1
|
Gao W, Kim MW, Dykstra T, Du S, Boskovic P, Lichti CF, Ruiz-Cardozo MA, Gu X, Weizman Shapira T, Rustenhoven J, Molina C, Smirnov I, Merbl Y, Ray WZ, Kipnis J. Engineered T cell therapy for central nervous system injury. Nature 2024; 634:693-701. [PMID: 39232158 DOI: 10.1038/s41586-024-07906-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 08/05/2024] [Indexed: 09/06/2024]
Abstract
Traumatic injuries to the central nervous system (CNS) afflict millions of individuals worldwide1, yet an effective treatment remains elusive. Following such injuries, the site is populated by a multitude of peripheral immune cells, including T cells, but a comprehensive understanding of the roles and antigen specificity of these endogenous T cells at the injury site has been lacking. This gap has impeded the development of immune-mediated cellular therapies for CNS injuries. Here, using single-cell RNA sequencing, we demonstrated the clonal expansion of mouse and human spinal cord injury-associated T cells and identified that CD4+ T cell clones in mice exhibit antigen specificity towards self-peptides of myelin and neuronal proteins. Leveraging mRNA-based T cell receptor (TCR) reconstitution, a strategy aimed to minimize potential adverse effects from prolonged activation of self-reactive T cells, we generated engineered transiently autoimmune T cells. These cells demonstrated notable neuroprotective efficacy in CNS injury models, in part by modulating myeloid cells via IFNγ. Our findings elucidate mechanistic insight underlying the neuroprotective function of injury-responsive T cells and pave the way for the future development of T cell therapies for CNS injuries.
Collapse
Affiliation(s)
- Wenqing Gao
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
| | - Min Woo Kim
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Immunology Program, School of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Medical Scientist Training Program, School of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Taitea Dykstra
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Siling Du
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Immunology Program, School of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Pavle Boskovic
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Miguel A Ruiz-Cardozo
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Xingxing Gu
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Tal Weizman Shapira
- Systems Immunology Department, The Weizmann Institute of Science, Rehovot, Israel
| | - Justin Rustenhoven
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Camilo Molina
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Igor Smirnov
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Yifat Merbl
- Systems Immunology Department, The Weizmann Institute of Science, Rehovot, Israel
| | - Wilson Z Ray
- Department of Neurological Surgery, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
- Immunology Program, School of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
- Medical Scientist Training Program, School of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
2
|
Liu S, Yang X, Zhao H, Zhao X, Fan K, Liu G, Li X, Du C, Liu J, Ma J. Cathepsin C exacerbates EAE by promoting the expansion of Tfh cells and the formation of TLSs in the CNS. Brain Behav Immun 2024; 123:123-142. [PMID: 39243987 DOI: 10.1016/j.bbi.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/05/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) mediated by CD4+ T helper (Th) cells, and characterized by immune cell infiltration, demyelination and neurodegeneration, with no definitive cure available. Thus, it is pivotal and imperative to acquire more profound comprehension of the underlying mechanisms implicated in MS. Dysregulated immune responses are widely believed to play a primary role in the pathogenesis of MS. Recently, a plethora of studies have demonstrated the involvement of T follicular helper (Tfh) cells and tertiary lymphoid-like structures (TLSs) in the pathogenesis and progression of MS. Cathepsin C (CatC) is a cysteine exopeptidase which is crucial for the activation of immune-cell-associated serine proteinases in many inflammatory diseases in peripheral system, such as rheumatoid arthritis and septicemia. We have previously demonstrated that CatC is involved in neuroinflammation and exacerbates demyelination in both cuprizone-induced and experimental autoimmune encephalomyelitis (EAE) mouse models. However, the underlying immunopathological mechanism remains elusive. In the present study, we established a recombinant myelin oligodendrocyte glycoprotein 35-55 peptide-induced EAE model using conditional CatC overexpression mice to investigate the effects of CatC on the alteration of CD4+ Th subsets, including Th1, Th2, Th17, Tfh and T regulatory cells. Our findings demonstrated that CatC particularly enhanced the population of Tfh cell in the brain, resulting in the earlier onset and more severe chronic syndrome of EAE. Furthermore, CatC promoted the formation of TLSs in the brain, leading to persistent neuroinflammation and exacerbating the severity of EAE in the chronic phase. Conversely, treatment with AZD7986, a specific inhibitor of CatC, effectively attenuated the syndrome of EAE and its effects caused by CatC both in vivo and in vitro. These findings provide a novel insight into the critical role of CatC in innate and adaptive immunity in EAE, and specific inhibitor of CatC, AZD7986, may contribute to potential therapeutic strategies for MS.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xiaohan Yang
- Department of Morphology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Henan Zhao
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xinnan Zhao
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Kai Fan
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Gang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Cong Du
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jianmei Ma
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China.
| |
Collapse
|
3
|
Kveštak D, Mihalić A, Jonjić S, Brizić I. Innate lymphoid cells in neuroinflammation. Front Cell Neurosci 2024; 18:1364485. [PMID: 38450285 PMCID: PMC10915051 DOI: 10.3389/fncel.2024.1364485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
Innate lymphoid cells (ILCs) are largely tissue-resident cells that participate in the maintenance of tissue homeostasis and react early to inflammatory events. Mature ILCs are divided into three major groups based on the transcription factors required for their development and function. Under physiological conditions, ILCs are present within the choroid plexus and meninges while the CNS parenchyma is almost devoid of these cells. However, pathological conditions such as autoimmune neuroinflammation and viral infections of the CNS result in the infiltration of ILCs into parenchyma. In this article, we provide an overview of the involvement and function of the ILCs within the CNS during physiological conditions and in infections, autoimmune diseases, neurodegeneration, and injury.
Collapse
Affiliation(s)
- Daria Kveštak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Andrea Mihalić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Department of Biomedical Sciences, Croatian Academy of Sciences and Arts, Rijeka, Croatia
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
4
|
Shulman Y, Finkelstein L, Levi Y, Kovalchuk D, Weksler A, Reichstein A, Kigel-Tsur K, Davidi M, Levi I, Schauder A, Rubin K, Achituv E, Castel D, Meilin S. A Novel Sensory Wave (P25) in Myelin Oligodendrocyte Glycoprotein-induced Experimental Autoimmune Encephalomyelitis Murine Model. THE JOURNAL OF PAIN 2024; 25:73-87. [PMID: 37524220 DOI: 10.1016/j.jpain.2023.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 07/09/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023]
Abstract
Myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) is a murine model for multiple sclerosis. This model is characterized by chronic and progressive demyelination, leading to impairment of motor function and paralysis. While the outcomes of the disease, including impaired motor function and immunological changes, are well-characterized, little is known about the impact of EAE on the electrophysiology of the motor and sensory systems. In this study, we assessed evoked potentials as a quantitative marker for in vivo monitoring of nervous system damage. Motor-evoked potentials (MEPs) and sensory-evoked potentials (SEPs) were first standardized in naïve C57BL mice and studied thoroughly in EAE mice. The duration of MEPs and the number of connotative potentials increased significantly alongside an increase in temporal SEP amplitudes. Moreover, a new SEP wave was identified in naïve animals, which significantly increased in MOG-induced EAE animals with no or mild symptoms (clinical score 0-2, 0-5 scale). This wave occurred ∼25 milliseconds poststimulation, thus named p25. P25 was correlated with increased vocalization and was also reduced in amplitude following treatment with morphine. As the EAE score progressed (clinical score 3-4, 0-5 scale), the amplitude of MEPs and SEPs decreased drastically. Our results demonstrate that desynchronized neural motor activity, along with hypersensitivity in the early stages of EAE, leads to a complete loss of motor and sensory functions in the late stages of the disease. The findings also suggest an increase in p25 amplitude before motor deficits appear, indicating SEP as a predictive marker for disease progression. PERSPECTIVE: This article assesses p25, a new sensory electrophysiology wave that correlates with pain-related behavior in MOG-induced EAE mice and appears prior to the clinical symptoms. Motor electrophysiology correlates with traditional motor behavior scoring and histology.
