 0.078). The Treg increase was unrelated to weight loss or metabolic improvement. Mean sCD163 did not change significantly upon intervention, nor did the cytokines, chemokines, and growth factors (except IP-10/CXCL10). CONCLUSION In obesity, T-cell homeostasis improves after a lifestyle intervention. Immunologic alterations can occur independently of metabolic improvement.
Collapse
|
177
|
Jahangard L, Behzad M. Diminished functional properties of T regulatory cells in major depressive disorder: The influence of selective serotonin reuptake inhibitor. J Neuroimmunol 2020; 344:577250. [PMID: 32344162 DOI: 10.1016/j.jneuroim.2020.577250] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 11/15/2022]
Abstract
The properties of CD4+CD25hi T regulatory cells (Tregs), and interleukin (IL)-2 pathway were investigated in major depressive disorder (MDD) patients treated with or without selective serotonin reuptake inhibitor (SSRI). The frequencies of FOXP3 and pSTAT5 in peripheral Tregs were found to be diminished in untreated patients (SSRI-) versus HCs (p < .001 for both), while their percentages were increased in treated patients (SSRI+) versus untreated patients (p < .001 and p = .04). The proliferation of CD4+ T cells was higher in SSRI-MDD patients versus HCs (p = .03). The SSRI-MDD patients showed a lower concentration of supernatant TGF-β than HCs (p = .001), while the production of TGF-β was enhanced in SSRI+MDD versus SSRI-MDD patients (p = .003). The number of CD45RA-expressing Tregs, the expression of JAK1 and JAK3, and the levels of IL-2 and IL-10 were similar between the patients and HCs. The study results showed that untreated patients have an impaired IL-2 signaling pathway and defective Tregs, and SSRI treatment may improve the Tregs function.
Collapse
Affiliation(s)
- Leila Jahangard
- Research Center for Behavioral Disorders and Substances Abuse, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahdi Behzad
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
178
|
Song JH, Yu DH, Hwang TS, Seung BJ, Sur JH, Kim YJ, Jung DI. Expression of platelet-derived growth factor receptor-α/ß, vascular endothelial growth factor receptor-2, c-Abl, and c-Kit in canine granulomatous meningoencephalitis and necrotizing encephalitis. Vet Med Sci 2020; 6:965-974. [PMID: 32585777 PMCID: PMC7738704 DOI: 10.1002/vms3.314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/11/2020] [Accepted: 05/27/2020] [Indexed: 01/21/2023] Open
Abstract
Background Given the active research on targeted therapy using tyrosine kinase (TK) inhibitors (TKIs) in the field of oncology, further studies have recently been conducted to evaluate their use in autoimmune disorders. Based on immunological investigations, previous studies have suggested that granulomatous meningoencephalomyelitis (GME) and necrotizing encephalomyelitis (NE) are similar to multiple sclerosis (MS), which is a human autoimmune demyelinating central nervous system disease. Objectives Considering this perspective, we hypothesized that canine GME and NE have significant expression of one or more TKs, which are associated with human MS pathogenesis. Methods To determine the possible use of conventional multi‐targeted TKIs as a treatment for canine GME and NE, we characterized the immunohistochemical expression of platelet‐derived growth factor receptor (PDGFR)‐α, PDGFR‐ß, vascular endothelial growth factor receptor (VEGFR)‐2, c‐Abl and c‐Kit in GME and NE samples. Results Histological samples from four dogs with GME and three with NE were retrieved. All samples stained positive for PDGFR‐ß (7/7 [100%]). PDGFR‐α and c‐Kit were expressed in 3/7 (42.8%) samples each. c‐Abl was identified in 2/7 (28.5%) samples; no sample showed VEGFR‐2 (0%) expression. Co‐expression of TKs was identified in 6/7 (85.7%) dogs. Conclusions All samples were positive for at least one or more of PDGFR‐α, PDGFR‐ß, c‐Kit and c‐Abl, which are known as the target TKs of conventional multi‐targeted TKIs. Their presence does suggest that these TKs may play a role in the pathogenesis of GME and NE. Therefore, multi‐targeted TKIs may provide benefits in the treatment of canine GME and NE by suppressing the activity of these TKs.
Collapse
Affiliation(s)
- Joong-Hyun Song
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Do-Hyeon Yu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Tae-Sung Hwang
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Byung-Joon Seung
- Department of Pathobiology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jung-Hyang Sur
- Department of Pathobiology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Young Joo Kim
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, 91766-1854, USA
| | - Dong-In Jung
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| |
Collapse
|
179
|
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: 32] [Impact Index Per Article: 8.0] [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
|
180
|
Rojas C, Campos-Mora M, Cárcamo I, Villalón N, Elhusseiny A, Contreras-Kallens P, Refisch A, Gálvez-Jirón F, Emparán I, Montoya-Riveros A, Vernal R, Pino-Lagos K. T regulatory cells-derived extracellular vesicles and their contribution to the generation of immune tolerance. J Leukoc Biol 2020; 108:813-824. [PMID: 32531824 DOI: 10.1002/jlb.3mr0420-533rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
T regulatory (Treg) cells have a major role in the maintenance of immune tolerance against self and foreign antigens through the control of harmful inflammation. Treg cells exert immunosuppressive function by several mechanisms, which can be distinguished as contact dependent or independent. Recently, the secretion of extracellular vesicles (EVs) by Treg cells has been reported as a novel suppressive mechanism capable of modulating immunity in a cell-contact independent and targeted manner, which has been identified in different pathologic scenarios. EVs are cell-derived membranous structures involved in physiologic and pathologic processes through protein, lipid, and genetic material exchange, which allow intercellular communication. In this review, we revise and discuss current knowledge on Treg cells-mediated immune tolerance giving special attention to the production and release of EVs. Multiple studies support that Treg cells-derived EVs represent a refined intercellular exchange device with the capacity of modulating immune responses, thus creating a tolerogenic microenvironment in a cell-free manner. The mechanisms proposed encompass miRNAs-induced gene silencing, the action of surface proteins and the transmission of enzymes. These observations gain relevance by the fact that Treg cells are susceptible to converting into effector T cells after exposition to inflammatory environments. Yet, in contrast to their cells of origin, EVs are unlikely to be modified under inflammatory conditions, highlighting the advantage of their use. Moreover, we speculate in the possibility that Treg cells may contribute to infectious tolerance via vesicle secretion, intervening with CD4+ T cells differentiation and/or stability.