Collapse
Affiliation(s)
- Yoav Shulman
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - Lena Finkelstein
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - Yakir Levi
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | | | - Ayelet Weksler
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | | | - Keren Kigel-Tsur
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - Mazal Davidi
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - Isaac Levi
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - Avital Schauder
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - Keren Rubin
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - Elhanan Achituv
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| | - David Castel
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel; The Neufeld Cardiac Research Institute and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sigal Meilin
- Neurology Division, MD Biosciences Innovalora, Ltd, Rehovot, Israel
| |
Collapse
|
5
|
Karaca E, Yarim M. Naringenin stimulates aromatase expression and alleviates the clinical and histopathological findings of experimental autoimmune encephalomyelitis in C57bl6 mice. Histochem Cell Biol 2023; 160:477-490. [PMID: 37378907 DOI: 10.1007/s00418-023-02217-1] [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] [Accepted: 06/02/2023] [Indexed: 06/29/2023]
Abstract
This study was conducted to demonstrate the possible protective and therapeutic effects of naringenin, an estrogenically effective flavonoid, in experimental autoimmune encephalomyelitis (EAE), which is the rodent model of multiple sclerosis. For this purpose, 50 12-week-old C57BL6 male mice were divided into five groups; control, naringenin, EAE, prophylactic naringenin + EAE, and EAE + therapeutic naringenin. The EAE model was induced with myelin oligodendrocyte glycoprotein(35-55), and naringenin (50 mg/kg) was administered by oral gavage. The prophylactic and therapeutic effects of naringenin were examined according to clinical, histopathological, immunohistochemical, electron microscopic, and RT-PCR (aromatase, 3βHSD, estrogen receptors, and progesterone receptor expression) parameters. The acute EAE model was successfully induced, along with its clinical and histopathological findings. RT-PCR showed that expression of aromatase, 3βHSD, estrogen receptor-β, and progesterone receptor gene decreased, while estrogen receptor-α increased after EAE induction. Electron microscopic analysis showed mitochondrial damage and degenerative changes in myelinated axons and neurons in EAE, which could be behind the downregulation in the expressions of neurosteroid enzymes. Aromatase immunopositivity rates also decreased in EAE, while estrogen receptor α and β, and progesterone receptor immunopositivity rates increased. Naringenin improved aromatase immunopositivity rates and gene expression in both prophylactic and therapeutic use. Clinical and histopathological findings revealed that EAE findings were alleviated in both prophylactic and therapeutic groups, along with significantly decreased inflammatory cell infiltrations in the white matter of the spinal cords. In conclusion, naringenin could provide long-term beneficial effects even in prophylactic use due to stimulating aromatase expression, but it could not prevent or eliminate the EAE model's lesions completely.
Collapse
Affiliation(s)
- Efe Karaca
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, 55200, Atakum, Samsun, Turkey.
| | - Murat Yarim
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Ondokuz Mayıs University, 55200, Atakum, Samsun, Turkey
| |
Collapse
|
6
|
Castro-Martínez G, Herrera-Ruiz M, González-Cortázar M, Porras-Dávila SL, Almanza Pérez JC, Jimenez-Ferrer E. Effects of Five Coumarins and Standardized Extracts from Tagetes lucida Cav. on Motor Impairment and Neuroinflammation Induced with Cuprizone. Pharmaceuticals (Basel) 2023; 16:1391. [PMID: 37895861 PMCID: PMC10610053 DOI: 10.3390/ph16101391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) with no curative treatment, and the available therapies aim to modify the course of the disease. It has been demonstrated that extracts of Tagetes lucida have immunomodulatory and neuroprotective effects. This work induced motor damage and neuroinflammation in male BALB/c mice by oral administration of cuprizone (CPZ) (40 mg/kg) for five weeks. In addition, the extracts and coumarins of Tagetes lucida (25 mg/kg) were used to control these damage variables; during the experiment, animals were subject to behavioral tests, and at the end of 5 weeks, mice from each group were used to measure the integrity of biological barriers (brain, kidneys, and spleen) through the extravasation test with blue Evans dye. In another group of animals, the ELISA method measured the brain concentrations of IL-1β, IL-4, IL-10, and TNF-α. The results presented here allow us to conclude that the extracts and coumarins IC, HN, PE, DF, and SC of Tagetes lucida exert a neuroprotective effect by controlling the motor damage and neuroinflammation by increasing the expression of IL-4 and IL-10 and decreasing IL-1β and TNF-α; notably, these treatments also protect organs from vascular permeability increase, mainly the BBB, in mice with CPZ-induced experimental encephalomyelitis (VEH * p < 0.05). However, more studies must be carried out to elucidate the molecular mechanisms of the pharmacological effects of this Mexican medicinal plant.
Collapse
Affiliation(s)
- Gabriela Castro-Martínez
- Doctorate in Biological and Health Sciences, Metropolitan Autonomous University, Mexico City 14387, Mexico;
- Southern Biomedical Research Center, Mexican Social Security Institute, Guadalajara 44340, Mexico; (M.H.-R.); (M.G.-C.); (S.L.P.-D.)
| | - Maribel Herrera-Ruiz
- Southern Biomedical Research Center, Mexican Social Security Institute, Guadalajara 44340, Mexico; (M.H.-R.); (M.G.-C.); (S.L.P.-D.)
| | - Manases González-Cortázar
- Southern Biomedical Research Center, Mexican Social Security Institute, Guadalajara 44340, Mexico; (M.H.-R.); (M.G.-C.); (S.L.P.-D.)
| | - Sandra Liliana Porras-Dávila
- Southern Biomedical Research Center, Mexican Social Security Institute, Guadalajara 44340, Mexico; (M.H.-R.); (M.G.-C.); (S.L.P.-D.)
| | - Julio Cesar Almanza Pérez
- Department of Health Sciences, Division of Biological and Health Sciences, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
| | - Enrique Jimenez-Ferrer
- Southern Biomedical Research Center, Mexican Social Security Institute, Guadalajara 44340, Mexico; (M.H.-R.); (M.G.-C.); (S.L.P.-D.)
| |
Collapse
|
7
|
Sadek MA, Kandil EA, El Sayed NS, Sayed HM, Rabie MA. Semaglutide, a novel glucagon-like peptide-1 agonist, amends experimental autoimmune encephalomyelitis-induced multiple sclerosis in mice: Involvement of the PI3K/Akt/GSK-3β pathway. Int Immunopharmacol 2023; 115:109647. [PMID: 36584570 DOI: 10.1016/j.intimp.2022.109647] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/17/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis (MS) is a disabling neurodegenerative disease that causes demyelination and axonal degeneration of the central nervous system. Current treatments are partially effective in managing MS relapses and have a negligible impact on treating MS cognitive deficits and cannot enhance neuronal remyelination, imposing a need for a new MS remedy. Semaglutide, a novel glucagon-like peptide-1 agonist, has recently displayed a neuroprotective effect on several neurodegenerative diseases, suggesting that it may have a protective effect in MS. Therefore, this study was conducted to investigate the influence of semaglutide on experimental autoimmune encephalomyelitis (EAE)-induced MS in mice. Here, EAE was induced in mice using spinal cord homogenate, which eventually altered the mice's cognitive and motor functions, similar to what is observed in MS. Interestingly, intraperitoneally administered semaglutide (25 nmol/kg/day) amended EAE-induced cognitive and motor deficits observed in novel object recognition, open field, rotarod, and grip strength tests. Moreover, histological examination revealed that semaglutide treatment attenuated hippocampal damage and corpus callosum demyelination caused by EAE. Additionally, biochemical testing revealed that semaglutide activates the PI3K/Akt axis, which eventually hampers GSK-3β activity. GSK-3β activity inhibition attenuates demyelination and triggers remyelination through CREB/BDNF; furthermore, it boosts Nrf2 and SOD levels, protecting the mice from EAE-induced oxidative stress. Additionally, GSK-3β inhibition minimizes neuroinflammation, as reflected by decreased NF-kβ and TNF-α levels. In conclusion, semaglutide has a neuroprotective effect in EAE-induced MS in mice, which is mediated by activating the ramified PI3K/Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Mohamed A Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Helmy M Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
8
|
Casey LM, Decker JT, Podojil JR, Rad L, Hughes KR, Rose JA, Pearson RM, Miller SD, Shea LD. Nanoparticle dose and antigen loading attenuate antigen-specific T-cell responses. Biotechnol Bioeng 2023; 120:284-296. [PMID: 36221192 PMCID: PMC9999438 DOI: 10.1002/bit.28252] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022]
Abstract
Immune-mediated hypersensitivities such as autoimmunity, allergy, and allogeneic graft rejection are treated with therapeutics that suppress the immune system, and the lack of specificity is associated with significant side effects. The delivery of disease-relevant antigens (Ags) by carrier systems such as poly(lactide-co-glycolide) nanoparticles (PLG-Ag) and carbodiimide (ECDI)-fixed splenocytes (SP-Ag) has demonstrated Ag-specific tolerance induction in model systems of these diseases. Despite therapeutic outcomes by both platforms, tolerance is conferred with different efficacy. This investigation evaluated Ag loading and total particle dose of PLG-Ag on Ag presentation in a coculture system of dendritic cells (DCs) and Ag-restricted T cells, with SP-Ag employed as a control. CD25 expression was observed in nearly all T cells even at low concentrations of PLG-Ag, indicating efficient presentation of Ag by dendritic cells. However, the secretion of IL-2, Th1, and Th2 cytokines (IFNγ and IL-4, respectively) varied depending on PLG-Ag concentration and Ag loading. Concentration escalation of soluble Ag resulted in an increase in IL-2 and IFNγ and a decrease in IL-4. Treatment with PLG-Ag followed a similar trend but with lower levels of IL-2 and IFNγ secreted. Transcriptional Activity CEll ARrays (TRACER) were employed to measure the real-time transcription factor (TF) activity in Ag-presenting DCs. The kinetics and magnitude of TF activity was dependent on the Ag delivery method, concentration, and Ag loading. Ag positively regulated IRF1 activity and, as carriers, NPs and ECDI-treated SP negatively regulated this signaling. The effect of Ag loading and dose on tolerance induction were corroborated in vivo using the delayed-type hypersensitivity (DTH) and experimental autoimmune encephalomyelitis (EAE) mouse models where a threshold of 8 μg/mg Ag loading and 0.5 mg PLG-Ag dose were required for tolerance. Together, the effect of Ag loading and dosing on in vitro and in vivo immune regulation provide useful insights for translating Ag-carrier systems for the clinical treatment of immune disorders.
Collapse
Affiliation(s)
- Liam M. Casey
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Joseph T. Decker
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Joseph R. Podojil
- Department of Microbiology‐Immunology, Feinberg School of MedicineNorthwestern UniversityChicagollinoisUSA
| | - Laila Rad
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Kevin R. Hughes
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Justin A. Rose
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Ryan M. Pearson
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Stephen D. Miller
- Department of Microbiology‐Immunology, Feinberg School of MedicineNorthwestern UniversityChicagollinoisUSA
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology Center, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Lonnie D. Shea
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
9
|
Tredicine M, Ria F, Poerio N, Lucchini M, Bianco A, De Santis F, Valentini M, De Arcangelis V, Rende M, Stabile AM, Pistilli A, Camponeschi C, Nociti V, Mirabella M, Fraziano M, Di Sante G. Liposome-based nanoparticles impact on regulatory and effector phenotypes of macrophages and T cells in multiple Sclerosis patients. Biomaterials 2023; 292:121930. [PMID: 36493716 DOI: 10.1016/j.biomaterials.2022.121930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
Current available treatments of Multiple Sclerosis (MS) reduce neuroinflammation acting on different targets on the immune system, but potentially lead to severe side effects and have a limited efficacy in slowing the progression of the disease. Here, we evaluated in vitro the immunomodulatory potential of a new class of nanoparticles - liposomes, constituted by a double-layer of phosphatidylserine (PSCho/PS), and double-faced, with an outer layer of phosphatidylserine and an inner layer of phosphatidic acid (PSCho/PA), either alone or in the presence of the myelin basic protein (MBP) peptide (residues 85-99) (PSCho/PS-MBP and PSCho/PA-MBP). Results showed that PSCho/PS are equally and efficiently internalized by pro- and anti-inflammatory macrophages (M1 and M2 respectively), while PSCho/PA were internalized better by M2 than M1. PSCho/PS liposomes were able to inhibit the secretion of innate pro-inflammatory cytokine IL-1β. PSCho/PS liposomes expanded Tregs, reducing Th1 and Th17 cells, while PSCho/PA liposomes were unable to dampen pro-inflammatory T cells and to promote immune-regulatory phenotype (Treg). The ability of PSCho/PS liposomes to up-regulate Treg cells was more pronounced in MS patients with high basal expression of M2 markers. PSCho/PS liposomes were more effective in decreasing Th1 (but not Th17) cells in MS patients with a disease duration >3 months. On the other hand, down-modulation of Th17 cells was evident in MS patients with active, Gadolinium enhancing lesions at MRI and in MS patients with a high basal expression of M1-associated markers in the monocytes. The same findings were observed for the modulation of MBP-driven Th1/Th17/Treg responses. These observations suggest that early MS associate to a hard-wired pro-Th1 phenotype of M1 that is lost later during disease course. On the other hand, acute inflammatory events reflect a temporary decrease of M2 phenotype that however is amenable to restauration upon treatment with PSCho/PS liposomes. Thus, together these data indicate that monocytes/macrophages may play an important regulatory function during MS course and suggest a role for PSCho/PS and PSCho/PS-MBP as new therapeutic tools to dampen the pro-inflammatory immune responses and to promote its regulatory branch.
Collapse
Affiliation(s)
- Maria Tredicine
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Francesco Ria
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Department Laboratory and Infectious diseases Sciences, Largo Agostino Gemelli 1-8, 00168, Rome, Italy.
| | - Noemi Poerio
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Matteo Lucchini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Assunta Bianco
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Federica De Santis
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Mariagrazia Valentini
- Section of Pathology, Department of Woman, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 1-8, 00168, Rome, Italy.
| | - Valeria De Arcangelis
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Mario Rende
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Anna Maria Stabile
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Alessandra Pistilli
- Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| | - Chiara Camponeschi
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Institute of Chemical Sciences and Technologies ''Giulio Natta'' (SCITEC)-CNR, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Viviana Nociti
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Massimiliano Mirabella
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOC of Neurology, Largo Agostino Gemelli 8, 00168, Rome, Italy; Department of Neurosciences, Centro di Ricerca Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Maurizio Fraziano
- Department of Biology, University of Rome "TorVergata", Via della Ricerca Scientifica 1, 00173, Rome, Italy.
| | - Gabriele Di Sante
- Section of General Pathology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Department of Surgery and Medicine, Institute of Human, Clinical and Forensic Anatomy, Piazza L. Severi 1, 06125, Perugia, Italy.