Collapse
Affiliation(s)
- Carolina Rojas
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile.,Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Las Condes, Santiago, Chile
| | - Mauricio Campos-Mora
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ignacio Cárcamo
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Natalia Villalón
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ahmed Elhusseiny
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Pamina Contreras-Kallens
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Aarón Refisch
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Felipe Gálvez-Jirón
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ivana Emparán
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Andro Montoya-Riveros
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Las Condes, Santiago, Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| |
Collapse
|
181
|
Machhi J, Kevadiya BD, Muhammad IK, Herskovitz J, Olson KE, Mosley RL, Gendelman HE. Harnessing regulatory T cell neuroprotective activities for treatment of neurodegenerative disorders. Mol Neurodegener 2020; 15:32. [PMID: 32503641 PMCID: PMC7275301 DOI: 10.1186/s13024-020-00375-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence demonstrates that adaptive immunity influences the pathobiology of neurodegenerative disorders. Misfolded aggregated self-proteins can break immune tolerance leading to the induction of autoreactive effector T cells (Teffs) with associated decreases in anti-inflammatory neuroprotective regulatory T cells (Tregs). An imbalance between Teffs and Tregs leads to microglial activation, inflammation and neuronal injury. The cascade of such a disordered immunity includes the drainage of the aggregated protein antigens into cervical lymph nodes serving to amplify effector immune responses. Both preclinical and clinical studies demonstrate transformation of this altered immunity for therapeutic gain. We posit that the signs and symptoms of common neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, and stroke can be attenuated by boosting Treg activities.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Bhavesh D. Kevadiya
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Radiology, School of Medicine, Stanford University, Palo Alto, 94304 USA
| | - Ijaz Khan Muhammad
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacy, University of Swabi, Anbar Swabi, 23561 Pakistan
| | - Jonathan Herskovitz
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
182
|
Ghavimi R, Alsahebfosoul F, Salehi R, Kazemi M, Etemadifar M, Zavaran Hosseini A. High-resolution melting curve analysis of polymorphisms within CD58, CD226, HLA-G genes and association with multiple sclerosis susceptibility in a subset of Iranian population: a case-control study. Acta Neurol Belg 2020; 120:645-652. [PMID: 30128676 DOI: 10.1007/s13760-018-0992-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system with unknown etiology, which typically is manifested in early to middle adulthood. Recently, genome-wide association studies have identified susceptibility of immune-related genes to be involved in MS predisposition. The goal of the current study was to investigate the association of single nucleotide polymorphisms (SNP) with the immunologically related genes responsible for the disease, composed of CD58 (rs2300747 A>G), CD226 (rs763361 C>T), and HLA-G (rs1611715 A>C), with MS susceptibility. In this case-control study, a total of 200 patients suffering from relapsing-remitting multiple sclerosis and 200 healthy individuals were recruited. DNA was extracted from blood and then all subjects were genotyped for the polymorphism within mentioned genes by high-resolution melting (HRM) real-time PCR method. Statistical analyses were performed using SPSS software (version 20; SPSS, Chicago, IL, USA). Our finding showed that there are significant differences in genotype and allele frequencies between two groups regarding rs763361 (P = 0.035, OR 0.64, CI 95% for C allele) and rs1611715 (P = 0.038, OR 1.57, CI 95% for AA genotype) polymorphisms within CD226 and HLA-G genes, respectively. Concerning rs2300747 polymorphism on CD58 gene, no significant differences were found between cases and controls. In general, results from the current study indicate that CD226 and HLA-G, but not CD58 genetic polymorphisms are associated with increased risk of MS in Isfahan population similar to European populations. However, to elucidate how these SNPs contribute to MS pathogenesis, functional studies are needed.
Collapse
|
183
|
Barp A, Gerardi F, Lizio A, Sansone VA, Lunetta C. Emerging Drugs for the Treatment of Amyotrophic Lateral Sclerosis: A Focus on Recent Phase 2 Trials. Expert Opin Emerg Drugs 2020; 25:145-164. [PMID: 32456491 DOI: 10.1080/14728214.2020.1769067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease involving both upper and lower motor neurons and resulting in increasing disability and death 3-5 years after onset of symptoms. Over 40 large clinical trials for ALS have been negative, except for Riluzole that offers a modest survival benefit, and Edaravone that modestly reduces disease progression in patients with specific characteristics. Thus, the discovery of efficient disease modifying therapy is an urgent need. AREAS COVERED Although the cause of ALS remains unclear, many studies have demonstrated that neuroinflammation, proteinopathies, glutamate-induced excitotoxicity, microglial activation, oxidative stress, and mitochondrial dysfunction may play a key role in the pathogenesis. This review highlights recent discoveries relating to these diverse mechanisms and their implications for the development of therapy. Ongoing phase 2 clinical trials aimed to interfere with these pathophysiological mechanisms are discussed. EXPERT OPINION This review describes the challenges that the discovery of an efficient drug therapy faces and how these issues may be addressed. With the continuous advances coming from basic research, we provided possible suggestions that may be considered to improve performance of clinical trials and turn ALS research into a 'fertile ground' for drug development for this devastating disease.
Collapse
Affiliation(s)
- Andrea Barp
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy.,Dept. Biomedical Sciences of Health, University of Milan , Milan, Italy
| | | | - Andrea Lizio
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy
| | - Valeria Ada Sansone
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy.,Dept. Biomedical Sciences of Health, University of Milan , Milan, Italy
| | | |
Collapse
|
184
|
Fan NW, Dohlman TH, Foulsham W, McSoley M, Singh RB, Chen Y, Dana R. The role of Th17 immunity in chronic ocular surface disorders. Ocul Surf 2020; 19:157-168. [PMID: 32470612 DOI: 10.1016/j.jtos.2020.05.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 12/23/2022]
Abstract
Th17 cells have been implicated in the pathogenesis of numerous inflammatory and autoimmune conditions. At the ocular surface, Th17 cells have been identified as key effector cells in chronic ocular surface disease. Evidence from murine studies indicates that following differentiation and expansion, Th17 cells migrate from the lymphoid tissues to the eye, where they release inflammatory cytokines including, but not limited to, their hallmark cytokine IL-17A. As the acute phase subsides, a population of long-lived memory Th17 cells persist, which predispose hosts both to chronic inflammation and severe exacerbations of disease; of great interest is the small subset of Th17/1 cells that secrete both IL-17A and IFN-γ in acute-on-chronic disease exacerbation. Over the past decade, substantial progress has been made in deciphering how Th17 cells interact with the immune and neuroimmune pathways that mediate chronic ocular surface disease. Here, we review (i) the evidence for Th17 immunity in chronic ocular surface disease, (ii) regulatory mechanisms that constrain the Th17 immune response, and (iii) novel therapeutic strategies targeting Th17 cells.
Collapse
Affiliation(s)
- Nai-Wen Fan
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Thomas H Dohlman
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Matthew McSoley
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA; University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Rohan Bir Singh
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Yihe Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
185
|
van Langelaar J, Rijvers L, Smolders J, van Luijn MM. B and T Cells Driving Multiple Sclerosis: Identity, Mechanisms and Potential Triggers. Front Immunol 2020; 11:760. [PMID: 32457742 PMCID: PMC7225320 DOI: 10.3389/fimmu.2020.00760] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/03/2020] [Indexed: 12/25/2022] Open
Abstract
Historically, multiple sclerosis (MS) has been viewed as being primarily driven by T cells. However, the effective use of anti-CD20 treatment now also reveals an important role for B cells in MS patients. The results from this treatment put forward T-cell activation rather than antibody production by B cells as a driving force behind MS. The main question of how their interaction provokes both B and T cells to infiltrate the CNS and cause local pathology remains to be answered. In this review, we highlight key pathogenic events involving B and T cells that most likely contribute to the pathogenesis of MS. These include (1) peripheral escape of B cells from T cell-mediated control, (2) interaction of pathogenic B and T cells in secondary lymph nodes, and (3) reactivation of B and T cells accumulating in the CNS. We will focus on the functional programs of CNS-infiltrating lymphocyte subsets in MS patients and discuss how these are defined by mechanisms such as antigen presentation, co-stimulation and cytokine production in the periphery. Furthermore, the potential impact of genetic variants and viral triggers on candidate subsets will be debated in the context of MS.
Collapse
Affiliation(s)
- Jamie van Langelaar
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Liza Rijvers
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Joost Smolders
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Department of Neurology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Marvin M. van Luijn
- Department of Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
186
|
Clough JN, Omer OS, Tasker S, Lord GM, Irving PM. Regulatory T-cell therapy in Crohn's disease: challenges and advances. Gut 2020; 69:942-952. [PMID: 31980447 PMCID: PMC7229901 DOI: 10.1136/gutjnl-2019-319850] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/21/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Abstract
The prevalence of IBD is rising in the Western world. Despite an increasing repertoire of therapeutic targets, a significant proportion of patients suffer chronic morbidity. Studies in mice and humans have highlighted the critical role of regulatory T cells in immune homeostasis, with defects in number and suppressive function of regulatory T cells seen in patients with Crohn's disease. We review the function of regulatory T cells and the pathways by which they exert immune tolerance in the intestinal mucosa. We explore the principles and challenges of manufacturing a cell therapy, and discuss clinical trial evidence to date for their safety and efficacy in human disease, with particular focus on the development of a regulatory T-cell therapy for Crohn's disease.
Collapse
Affiliation(s)
- Jennie N Clough
- School of Immunology and Microbial Sciences, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and Saint Thomas' NHS Foundation Trust and King's College, London, UK
| | - Omer S Omer
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Department of Gastroenterology, Guy's and Saint Thomas' Hospitals NHS Trust, London, UK
| | - Scott Tasker
- Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Peter M Irving
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Department of Gastroenterology, Guy's and Saint Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
187
|
Haddadi MH, Hajizadeh-Saffar E, Khosravi-Maharlooei M, Basiri M, Negahdari B, Baharvand H. Autoimmunity as a target for chimeric immune receptor therapy: A new vision to therapeutic potential. Blood Rev 2020; 41:100645. [DOI: 10.1016/j.blre.2019.100645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/12/2019] [Accepted: 11/22/2019] [Indexed: 12/25/2022]
|
188
|
Husseiny MI, Fahmy A, Du W, Gu A, Garcia P, Ferreri K, Kandeel F. Development of Quantitative Methylation-Specific Droplet Digital PCR (ddMSP) for Assessment of Natural Tregs. Front Genet 2020; 11:300. [PMID: 32318096 PMCID: PMC7154152 DOI: 10.3389/fgene.2020.00300] [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: 11/29/2019] [Accepted: 03/13/2020] [Indexed: 11/13/2022] Open
Abstract
Regulatory T cells (Tregs) suppress immune responses in vivo in an antigen-specific manner. Of clinical relevance, Tregs can be isolated and expanded in vitro while maintaining immunoregulatory function. Tregs are classified as CD4+CD25highCD127low FOXP3+ cells. Demethylation of the Treg-specific demethylation region (TSDR) of FOXP3 is found in natural Tregs (nTregs). We report a method for the characterization of the differential methylation pattern of the FOXP3 TSDR in patient-derived and expanded nTregs. Human TSDR sequences from nTregs (unmethylated sequence) and pancreatic (methylated sequence) cells were amplified and cloned into plasmids. A droplet digital TaqMan probe-based qPCR (ddPCR) assay using methylation-specific primers and probes was employed to quantify unmethylated and methylated sequences. The methylation-specific droplet digital PCR (ddMSP) assay was specific and selective for unmethylated DNA in mixtures with methylated DNA in the range of 5000 copies/μL to less than 1 copy/μL (R 2 = 0.99) even in the presence of non-selective gDNAs. CD4+CD25highCD127lowFOXP3+ human nTregs, in the presence of Dynabeads or activators, were expanded for 21 days. There was a decrease in the unmethylated ratio of Tregs after expansion with essentially the same ratio at days 10, 14, and 17. However, the activator expanded group showed a significant decrease in unmethylated targets at day 21. The suppression activity of activator-expanded nTregs at day 21 was decreased compared to cells expanded with Dynabeads. These data suggest that the ddMSP can quantitatively monitor nTreg expansion in vitro. These data also indicate that the assay is sensitive and specific at differentiating nTregs from other cells and may be useful for rapid screening of nTregs in clinical protocols.