| |
Collapse
|
10
|
Robles LM, Reichenberg LH, Grissom Ⅲ JH, Chi RJ, Piller KJ. Recombinant MBP-pσ1 expressed in soybean seeds delays onset and reduces developing disease in an animal model of multiple sclerosis. PLANT BIOTECHNOLOGY (TOKYO, JAPAN) 2022; 39:367-379. [PMID: 37283612 PMCID: PMC10240915 DOI: 10.5511/plantbiotechnology.22.0926a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/26/2022] [Indexed: 06/08/2023]
Abstract
It is estimated that multiple sclerosis (MS) affects over 2.8 million people worldwide, with a prevalence that is expected to continue growing over time. Unfortunately, there is no cure for this autoimmune disease. For several decades, antigen-specific treatments have been used in animal models of experimental autoimmune encephalomyelitis (EAE) to demonstrate their potential for suppressing autoimmune responses. Successes with preventing and limiting ongoing MS disease have been documented using a wide variety of myelin proteins, peptides, autoantigen-conjugates, and mimics when administered by various routes. While those successes were not translatable in the clinic, we have learned a great deal about the roadblocks and hurdles that must be addressed if such therapies are to be useful. Reovirus sigma1 protein (pσ1) is an attachment protein that allows the virus to target M cells with high affinity. Previous studies showed that autoantigens tethered to pσ1 delivered potent tolerogenic signals and diminished autoimmunity following therapeutic intervention. In this proof-of-concept study, we expressed a model multi-epitope autoantigen (human myelin basic protein, MBP) fused to pσ1 in soybean seeds. The expression of chimeric MBP-pσ1 was stable over multiple generations and formed the necessary multimeric structures required for binding to target cells. When administered to SJL mice prophylactically as an oral therapeutic, soymilk formulations containing MBP-pσ1 delayed the onset of clinical EAE and significantly reduced developing disease. These results demonstrate the practicality of soybean as a host for producing and formulating immune-modulating therapies to treat autoimmune diseases.
Collapse
Affiliation(s)
| | | | - James H. Grissom Ⅲ
- University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA
| | - Richard J. Chi
- University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA
| | - Kenneth J. Piller
- SoyMeds, Inc., Charlotte, North Carolina 28223, USA
- University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA
| |
Collapse
|
11
|
Tolmacheva AS, Aulova KS, Urusov AE, Doronin VB, Nevinsky GA. Antibodies-Abzymes with Antioxidant Activities in Two Th and 2D2 Experimental Autoimmune Encephalomyelitis Mice during the Development of EAE Pathology. Molecules 2022; 27:7527. [PMID: 36364362 PMCID: PMC9656754 DOI: 10.3390/molecules27217527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 08/30/2023] Open
Abstract
The exact mechanisms of multiple sclerosis development are still unknown. However, the development of EAE (experimental autoimmune encephalomyelitis) in Th and 2D2 mice is associated with the infringement of the differentiation profiles of bone marrow hematopoietic stem cells which are bound with the production of compounds that are harmful for human autoantibodies-abzymes that hydrolyze myelin oligodendrocyte glycoprotein, myelin basic protein, and DNA. It showed that autoimmune patients' antioxidant IgG antibodies oxidise some compounds due to their peroxidase (H2O2-dependent) and oxidoreductase (H2O2-independent) activities more effectively than those in healthy humans can. It was interesting to identify whether the redox activities of the antibodies change during the development of autoimmune diseases. Here, we analyzed the change in these redox activities of the IgGs from the blood of Th and 2D2 mice, which corresponded to different stages of the EAE development. The peroxidase activity in the oxidation of ABTS (2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)) in the Th (4-fold) and 2D2 (2-fold) mice IgGs, on average, is higher than the oxidoreductase activity is. The peroxidase activity of the Th (1.9-fold) and 2D2 (3.5-fold) mice IgGs remarkably increased during the 40 days of the spontaneous development of EAE. Forty days after the immunization of the MOG peroxidase activity, the IgGs of the Th and 2D2 mice increased 5.6-6.0 times when they were compared with those that presented no increase (3 months of age). The mice IgGs were oxidized with 3,3'-diaminobenzidine (2.4-4.3-fold) and o-phenylenediamine (139-143-fold) less efficiently than they were with ABTS. However, the temper of the change in the IgG activity in the oxidation of these substrates during the spontaneous and MOG-induced development of EAE was close to that which occurred for ABTS. All of the data show that the IgG peroxidase and oxidoreductase activities of EAE mice can play an important role in their protection from toxic compounds and oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | - Georgy A. Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
12
|
Postoak JL, Song W, Yang G, Guo X, Xiao S, Saffold CE, Zhang J, Joyce S, Manley NR, Wu L, Van Kaer L. Thymic epithelial cells require lipid kinase Vps34 for CD4 but not CD8 T cell selection. J Exp Med 2022; 219:e20212554. [PMID: 35997680 PMCID: PMC9402993 DOI: 10.1084/jem.20212554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 06/22/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022] Open
Abstract
The generation of a functional, self-tolerant T cell receptor (TCR) repertoire depends on interactions between developing thymocytes and antigen-presenting thymic epithelial cells (TECs). Cortical TECs (cTECs) rely on unique antigen-processing machinery to generate self-peptides specialized for T cell positive selection. In our current study, we focus on the lipid kinase Vps34, which has been implicated in autophagy and endocytic vesicle trafficking. We show that loss of Vps34 in TECs causes profound defects in the positive selection of the CD4 T cell lineage but not the CD8 T cell lineage. Utilizing TCR sequencing, we show that T cell selection in conditional mutants causes altered repertoire properties including reduced clonal sharing. cTECs from mutant mice display an increased abundance of invariant chain intermediates bound to surface MHC class II molecules, indicating altered antigen processing. Collectively, these studies identify lipid kinase Vps34 as an important contributor to the repertoire of selecting ligands processed and presented by TECs to developing CD4 T cells.
Collapse
Affiliation(s)
- J. Luke Postoak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Wenqiang Song
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Guan Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Xingyi Guo
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN
| | - Shiyun Xiao
- Department of Genetics, University of Georgia, Athens, GA
| | - Cherie E. Saffold
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
- Birmingham Veterans Affairs Medical Center, Birmingham, AL
| | - Sebastian Joyce
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| | | | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
13
|
Zhang M, Guo FR. BSDE: barycenter single-cell differential expression for case-control studies. Bioinformatics 2022; 38:2765-2772. [PMID: 35561165 PMCID: PMC9113363 DOI: 10.1093/bioinformatics/btac171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Single-cell sequencing brings about a revolutionarily high resolution for finding differentially expressed genes (DEGs) by disentangling highly heterogeneous cell tissues. Yet, such analysis is so far mostly focused on comparing between different cell types from the same individual. As single-cell sequencing becomes cheaper and easier to use, an increasing number of datasets from case-control studies are becoming available, which call for new methods for identifying differential expressions between case and control individuals. RESULTS To bridge this gap, we propose barycenter single-cell differential expression (BSDE), a nonparametric method for finding DEGs for case-control studies. Through the use of optimal transportation for aggregating distributions and computing their distances, our method overcomes the restrictive parametric assumptions imposed by standard mixed-effect-modeling approaches. Through simulations, we show that BSDE can accurately detect a variety of differential expressions while maintaining the type-I error at a prescribed level. Further, 1345 and 1568 cell type-specific DEGs are identified by BSDE from datasets on pulmonary fibrosis and multiple sclerosis, among which the top findings are supported by previous results from the literature. AVAILABILITY AND IMPLEMENTATION R package BSDE is freely available from doi.org/10.5281/zenodo.6332254. For real data analysis with the R package, see doi.org/10.5281/zenodo.6332566. These can also be accessed thorough GitHub at github.com/mqzhanglab/BSDE and github.com/mqzhanglab/BSDE_pipeline. The two single-cell sequencing datasets can be download with UCSC cell browser from cells.ucsc.edu/?ds=ms and cells.ucsc.edu/?ds=lung-pf-control. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mengqi Zhang
- Department of Surgery, Perelman Medical School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
14
|
Essential Protective Role of Catalytically Active Antibodies (Abzymes) with Redox Antioxidant Functions in Animals and Humans. Int J Mol Sci 2022; 23:ijms23073898. [PMID: 35409256 PMCID: PMC8999700 DOI: 10.3390/ijms23073898] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 12/13/2022] Open
Abstract
During the life of aerobic organisms, the oxygen resulting from numerous reactions is converted into reactive oxygen species (ROS). Many ROS are dangerous due to their high reactivity; they are strong oxidants, and react with various cell components, leading to their damage. To protect against ROS overproduction, enzymatic and non-enzymatic systems are evolved in aerobic cells. Several known non-enzymatic antioxidants have a relatively low specific antioxidant activity. Superoxide dismutases, catalase, glutathione peroxidase, glutathione S-transferase, thioredoxin, and the peroxiredoxin families are the most important enzyme antioxidants. Artificial antibodies catalyzing redox reactions using different approaches have been created. During the past several decades, it has been shown that the blood and various biological fluids of humans and animals contain natural antibodies that catalyze different redox reactions, such as classical enzymes. This review, for the first time, summarizes data on existing non-enzymatic antioxidants, canonical enzymes, and artificial or natural antibodies (abzymes) with redox functions. Comparing abzymes with superoxide dismutase, catalase, peroxide-dependent peroxidase, and H2O2-independent oxidoreductase activities with the same activities as classical enzymes was carried out. The features of abzymes with the redox activities are described, including their exceptional diversity in the optimal pH values, dependency and independence on various metal ions, and the reaction rate constants for healthy donors and patients with different autoimmune diseases. The entire body of evidence indicates that abzymes with redox antioxidant activities existing in the blood for a long time compared to enzymes are an essential part of the protection system of humans and animals from oxidative stress.