Collapse
Affiliation(s)
- Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, United States.,Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ahmed Fahmy
- East Lancashire Hospitals NHS Trust, Blackburn, United Kingdom
| | - Weiting Du
- Department of Translational Research & Cellular Therapeutics, Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Angel Gu
- Department of Translational Research & Cellular Therapeutics, Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Pablo Garcia
- Department of Translational Research & Cellular Therapeutics, Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Kevin Ferreri
- Department of Translational Research & Cellular Therapeutics, Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, United States
| |
Collapse
|
189
|
Zhang X, Olsen N, Zheng SG. The progress and prospect of regulatory T cells in autoimmune diseases. J Autoimmun 2020; 111:102461. [PMID: 32305296 DOI: 10.1016/j.jaut.2020.102461] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Treg) are an important immune cell population, playing a crucial role in regulating immune tolerance and preventing autoimmune diseases. These cells consist of various cell sub-populations and generally have an immunoregulatory or suppressive role against immune responses. They also have a different cell heterogeneity and each populations has own biological characteristics. Treg deficiency, reduction, instability, reduced vitality and dysfunction all account for multiple autoimmune diseases. In this review, we have systemically reviewed Treg classification, phenotypic features, regulation of Foxp3 expression, plasticity and stability of Treg as well as their relationship with several important autoimmune diseases. We particularly focus on why and how inflammatory and diet environments affect the functional capacity and underlying mechanisms of Treg cell populations. We also summarize new advances in technologies which help to analyze and dissect these cells in molecular levels in-depth. We also clarify the possible clinical relevance on application of these cells in patients with autoimmune diseases. The advantages and weaknesses have been carefully discussed as well. We also propose the possible approaches to overcome these weaknesses of Treg cells in complicate environments. Thus, we have displayed the updated knowledge of Treg cells, which provides an overall insight into the role and mechanisms of Treg cells in autoimmune diseases.
Collapse
Affiliation(s)
- Ximei Zhang
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China; Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA
| | - Nancy Olsen
- Division of Rheumatology, Department of Medicine at Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey, 17033, USA
| | - Song Guo Zheng
- Division of Rheumatology and Immunology, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, PA, 43201, USA.
| |
Collapse
|
190
|
Manuszak C, Brainard M, Thrash E, Hodi FS, Severgnini M. Standardized 11-color flow cytometry panel for the functional phenotyping of human T regulatory cells. J Biol Methods 2020; 7:e131. [PMID: 32313815 PMCID: PMC7163208 DOI: 10.14440/jbm.2020.325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/02/2020] [Accepted: 03/21/2020] [Indexed: 12/17/2022] Open
Abstract
T regulatory cells (Tregs) are a cell subset that can suppress immune responses to maintain homeostasis and self-tolerance. In some scenarios, the immunosuppressive nature could be associated to other pathological developments such as autoimmune diseases and cancers. Due to the importance of Tregs in disease pathogenesis, we developed and validated an 11-color flow cytometry panel for phenotypic and functional detection of Treg markers using healthy human donor peripheral blood mononuclear cells (PBMCs). Our panel contains 4 Treg surface proteins and 2 functional cytokines as well as T-lymphocyte lineage markers CD3, CD4, and CD8. Our data shows an increase in expression of markers CD25, FoxP3, CTLA4, GITR and intracellular cytokines IL4 and TGFβ when comparing unstimulated samples to CD3/CD28 bead stimulated samples. This 11-color panel can be used to functionally evaluate immunosuppressive Tregs in human PBMC samples.
Collapse
Affiliation(s)
- Claire Manuszak
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - Martha Brainard
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - Emily Thrash
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - Mariano Severgnini
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| |
Collapse
|
191
|
Melero-Jerez C, Alonso-Gómez A, Moñivas E, Lebrón-Galán R, Machín-Díaz I, de Castro F, Clemente D. The proportion of myeloid-derived suppressor cells in the spleen is related to the severity of the clinical course and tissue damage extent in a murine model of multiple sclerosis. Neurobiol Dis 2020; 140:104869. [PMID: 32278882 DOI: 10.1016/j.nbd.2020.104869] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/28/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple Sclerosis (MS) is the second cause of paraplegia among young adults, after all types of CNS traumatic lesions. In its most frequent relapsing-remitting form, the severity of the disease course is very heterogeneous, and its reliable evaluation remains a key issue for clinicians. Myeloid-Derived sSuppressor Cells (MDSCs) are immature myeloid cells that suppress the inflammatory response, a phenomenon related to the resolution or recovery of the clinical symptoms associated with experimental autoimmune encephalomyelitis (EAE), the most common model for MS. Here, we establish the severity index as a new parameter for the clinical assessment in EAE. It is derived from the relationship between the maximal clinical score and the time elapsed since disease onset. Moreover, we relate this new index with several histopathological hallmarks in EAE and with the peripheral content of MDSCs. Based on this new parameter, we show that the splenic MDSC content is related to the evolution of the clinical course of EAE, ranging from mild to severe. Indeed, when the severity index indicates a severe disease course, EAE mice display more intense lymphocyte infiltration, demyelination and axonal damage. A direct correlation was drawn between the MDSC population in the peripheral immune system, and the preservation of myelin and axons, which was also correlated with T cell apoptosis within the CNS (being these cells the main target for MDSC suppression). The data presented clearly indicated that the severity index is a suitable tool to analyze disease severity in EAE. Moreover, our data suggest a clear relationship between circulating MDSC enrichment and disease outcome, opening new perspectives for the future targeting of this population as an indicator of MS severity.
Collapse
Affiliation(s)
- Carolina Melero-Jerez
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain; Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Aitana Alonso-Gómez
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Esther Moñivas
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Rafael Lebrón-Galán
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Isabel Machín-Díaz
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Instituto Cajal-CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain.
| | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain.
| |
Collapse
|
192
|
Ching YM, Viswanathan S, Mohamed Nor N, Shuib S, Kamarudin B, Mansor S, Yusof AY, Arip M. Association of CD58 polymorphism and multiple sclerosis in Malaysia: a pilot study. AUTOIMMUNITY HIGHLIGHTS 2020; 10:13. [PMID: 32257069 PMCID: PMC7065364 DOI: 10.1186/s13317-019-0123-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/06/2019] [Indexed: 11/15/2022]
Abstract
Background Multiple sclerosis is an immune mediated disease targeting the central nervous system. Association of non-human leukocyte antigen gene, CD58, with multiple sclerosis has been reported in several populations but is unclear among Southeast Asians. This pilot study was conducted to explore the association between CD58 polymorphism and multiple sclerosis among the Malay population in Malaysia. Methods Blood samples were collected from 27 multiple sclerosis patients, and compared with 58 age- and gender matched healthy individuals. All patients were tested negative for anti-aquaporin 4. DNA was extracted from the blood and genotyped for 3 single nucleotide polymorphisms rs12044852, rs2300747 and rs1335532 of gene CD58 by real-time PCR. Results The majority of multiple sclerosis patients were female (85.2%). The general mean age of onset was 30.5 years. Genotyping results showed that frequencies of the alleles were between 40 and 50% for MS patients and healthy individuals. Association (allelic model) between multiple sclerosis and CD58 gene polymorphism alleles rs12044852 (p = 0.410), rs2300747 (p = 0.881) and rs1335532 (p = 0.407) were indistinct. Conclusions The impact of the CD58 gene polymorphism was not prominent in this pilot study, implying that genetic composition contributing to multiple sclerosis may be different between different populations, thus results in a heterogeneity of disease manifestation and distribution.
Collapse
Affiliation(s)
- Yee Ming Ching
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | | | - Nurhanani Mohamed Nor
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Shuwahida Shuib
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Balqis Kamarudin
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Salawati Mansor
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Ainur Yusniza Yusof
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia
| | - Masita Arip
- Autoimmune Unit, Allergy and Immunology Research Centre, Institute for Medical Research, National Institute of Health, Selangor, Malaysia.,3Allergy and Immunology Research Centre, Institute for Medical Research, Block C6, National Institute of Health Complex, No. 1, Jalan Setia Murni U13/52, Seksyen U13, Bandar Setia Alam, 40170 Shah Alam, Selangor Darul Ehsan Malaysia
| |
Collapse
|
193
|
Modulation of regulatory T cell function and stability by co-inhibitory receptors. Nat Rev Immunol 2020; 20:680-693. [PMID: 32269380 DOI: 10.1038/s41577-020-0296-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
Regulatory T (Treg) cells constitute a dynamic population that is essential for controlling immune responses in health and disease. Defects in Treg cell function and decreases in Treg cell numbers have been observed in patients with autoimmunity and the opposite effects on Treg cells occur in cancer settings. Current research on new therapies for these diseases is focused on modulating Treg cell function to increase or decrease suppressive activity in autoimmunity and cancer, respectively. In this regard, several co-inhibitory receptors that are preferentially expressed by Treg cells under homeostatic conditions have recently been shown to control Treg cell function and stability in different disease settings. These receptors could be amenable to therapeutic targeting aimed at modulating Treg cell function and plasticity. This Review summarizes recent data regarding the role of co-inhibitory molecules in the control of Treg cell function and stability, with a focus on their roles and potential therapeutic use in autoimmunity and cancer.
Collapse
|
194
|
Calvo-Barreiro L, Eixarch H, Ponce-Alonso M, Castillo M, Lebrón-Galán R, Mestre L, Guaza C, Clemente D, del Campo R, Montalban X, Espejo C. A Commercial Probiotic Induces Tolerogenic and Reduces Pathogenic Responses in Experimental Autoimmune Encephalomyelitis. Cells 2020; 9:cells9040906. [PMID: 32272791 PMCID: PMC7226819 DOI: 10.3390/cells9040906] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/28/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
Previous studies in experimental autoimmune encephalomyelitis (EAE) models have shown that some probiotic bacteria beneficially impact the development of this experimental disease. Here, we tested the therapeutic effect of two commercial multispecies probiotics—Lactibiane iki and Vivomixx—on the clinical outcome of established EAE. Lactibiane iki improves EAE clinical outcome in a dose-dependent manner and decreases central nervous system (CNS) demyelination and inflammation. This clinical improvement is related to the inhibition of pro-inflammatory and the stimulation of immunoregulatory mechanisms in the periphery. Moreover, both probiotics modulate the number and phenotype of dendritic cells (DCs). Specifically, Lactibiane iki promotes an immature, tolerogenic phenotype of DCs that can directly induce immune tolerance in the periphery, while Vivomixx decreases the percentage of DCs expressing co-stimulatory molecules. Finally, gut microbiome analysis reveals an altered microbiome composition related to clinical condition and disease progression. This is the first preclinical assay that demonstrates that a commercial probiotic performs a beneficial and dose-dependent effect in EAE mice and one of the few that demonstrates a therapeutic effect once the experimental disease is established. Because this probiotic is already available for clinical trials, further studies are being planned to explore its therapeutic potential in multiple sclerosis patients.
Collapse
Affiliation(s)
- Laura Calvo-Barreiro
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya, Vall d’Hebron Institut de Recerca, Hospital Universitari Vall d’Hebron, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (L.C.-B.); (H.E.); (M.C.); (X.M.)