Collapse
|
15
|
Cell Differentiation and Proliferation in the Bone Marrow and Other Organs of 2D2 Mice during Spontaneous Development of EAE Leading to the Production of Abzymes. Molecules 2022; 27:molecules27072195. [PMID: 35408594 PMCID: PMC9000721 DOI: 10.3390/molecules27072195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/25/2022] [Indexed: 01/27/2023] Open
Abstract
The exact cellular and molecular mechanisms of multiple sclerosis and other autoimmune diseases have not been established. Autoimmune pathologies are known to be associated with faults in the immune system and changes in the differentiation profiles of bone marrow stem cells. This study analyzed various characteristics of experimental autoimmune encephalomyelitis (EAE) in 2D2 mice. Differentiation profiles of six hematopoietic stem cells of bone marrow were found to significantly differ in 2D2 male and female mice during the spontaneous development of EAE. In addition, we found various properties of B and T cells, CD4+ and CD8+ lymphocytes in blood and several organs (bone marrow, spleen, thymus, and lymph nodes) of 2D2 male and female mice to be considerably different. These changes in hematopoietic stem cells differentiation profiles and level of lymphocyte proliferation in various organs of 2D2 mice were found to induce the production of IgGs against DNA, myelin basic protein, and myelin oligodendrocyte glycoprotein, increasing the number of autoantibodies hydrolyzing these substrates. We compared the changes of these immunological and biochemical parameters in 2D2 mice with those of mice of two other lines (Th and C57BL/6), also prone to spontaneous development of EAE. Some noticeable and even extreme variations were found in the time-related development of parameters between male and female mice of 2D2, Th, and C57BL/6 lines. Despite some differences, mice of all three lines demonstrated the changes in hematopoietic stem cells profiles, lymphocyte content, and production of catalytic autoantibodies. Given that these changes are harmful to mice, we believe them to cause the development of experimental autoimmune encephalomyelitis.
Collapse
|
16
|
Longoria V, Parcel H, Toma B, Minhas A, Zeine R. Neurological Benefits, Clinical Challenges, and Neuropathologic Promise of Medical Marijuana: A Systematic Review of Cannabinoid Effects in Multiple Sclerosis and Experimental Models of Demyelination. Biomedicines 2022; 10:539. [PMID: 35327341 PMCID: PMC8945692 DOI: 10.3390/biomedicines10030539] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/22/2022] Open
Abstract
Despite current therapeutic strategies for immunomodulation and relief of symptoms in multiple sclerosis (MS), remyelination falls short due to dynamic neuropathologic deterioration and relapses, leading to accrual of disability and associated patient dissatisfaction. The potential of cannabinoids includes add-on immunosuppressive, analgesic, neuroprotective, and remyelinative effects. This study evaluates the efficacy of medical marijuana in MS and its experimental animal models. A systematic review was conducted by a literature search through PubMed, ProQuest, and EBSCO electronic databases for studies reported since 2007 on the use of cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) in MS and in experimental autoimmune encephalomyelitis (EAE), Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), and toxin-induced demyelination models. Study selection and data extraction were performed by 3 reviewers, and 28 studies were selected for inclusion. The certainty of evidence was appraised using the Cochrane GRADE approach. In clinical studies, there was low- and moderate-quality evidence that treatment with ~1:1 CBD/THC mixtures as a nabiximols (Sativex®) oromucosal spray reduced numerical rating scale (NRS) scores for spasticity, pain, and sleep disturbance, diminished bladder overactivity, and decreased proinflammatory cytokine and transcription factor expression levels. Preclinical studies demonstrated decreases in disease severity, hindlimb stiffness, motor function, neuroinflammation, and demyelination. Other experimental systems showed the capacity of cannabinoids to promote remyelination in vitro and by electron microscopy. Modest short-term benefits were realized in MS responders to adjunctive therapy with CBD/THC mixtures. Future studies are recommended to investigate the cellular and molecular mechanisms of cannabinoid effects on MS lesions and to evaluate whether medical marijuana can accelerate remyelination and retard the accrual of disability over the long term.
Collapse
Affiliation(s)
- Victor Longoria
- Basic Medical Sciences, St. Vincent Campus, Saint James School of Medicine, 1480 Renaissance Drive, Park Ridge, IL 60068, USA; (V.L.); (H.P.); (B.T.); (A.M.)
| | - Hannah Parcel
- Basic Medical Sciences, St. Vincent Campus, Saint James School of Medicine, 1480 Renaissance Drive, Park Ridge, IL 60068, USA; (V.L.); (H.P.); (B.T.); (A.M.)
| | - Bameelia Toma
- Basic Medical Sciences, St. Vincent Campus, Saint James School of Medicine, 1480 Renaissance Drive, Park Ridge, IL 60068, USA; (V.L.); (H.P.); (B.T.); (A.M.)
| | - Annu Minhas
- Basic Medical Sciences, St. Vincent Campus, Saint James School of Medicine, 1480 Renaissance Drive, Park Ridge, IL 60068, USA; (V.L.); (H.P.); (B.T.); (A.M.)
| | - Rana Zeine
- School of Natural Sciences, Kean University, 1000 Morris Ave., Union, NJ 07083, USA
| |
Collapse
|
17
|
Tredicine M, Camponeschi C, Pirolli D, Lucchini M, Valentini M, Geloso MC, Mirabella M, Fidaleo M, Righino B, Moliterni C, Giorda E, Rende M, De Rosa MC, Foti M, Constantin G, Ria F, Di Sante G. A TLR/CD44 axis regulates T cell trafficking in experimental and human multiple sclerosis. iScience 2022; 25:103763. [PMID: 35128357 PMCID: PMC8804271 DOI: 10.1016/j.isci.2022.103763] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/28/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
In the pathogenesis of autoimmune disorders, the modulation of leukocytes' trafficking plays a central role, still poorly understood. Here, we focused on the effect of TLR2 ligands in trafficking of T helper cells through reshuffling of CD44 isoforms repertoire. Concurrently, strain background and TLR2 haplotype affected Wnt/β-catenin signaling pathway and expression of splicing factors. During EAE, mCD44 v9- v 10 was specifically enriched in the forebrain and showed an increased ability to bind stably to osteopontin. Similarly, we observed that hCD44 v7 was highly enriched in cells of cerebrospinal fluid from MS patients with active lesions. Moreover, TLRs engagement modulated the composition of CD44 variants also in human T helper cells, supporting the hypothesis that pathogens or commensals, through TLRs, in turn modulate the repertoire of CD44 isoforms, thereby controlling the distribution of lesions in the CNS. The interference with this mechanism(s) represents a potential tool for prevention and treatment of autoimmune relapses and exacerbations.