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
| | - Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya, Vall d’Hebron Institut de Recerca, Hospital Universitari Vall d’Hebron, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (L.C.-B.); (H.E.); (M.C.); (X.M.)
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
| | - Manuel Ponce-Alonso
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Carretera de Colmenar km. 9.1, 28034 Madrid, Spain; (M.P.-A.); (R.d.C.)
| | - Mireia Castillo
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya, Vall d’Hebron Institut de Recerca, Hospital Universitari Vall d’Hebron, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (L.C.-B.); (H.E.); (M.C.); (X.M.)
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
| | - Rafael Lebrón-Galán
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
- Grupo de Neuroinmuno-Reparación, Unidad de Investigación, Hospital Nacional de Parapléjicos, Finca “La Peraleda” s/n, 45071 Toledo, Spain
| | - Leyre Mestre
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
- Grupo de Neuroinmunología, Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Carmen Guaza
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
- Grupo de Neuroinmunología, Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Diego Clemente
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
- Grupo de Neuroinmuno-Reparación, Unidad de Investigación, Hospital Nacional de Parapléjicos, Finca “La Peraleda” s/n, 45071 Toledo, Spain
| | - Rosa del Campo
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Carretera de Colmenar km. 9.1, 28034 Madrid, Spain; (M.P.-A.); (R.d.C.)
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya, Vall d’Hebron Institut de Recerca, Hospital Universitari Vall d’Hebron, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (L.C.-B.); (H.E.); (M.C.); (X.M.)
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
- Division of Neurology, University of Toronto, St. Michael’s Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya, Vall d’Hebron Institut de Recerca, Hospital Universitari Vall d’Hebron, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (L.C.-B.); (H.E.); (M.C.); (X.M.)
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain
- Red Española de Esclerosis Múltiple (REEM), Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, 28801 Madrid, Spain; (R.L.-G.); (L.M.); (C.G.); (D.C.)
- Correspondence: ; Tel.: +34-93-489-3599
| |
Collapse
|
195
|
Bahrini K, Belghith M, Maghrebi O, Bekir J, Kchaou M, Jeridi C, Amouri R, Hentati F, Belal S, Ben Sassi S, Barbouche MR. Discriminative expression of CD39 and CD73 in Cerebrospinal fluid of patients with Multiple Sclerosis and Neuro-Behçet's disease. Cytokine 2020; 130:155054. [PMID: 32151963 DOI: 10.1016/j.cyto.2020.155054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 12/11/2022]
Abstract
Treg-mediated immune suppression involves many molecular mechanisms including the cleavage of inflammatory extracellular ATP to adenosine by CD39 ectoenzyme. In the peripheral blood of Multiple Sclerosis (MS) patients, it has been suggested that CD39+ Treg cells have the potential to suppress pro-inflammatory IL-17 secreting cells. Herein, we studied cellular phenotype and mRNA expression of CD39 and CD73 ectoenzymes in the Cerebrospinal fluid (CSF) of MS patients and another neuro-inflammatory disease: the Neuro-behçet's disease (NBD). Using qRT-PCR, we assessed mRNA expression of CD39 and CD73 as well as anti-inflammatory (IL-10) and pro-inflammatory (IL-6, TNF-α, IL-1β) cytokines in patients Peripheral blood mononuclear cells (PBMCs) and CSF of 28 relapsing-remitting multiple sclerosis (RRMS), 20 NBD and 22 controls with non inflammatory neurological disorders (NIND). The most substantial result in the CSF was the higher expression of CD39 in both RRMS and NBD patients compared to NIND. While, the expression of CD73 in CSF samples of NBD was low. In RRMS samples, we detected a significant positive correlation of both CD39 and CD73 with IL-10 expression. Moreover, results by flow cytometry revealed a high percentage of CD39 Treg cells in RRMS CSF. CD39 was preferentially expressed on B cells of NBD. Regarding inflammatory response, we showed a significant increase of IL-6 mRNA expression in NBD patients CSF while in RRMS this increase concerned TNF-α. These results bring evidence that CD39 correlates positively with an anti-inflammatory IL-10 response in RRMS. In contrast, no such association was observed in CSF of NBD patients and CD39 was preferentially expressed on B cells.
Collapse
Affiliation(s)
- Khadija Bahrini
- Institut Pasteur de Tunis, LTCII, LR11IPT02, Tunis 1002, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Meriam Belghith
- Institut Pasteur de Tunis, LTCII, LR11IPT02, Tunis 1002, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Olfa Maghrebi
- Institut Pasteur de Tunis, LTCII, LR11IPT02, Tunis 1002, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Jihène Bekir
- Institut Pasteur de Tunis, LTCII, LR11IPT02, Tunis 1002, Tunisia; Tunis El Manar University, Tunis 1068, Tunisia.
| | - Mariem Kchaou
- Neurological Department of Charles Nicolle Hospital, Tunis, Tunisia.
| | - Cyrine Jeridi
- Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia.
| | - Rim Amouri
- Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia.
| | - Faycel Hentati
- Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia.
| | - Samir Belal
- Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia; Faculty of Medicine of Tunis, 1006, Tunisia.
| | - Samia Ben Sassi
- Neurology's Department, Mongi Ben Hmida National Institute of Neurology, Tunis, Tunisia.
| | - Mohamed-Ridha Barbouche
- Institut Pasteur de Tunis, LTCII, LR11IPT02, Tunis 1002, Tunisia; Faculty of Medicine of Tunis, 1006, Tunisia.
| |
Collapse
|
196
|
Cellerino M, Ivaldi F, Pardini M, Rotta G, Vila G, Bäcker-Koduah P, Berge T, Laroni A, Lapucci C, Novi G, Boffa G, Sbragia E, Palmeri S, Asseyer S, Høgestøl E, Campi C, Piana M, Inglese M, Paul F, Harbo HF, Villoslada P, Kerlero de Rosbo N, Uccelli A. Impact of treatment on cellular immunophenotype in MS: A cross-sectional study. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/3/e693. [PMID: 32139439 PMCID: PMC7136062 DOI: 10.1212/nxi.0000000000000693] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/05/2020] [Indexed: 12/22/2022]
Abstract
Objective To establish cytometry profiles associated with disease stages and immunotherapy in MS. Methods Demographic/clinical data and peripheral blood samples were collected from 227 patients with MS and 82 sex- and age-matched healthy controls (HCs) enrolled in a cross-sectional study at 4 European MS centers (Spain, Italy, Germany, and Norway). Flow cytometry of isolated peripheral blood mononuclear cells was performed in each center using specifically prepared antibody-cocktail Lyotubes; data analysis was centralized at the Genoa center. Differences in immune cell subsets were assessed between groups of untreated patients with relapsing-remitting or progressive MS (RRMS or PMS) and HCs and between groups of patients with RRMS taking 6 commonly used disease-modifying drugs. Results In untreated patients with MS, significantly higher frequencies of Th17 cells in the RRMS population compared with HC and lower frequencies of B-memory/B-regulatory cells as well as higher percentages of B-mature cells in patients with PMS compared with HCs emerged. Overall, the greatest deviation in immunophenotype in MS was observed by treatment rather than disease course, with the strongest impact found in fingolimod-treated patients. Fingolimod induced a decrease in total CD4+ T cells and in B-mature and B-memory cells and increases in CD4+ and CD8+ T-regulatory and B-regulatory cells. Conclusions Our highly standardized, multisite cytomics data provide further understanding of treatment impact on MS immunophenotype and could pave the way toward monitoring immune cells to help clinical management of MS individuals.
Collapse
Affiliation(s)
- Maria Cellerino
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Federico Ivaldi
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Matteo Pardini
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Gianluca Rotta
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Gemma Vila
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Priscilla Bäcker-Koduah
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Tone Berge
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Alice Laroni
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Caterina Lapucci
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Giovanni Novi
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Giacomo Boffa
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Elvira Sbragia
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Serena Palmeri
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Susanna Asseyer
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Einar Høgestøl
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Cristina Campi
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Michele Piana
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Matilde Inglese
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Friedemann Paul
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Hanne F Harbo
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Pablo Villoslada
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Nicole Kerlero de Rosbo
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy
| | - Antonio Uccelli
- From the Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (M.C., F.I., M.P., A.L., C.L., G.N., G.B., E.S., S.P., M.I., N.K.d.R.) and Center of Excellence for Biomedical Research (A.U.), University of Genoa, Italy; BD Biosciences Italy (G.R.), Milan; Institut d'Investigacions Biomediques August Pi Sunyer (G.V., P.V.), Barcelona, Spain; Charité Universitaetsmedizin Berlin and Max Delbrueck Center for Molecular Medicine (P.B.-K., S.A., F.P.), Germany; Department of Research, Innovation and Education (T.B.), Neuroscience Research Unit, Oslo University Hospital; Department of Mechanical Electronics and Chemical Engineering (T.B.), Oslo Metropolitan University, Norway; University of Oslo (E.H., H.F.H.) and Oslo University Hospital (H.F.H.), Norway; Department of Mathematics and Padova Neuroscience Center (C.C.), University of Padua, Italy; Department of Mathematics (M.P.), University of Genoa, Italy; and IRCCS Ospedale Policlinico San Martino (A.L., M.P., M.I., A.U.), Genoa, Italy.
| |
Collapse
|
197
|
Arroyo Hornero R, Hamad I, Côrte-Real B, Kleinewietfeld M. The Impact of Dietary Components on Regulatory T Cells and Disease. Front Immunol 2020; 11:253. [PMID: 32153577 PMCID: PMC7047770 DOI: 10.3389/fimmu.2020.00253] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
The rise in the prevalence of autoimmune diseases in developed societies has been associated with a change in lifestyle patterns. Among other factors, increased consumption of certain dietary components, such as table salt and fatty acids and excessive caloric intake has been associated with defective immunological tolerance. Dietary nutrients have shown to modulate the immune response by a direct effect on the function of immune cells or, indirectly, by acting on the microbiome of the gastrointestinal tract. FOXP3+ regulatory T cells (Tregs) suppress immune responses and are critical for maintaining peripheral tolerance and immune homeostasis, modulating chronic tissue inflammation and autoimmune disease. It is now well-recognized that Tregs show certain degree of plasticity and can gain effector functions to adapt their regulatory function to different physiological situations during an immune response. However, plasticity of Tregs might also result in conversion into effector T cells that may contribute to autoimmune pathogenesis. Yet, which environmental cues regulate Treg plasticity and function is currently poorly understood, but it is of significant importance for therapeutic purposes. Here we review the current understanding on the effect of certain dietary nutrients that characterize Western diets in Treg metabolism, stability, and function. Moreover, we will discuss the role of Tregs linking diet and autoimmunity and the potential of dietary-based interventions to modulate Treg function in disease.
Collapse
Affiliation(s)
- Rebeca Arroyo Hornero
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Ibrahim Hamad
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Beatriz Côrte-Real
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), University of Hasselt, Hasselt, Belgium
| |
Collapse
|
198
|
Colamatteo A, Carbone F, Bruzzaniti S, Galgani M, Fusco C, Maniscalco GT, Di Rella F, de Candia P, De Rosa V. Molecular Mechanisms Controlling Foxp3 Expression in Health and Autoimmunity: From Epigenetic to Post-translational Regulation. Front Immunol 2020; 10:3136. [PMID: 32117202 PMCID: PMC7008726 DOI: 10.3389/fimmu.2019.03136] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
The discovery of the transcription factor Forkhead box-p3 (Foxp3) has shed fundamental insights into the understanding of the molecular determinants leading to generation and maintenance of T regulatory (Treg) cells, a cell population with a key immunoregulatory role. Work over the past few years has shown that fine-tuned transcriptional and epigenetic events are required to ensure stable expression of Foxp3 in Treg cells. The equilibrium between phenotypic plasticity and stability of Treg cells is controlled at the molecular level by networks of transcription factors that bind regulatory sequences, such as enhancers and promoters, to regulate Foxp3 expression. Recent reports have suggested that specific modifications of DNA and histones are required for the establishment of the chromatin structure in conventional CD4+ T (Tconv) cells for their future differentiation into the Treg cell lineage. In this review, we discuss the molecular events that control Foxp3 gene expression and address the associated alterations observed in human diseases. Also, we explore how Foxp3 influences the gene expression programs in Treg cells and how unique properties of Treg cell subsets are defined by other transcription factors.