Collapse
Affiliation(s)
- Maria Tredicine
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Chiara Camponeschi
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Davide Pirolli
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC) -CNR, Largo Francesco Vito 1,00168 Rome, Italy
| | - Matteo Lucchini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
- Centro di ricerca per la Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1,00168 Rome, Italy
| | - Mariagrazia Valentini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
| | - Maria Concetta Geloso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1,00168 Rome, Italy
| | - Massimiliano Mirabella
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
- Centro di ricerca per la Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1,00168 Rome, Italy
| | - Marco Fidaleo
- Department of Biology and Biotechnology Charles Darwin, University of Rome Sapienza,00185 Rome, Italy
| | - Benedetta Righino
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC) -CNR, Largo Francesco Vito 1,00168 Rome, Italy
| | - Camilla Moliterni
- Department of Biology and Biotechnology Charles Darwin, University of Rome Sapienza,00185 Rome, Italy
| | - Ezio Giorda
- Core Facilities di Ricerca, Ospedale Pediatrico Bambino Gesù Roma – IRCCS, V.le Ferdinando Baldelli,40,00146 Roma, Italy
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinic and Forensic Anatomy, University of Perugia, Piazza L. Severi, 06132 Perugia, Italy
| | - Maria Cristina De Rosa
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC) -CNR, Largo Francesco Vito 1,00168 Rome, Italy
| | - Maria Foti
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8,37134 Verona, Italy
| | - Francesco Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
| | - Gabriele Di Sante
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Medicine and Surgery, Section of Human, Clinic and Forensic Anatomy, University of Perugia, Piazza L. Severi, 06132 Perugia, Italy
| |
Collapse
|
18
|
Farnesol induces protection against murine CNS inflammatory demyelination and modifies gut microbiome. Clin Immunol 2022; 235:108766. [PMID: 34091018 PMCID: PMC8660955 DOI: 10.1016/j.clim.2021.108766] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/21/2021] [Accepted: 05/31/2021] [Indexed: 02/08/2023]
Abstract
Farnesol is a 15‑carbon organic isoprenol synthesized by plants and mammals with anti-oxidant, anti-inflammatory, and neuroprotective activities. We sought to determine whether farnesol treatment would result in protection against murine experimental autoimmune encephalomyelitis (EAE), a well-established model of multiple sclerosis (MS). We compared disease progression and severity in C57BL/6 mice treated orally with 100 mg/kg/day farnesol solubilized in corn oil to corn-oil treated and untreated EAE mice. Farnesol significantly delayed the onset of EAE (by ~2 days) and dramatically decreased disease severity (~80%) compared to controls. Disease protection by farnesol was associated with a significant reduction in spinal cord infiltration by monocytes-macrophages, dendritic cells, CD4+ T cells, and a significant change in gut microbiota composition, including a decrease in the Firmicutes:Bacteroidetes ratio. The study suggests FOL could protect MS patients against CNS inflammatory demyelination by partially modulating the gut microbiome composition.
Collapse
|
19
|
Bourque J, Hawiger D. Applications of Antibody-Based Antigen Delivery Targeted to Dendritic Cells In Vivo. Antibodies (Basel) 2022; 11:antib11010008. [PMID: 35225867 PMCID: PMC8884005 DOI: 10.3390/antib11010008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
Recombinant immunoglobulins, derived from monoclonal antibodies recognizing the defined surface epitopes expressed on dendritic cells, have been employed for the past two decades to deliver antigens to dendritic cells in vivo, serving as critical tools for the investigation of the corresponding T cell responses. These approaches originated with the development of the recombinant chimeric antibody against a multilectin receptor, DEC-205, which is present on subsets of murine and human conventional dendritic cells. Following the widespread application of antigen targeting through DEC-205, similar approaches then utilized other epitopes as entry points for antigens delivered by specific antibodies to multiple types of dendritic cells. Overall, these antigen-delivery methodologies helped to reveal the mechanisms underlying tolerogenic and immunogenic T cell responses orchestrated by dendritic cells. Here, we discuss the relevant experimental strategies as well as their future perspectives, including their translational relevance.
Collapse
Affiliation(s)
| | - Daniel Hawiger
- Correspondence: ; Tel.: +1-314-977-8875; Fax: +1-314-977-8717
| |
Collapse
|
20
|
Nissen JC, Tsirka SE. Preclinical model of multiple sclerosis: Methods in autoimmune demyelination. Methods Cell Biol 2022; 168:67-86. [DOI: 10.1016/bs.mcb.2021.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Grigg JB, Shanmugavadivu A, Regen T, Parkhurst CN, Ahmed A, Joseph AM, Mazzucco M, Gronke K, Diefenbach A, Eberl G, Vartanian T, Waisman A, Sonnenberg GF. Antigen-presenting innate lymphoid cells orchestrate neuroinflammation. Nature 2021; 600:707-712. [PMID: 34853467 PMCID: PMC8702489 DOI: 10.1038/s41586-021-04136-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/14/2021] [Indexed: 01/03/2023]
Abstract
Pro-inflammatory T cells in the central nervous system (CNS) are causally associated with multiple demyelinating and neurodegenerative diseases1-6, but the pathways that control these responses remain unclear. Here we define a population of inflammatory group 3 innate lymphoid cells (ILC3s) that infiltrate the CNS in a mouse model of multiple sclerosis. These ILC3s are derived from the circulation, localize in proximity to infiltrating T cells in the CNS, function as antigen-presenting cells that restimulate myelin-specific T cells, and are increased in individuals with multiple sclerosis. Notably, antigen presentation by inflammatory ILC3s is required to promote T cell responses in the CNS and the development of multiple-sclerosis-like disease in mouse models. By contrast, conventional and tissue-resident ILC3s in the periphery do not appear to contribute to disease induction, but instead limit autoimmune T cell responses and prevent multiple-sclerosis-like disease when experimentally targeted to present myelin antigen. Collectively, our data define a population of inflammatory ILC3s that is essential for directly promoting T-cell-dependent neuroinflammation in the CNS and reveal the potential of harnessing peripheral tissue-resident ILC3s for the prevention of autoimmune disease.
Collapse
Affiliation(s)
- John B Grigg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Arthi Shanmugavadivu
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christopher N Parkhurst
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Anees Ahmed
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ann M Joseph
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Michael Mazzucco
- Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Konrad Gronke
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
| | - Gerard Eberl
- Microenvironment and Immunity Unit, Institut Pasteur, Paris, France
| | - Timothy Vartanian
- Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
22
|
Tiberi M, Evron T, Saracini S, Boffa L, Mercuri NB, Chintalacharuvu SR, Atamas SP, Chiurchiù V. Potent T cell-mediated anti-inflammatory role of the selective CB2 agonist lenabasum in multiple sclerosis. Neuropathol Appl Neurobiol 2021; 48:e12768. [PMID: 34543449 DOI: 10.1111/nan.12768] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/27/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Lenabasum is a synthetic cannabinoid receptor type-2 (CB2) agonist able to exert potent anti-inflammatory effects, but its role on T cells remains unknown. OBJECTIVES The present study was undertaken to investigate anti-inflammatory mechanisms of lenabasum in T lymphocyte subsets and its in vivo therapeutic efficacy in experimental autoimmune encephalomyelitis (EAE). METHODS Mononuclear cells from 17 healthy subjects (HS) and 25 relapsing-remitting multiple sclerosis (RRMS) patients were activated in presence or absence of lenabasum and analysed by flow cytometry and qRT-PCR. EAE mice were treated with lenabasum, and clinical score and neuroinflammation were evaluated. RESULTS Lenabasum significantly reduced TNF-a production from CD4+ T cells and CD8+ T cells in a dose-dependent manner in both HS and RRMS patients. In MS patients, lenabasum also reduced activation marker CD25 and inhibited IL-2 production from both T cell subsets and IFN-γ and IL-17 from committed Th1 and Th17 cells, respectively. These effects were blocked by the pretreatment with selective CB2 inverse agonist SR144528. In vivo treatment of EAE mice with lenabasum significantly ameliorated disease severity, reduced neuroinflammation and demyelination in spinal cord. CONCLUSION Lenabasum exerts potent T cell-mediated immunomodulatory effects, suggesting CB2 as a promising pharmacological target to counteract neuroinflammation in MS.