Collapse
Affiliation(s)
- Alessandra Colamatteo
- Treg Cell Laboratory, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Fortunata Carbone
- Laboratorio di Immunologia, Istituto per L'Endocrinologia e L'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), Naples, Italy.,Unità di NeuroImmunologia, Fondazione Santa Lucia, Rome, Italy
| | - Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per L'Endocrinologia e L'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), Naples, Italy.,Dipartimento di Biologia, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Mario Galgani
- Treg Cell Laboratory, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy.,Laboratorio di Immunologia, Istituto per L'Endocrinologia e L'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), Naples, Italy
| | - Clorinda Fusco
- Treg Cell Laboratory, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Giorgia Teresa Maniscalco
- Dipartimento di Neurologia, Centro Regionale Sclerosi Multipla, Azienda Ospedaliera "A. Cardarelli", Naples, Italy
| | - Francesca Di Rella
- Clinical and Experimental Senology, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | | | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto per L'Endocrinologia e L'Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche (IEOS-CNR), Naples, Italy.,Unità di NeuroImmunologia, Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
199
|
Cavallo S. Immune-mediated genesis of multiple sclerosis. J Transl Autoimmun 2020; 3:100039. [PMID: 32743522 PMCID: PMC7388381 DOI: 10.1016/j.jtauto.2020.100039] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is widely acknowledged to be an autoimmune disease affecting the neuronal myelin structure of the CNS. Autoantigens recognized as the target of this autoimmune process are: myelin basal protein, anti-proteolipid protein, antimyelin-associated glycoprotein and antimyelin-based oligodendrocytic basic protein. Ample evidence supports the idea of a dysregulation of immunological tolerance towards self-antigens of neuronal myelin structure triggered by one or more viral or bacterial microbial agents in predisposed HLA gene subjects. Genetic predisposition to MS has been highlighted by numerous studies associating the disease to specific HLA haplotypes. Moreover, a wide range of evidence supports the fact that MS may be consequence of one or more viral or bacterial infections such as measles virus, EBV, HHV6, HZV, Chlamydia pneumoniae, Helicobacter Pylori, and other microbial agents. Microbiota elements also seems to have a role on the determinism of the disease as a pathogenic or protective factor. The autoimmune pathogenetic process could arise when a molecular mimicry between a foreign microbial antigen and an auto-antigen occurs in an HLA gene subject competent for that particular antigen. The antigen-presenting cells in this case would induce the activation of a specific Th clone causing a cross-reaction between a foreign antigen and an autoantigen resulting in an autoimmune response. A multifactorial ethiopathogenetic model based on immunomediation is a reliable hypothesis for multiple sclerosis. Evidence found in the scientific literature makes it possible to reconstruct this etiopathogenetic hypothesis for MS. HLA gene predisposition, correlation with infections, molecular mimicry and other immunological data are reported.
Collapse
Affiliation(s)
- Salvatore Cavallo
- Expert Doctor in Non-Conventional Medicine, Professor and Member of the Board of the MMS, MMS (Medicina di Modulazione Dei Sistemi) Roma, Salvatore Cavallo Via G.B. Pergolesi, 28, 75100, Matera, Italy
| |
Collapse
|
200
|
Pape K, Tamouza R, Leboyer M, Zipp F. Immunoneuropsychiatry - novel perspectives on brain disorders. Nat Rev Neurol 2020; 15:317-328. [PMID: 30988501 DOI: 10.1038/s41582-019-0174-4] [Citation(s) in RCA: 281] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Immune processes have a vital role in CNS homeostasis, resilience and brain reserve. Our cognitive and social abilities rely on a highly sensitive and fine-tuned equilibrium of immune responses that involve both innate and adaptive immunity. Autoimmunity, chronic inflammation, infection and psychosocial stress can tip the scales towards disruption of higher-order networks. However, not only classical neuroinflammatory diseases, such as multiple sclerosis and autoimmune encephalitis, are caused by immune dysregulation that affects CNS function. Recent insight indicates that similar processes are involved in psychiatric diseases such as schizophrenia, autism spectrum disorder, bipolar disorder and depression. Pathways that are common to these disorders include microglial activation, pro-inflammatory cytokines, molecular mimicry, anti-neuronal autoantibodies, self-reactive T cells and disturbance of the blood-brain barrier. These discoveries challenge our traditional classification of neurological and psychiatric diseases. New clinical paths are required to identify subgroups of neuropsychiatric disorders that are phenotypically distinct but pathogenically related and to pave the way for mechanism-based immune treatments. Combined expertise from neurologists and psychiatrists will foster translation of these paths into clinical practice. The aim of this Review is to highlight outstanding findings that have transformed our understanding of neuropsychiatric diseases and to suggest new diagnostic and therapeutic criteria for the emerging field of immunoneuropsychiatry.
Collapse
Affiliation(s)
- Katrin Pape
- Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ryad Tamouza
- Inserm, U955, Institut Mondor de la Recherche Biomédicale, Créteil, France.,Fondation FondaMental, Créteil, France.,AP-HP, Department of Psychiatry of Mondor University Hospital, DHU PePsy, University of Paris-Est-Créteil, Créteil, France
| | - Marion Leboyer
- Inserm, U955, Institut Mondor de la Recherche Biomédicale, Créteil, France.,Fondation FondaMental, Créteil, France.,AP-HP, Department of Psychiatry of Mondor University Hospital, DHU PePsy, University of Paris-Est-Créteil, Créteil, France
| | - Frauke Zipp
- Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|