Collapse
Affiliation(s)
- Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Tama Evron
- Department of Discovery and Preclinical Development, Corbus Pharmaceuticals, Inc., Norwood, MA, USA
| | - Stefano Saracini
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Laura Boffa
- Neurology Unit, Tor Vergata Hospital, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Tor Vergata Hospital, Rome, Italy.,Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Subba R Chintalacharuvu
- Department of Discovery and Preclinical Development, Corbus Pharmaceuticals, Inc., Norwood, MA, USA
| | - Sergei P Atamas
- Department of Discovery and Preclinical Development, Corbus Pharmaceuticals, Inc., Norwood, MA, USA
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, Rome, Italy.,Institute of Translational Pharmacology, National Research Council, Rome, Italy
| |
Collapse
|
23
|
Pfeil J, Simonetti M, Lauer U, von Thülen B, Durek P, Poulsen C, Pawlowska J, Kröger M, Krähmer R, Leenders F, Hoffmann U, Hamann A. Prevention of EAE by tolerogenic vaccination with PEGylated antigenic peptides. Ther Adv Chronic Dis 2021; 12:20406223211037830. [PMID: 34408824 PMCID: PMC8366199 DOI: 10.1177/20406223211037830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/13/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Therapeutic treatment options for chronic autoimmune disorders such as multiple sclerosis (MS) rely largely on the use of non-specific immunosuppressive drugs, which are not able to cure the disease. Presently, approaches to induce antigen-specific tolerance as a therapeutic approach; for example, by peptide-based tolerogenic 'inverse' vaccines have regained great interest. We have previously shown that coupling of peptides to carriers can enhance their capacity to induce regulatory T cells in vivo. METHOD In this present study, we investigated whether the tolerogenic potential of immunodominant myelin T-cell epitopes can be improved by conjugation to the synthetic carrier polyethylene glycol (PEG) in an experimental autoimmune encephalomyelitis (EAE) mouse model for chronic MS (MOG C57BL/6). RESULTS Preventive administration of the PEGylated antigenic peptide could strongly suppress the development of EAE, accompanied by reduced immune cell infiltration in the central nervous system (CNS). Depletion of regulatory T cells (Tregs) abrogated the protective effect indicating that Tregs play a crucial role in induction of antigen-specific tolerance in EAE. Treatment during the acute phase of disease was safe and did not induce immune activation. However, treatment at the peak of disease did not affect the disease course, suggesting that either induction of Tregs does not occur in the highly inflamed situation, or that the immune system is refractory to regulation in this condition. CONCLUSION PEGylation of antigenic peptides is an effective and feasible strategy to improve tolerogenic (Treg-inducing) peptide-based vaccines, but application for immunotherapy of overt disease might require modifications or combination therapies that simultaneously suppress effector mechanisms.
Collapse
Affiliation(s)
- Jennifer Pfeil
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum, a Leibniz-Institute, Berlin, Germany
| | - Mario Simonetti
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin, Berlin, Germany
| | - Uta Lauer
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum, a Leibniz-Institute, Berlin, Germany
| | | | - Pawel Durek
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum, a Leibniz-Institute, Berlin, Germany
| | - Christina Poulsen
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum, a Leibniz-Institute, Berlin, Germany
| | - Justyna Pawlowska
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum, a Leibniz-Institute, Berlin, Germany
| | - Matthias Kröger
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum, a Leibniz-Institute, Berlin, Germany
| | | | | | - Ute Hoffmann
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum, a Leibniz-Institute, Berlin, Germany
| | - Alf Hamann
- Experimental Rheumatology, Deutsches Rheuma-Forschungszentrum Berlin, Charitéplatz 1, Berlin 10117, Germany
| |
Collapse
|
24
|
Gharagozloo M, Smith MD, Sotirchos ES, Jin J, Meyers K, Taylor M, Garton T, Bannon R, Lord HN, Dawson TM, Dawson VL, Lee S, Calabresi PA. Therapeutic Potential of a Novel Glucagon-like Peptide-1 Receptor Agonist, NLY01, in Experimental Autoimmune Encephalomyelitis. Neurotherapeutics 2021; 18:1834-1848. [PMID: 34260042 PMCID: PMC8608955 DOI: 10.1007/s13311-021-01088-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2021] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS), characterized by demyelination, gliosis, and neurodegeneration. While the currently available disease-modifying therapies effectively suppress the immune attack on the CNS, there are no therapies to date that directly mitigate neurodegeneration. Glucagon-like peptide-1 (GLP-1) is a small peptide hormone that maintains glucose homeostasis. A novel GLP-1 receptor (GLP-1R) agonist, NLY01, was recently shown to have neuroprotective effects in the animal models of Parkinson's disease and is now in a phase 2 clinical trial. In this study, we investigated the therapeutic potential of NLY01 in a mouse model of MS, experimental autoimmune encephalomyelitis (EAE). Our data show that NLY01 delays the onset and attenuates the severity of EAE in a prevention paradigm, when given before disease onset. NLY01 inhibits the activation of immune cells in the spleen and reduces their trafficking into the CNS. In addition, we show that NLY01 suppresses the production of chemokines that are involved in leukocyte recruitment to the site of inflammation. The anti-inflammatory effect of NLY01 at the early stage of EAE may block the expression of the genes associated with neurotoxic astrocytes in the optic nerves, thereby preventing retinal ganglion cell (RGC) loss in the progressive stage of EAE. In the therapeutic paradigm, NLY01 significantly decreases the clinical score and second attack in a model of relapsing-remitting EAE. GLP-1R agonists may have dual efficacy in MS by suppressing peripheral and CNS inflammation, thereby limiting neuronal loss.
Collapse
Affiliation(s)
| | | | | | - Jing Jin
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA
| | - Keya Meyers
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA
| | | | - Thomas Garton
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA
| | - Riley Bannon
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA
| | | | - Ted M Dawson
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Peter A Calabresi
- Department of Neurology, Johns Hopkins, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, 600 N. Wolfe St, Baltimore, MD, 21287, USA.
| |
Collapse
|
25
|
Ondek K, Nasirishargh A, Dayton JR, Nuño MA, Cruz-Orengo L. Strain and sex differences in somatosensation and sociability during experimental autoimmune encephalomyelitis. Brain Behav Immun Health 2021; 14:100262. [PMID: 34589768 PMCID: PMC8474462 DOI: 10.1016/j.bbih.2021.100262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 12/01/2022] Open
Abstract
Multiple Sclerosis (MS) is an immune-mediated disease that results in major locomotor deficits. However, recent studies have revealed that fatigue, slow processing speed, and memory impairment are the top variables impacting employment status for MS patients. These suggest that cognitive effects may have a greater impact on productivity, lifestyle, and quality of life than do disease-related motor deficits. However, these debilitating non-locomotive effects have been largely overlooked in rodent models of the disease, such as experimental autoimmune encephalomyelitis (EAE). We hypothesized that murine EAE can also be used to assess non-locomotive dysfunctions (mood, sociability, muscle strength, and balance), as well as potential biases in these dysfunctions due to sex and/or strain. We actively immunized male and female C57BL/6 (B6) and SJL mice for EAE and evaluated their performance on the Deacon's weight grip test, Kondziela's inverted screen test, Hall's rope grip test, manual von Frey test for somatic nociception, and a three-chamber social preference paradigm. We hypothesized that EAE progression is associated with changes in muscle strength, balance, pain, and sociability and that these variations are linked to sex and/or strain. Our results indicate that strain but not sex influenced differences in muscle strength and balance during EAE, and both sex and strain have an impact on mechanical nociception, regardless of EAE disease status. Furthermore, both sex and strain had complex effects on differences in sociability. In conclusion, testing these additional modalities during EAE helps to unveil other signs and symptoms that could be used to determine the efficacy of a drug or treatment in the modulation of a MS-like behavior.
Collapse
Affiliation(s)
- Katelynn Ondek
- University of California, Davis. Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Aida Nasirishargh
- University of California, Davis. Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Jacquelyn R. Dayton
- University of California, Davis. Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| | - Miriam A. Nuño
- University of California, Davis. Department of Public Health, Division of Biostatistics, School of Medicine, Public Health/Medical Sciences Bldg. 1-C, Davis, CA 95616, USA
| | - Lillian Cruz-Orengo
- University of California, Davis. Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA, 95616, USA
| |
Collapse
|
26
|
Psenicka MW, Smith BC, Tinkey RA, Williams JL. Connecting Neuroinflammation and Neurodegeneration in Multiple Sclerosis: Are Oligodendrocyte Precursor Cells a Nexus of Disease? Front Cell Neurosci 2021; 15:654284. [PMID: 34234647 PMCID: PMC8255483 DOI: 10.3389/fncel.2021.654284] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
The pathology in neurodegenerative diseases is often accompanied by inflammation. It is well-known that many cells within the central nervous system (CNS) also contribute to ongoing neuroinflammation, which can promote neurodegeneration. Multiple sclerosis (MS) is both an inflammatory and neurodegenerative disease in which there is a complex interplay between resident CNS cells to mediate myelin and axonal damage, and this communication network can vary depending on the subtype and chronicity of disease. Oligodendrocytes, the myelinating cell of the CNS, and their precursors, oligodendrocyte precursor cells (OPCs), are often thought of as the targets of autoimmune pathology during MS and in several animal models of MS; however, there is emerging evidence that OPCs actively contribute to inflammation that directly and indirectly contributes to neurodegeneration. Here we discuss several contributors to MS disease progression starting with lesion pathology and murine models amenable to studying particular aspects of disease. We then review how OPCs themselves can play an active role in promoting neuroinflammation and neurodegeneration, and how other resident CNS cells including microglia, astrocytes, and neurons can impact OPC function. Further, we outline the very complex and pleiotropic role(s) of several inflammatory cytokines and other secreted factors classically described as solely deleterious during MS and its animal models, but in fact, have many neuroprotective functions and promote a return to homeostasis, in part via modulation of OPC function. Finally, since MS affects patients from the onset of disease throughout their lifespan, we discuss the impact of aging on OPC function and CNS recovery. It is becoming clear that OPCs are not simply a bystander during MS progression and uncovering the active roles they play during different stages of disease will help uncover potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Morgan W. Psenicka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brandon C. Smith
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Brain Health Research Institute, Kent State University, Kent, OH, United States
| |
Collapse
|
27
|
Tolmacheva AS, Aulova KS, Urusov AE, Orlovskaya IA, Nevinsky GA. Increase in Autoantibodies-Abzymes with Peroxidase and Oxidoreductase Activities in Experimental Autoimmune Encephalomyelitis Mice during the Development of EAE Pathology. Molecules 2021; 26:molecules26072077. [PMID: 33916567 PMCID: PMC8038483 DOI: 10.3390/molecules26072077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 11/16/2022] Open
Abstract
The exact mechanisms of multiple sclerosis (MS) development are still unknown, but the development of experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice is associated with the violation of bone marrow hematopoietic stem cells (HSCs) differentiation profiles associated with the production of harmful for human’s autoantibodies hydrolyzing myelin basic protein, myelin oligodendrocyte glycoprotein (MOG35–55), and DNA. It was shown that IgGs from the sera of healthy humans and autoimmune patients oxidize many different compounds due to their H2O2-dependent peroxidase and oxidoreductase activity in the absence of H2O2. Here we first analyzed the change in the relative redox activities of IgGs antibodies from the blood of C57BL/6 mice over time at different stages of the EAE development. It was shown that the peroxidase activity of mice IgGs in the oxidation of ABTS (2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) is on average 6.9-fold higher than the oxidoreductase activity. The peroxidase activity of IgGs increased during the spontaneous development of EAE during 40 days, 1.4-fold. After EAE development acceleration due to mice immunization with MOG35–55 (5.3-fold), complexes of bovine DNA with methylated bovine serum albumin (DNA-metBSA; 3.5-fold), or with histones (2.6-fold), the activity was increased much faster. The increase in peroxidase activity after mice immunization with MOG35–55 and DNA-metBSA up to 40 days of experiments was relatively gradual, while for DNA-histones complex was observed its sharp increase at the acute phase of EAE (14–20 days). All data show that IgGs’ redox activities can play an important role in the protection of mice from toxic compounds and oxidative stress.
Collapse
MESH Headings
- Animals
- Antibodies, Catalytic/metabolism
- Autoantibodies/metabolism
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/pathology
- Humans
- Hydrogen Peroxide/pharmacology
- Immunoglobulin G/metabolism
- Mice
- Mice, Inbred C57BL
- Myelin Basic Protein/metabolism
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- Oxidation-Reduction
- Oxidoreductases/immunology
- Oxidoreductases/metabolism
- Peptide Fragments/administration & dosage
- Peroxidases/immunology
- Peroxidases/metabolism
Collapse
Affiliation(s)
- Anna S. Tolmacheva
- Institute of Chemical Biology and Fundamental Medicine, SB of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.T.); (K.S.A.); (A.E.U.)
| | - Kseniya S. Aulova
- Institute of Chemical Biology and Fundamental Medicine, SB of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.T.); (K.S.A.); (A.E.U.)
| | - Andrey E. Urusov
- Institute of Chemical Biology and Fundamental Medicine, SB of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.T.); (K.S.A.); (A.E.U.)
| | - Irina A. Orlovskaya
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Medical Sciences, 630090 Novosibirsk, Russia;
| | - Georgy A. Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, SB of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.T.); (K.S.A.); (A.E.U.)
- Correspondence: ; Tel.: +7-383-3635126; Fax: +7-383-3635153
| |
Collapse
|
28
|
Gómez-López AR, Manich G, Recasens M, Almolda B, González B, Castellano B. Evaluation of Myelin Phagocytosis by Microglia/Macrophages in Nervous Tissue Using Flow Cytometry. Curr Protoc 2021; 1:e73. [PMID: 33687792 DOI: 10.1002/cpz1.73] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Determination of microglial phagocytosis of myelin has acquired importance in the study of demyelinating diseases. One strategy to determine microglial phagocytosis capacity consists of assaying microglia with fluorescently labeled myelin; however, most approaches are performed in cell culture, where microglia usually show important phenotypic differences compared with in vivo conditions. In this article we describe an adapted flow cytometry protocol to assay myelin phagocytosis by microglia obtained directly from in vivo tissue of the central nervous system. Key steps for a first analysis of phagocytic microglia are provided. Additionally, we describe how to fluorescently label myelin using a pH-sensitive tag, pHrodo™ Green STP Ester. © 2021 Wiley Periodicals LLC. Basic Protocol: Assay for determination of myelin phagocytosis by microglia/macrophages using flow cytometry Support Protocol 1: Conjugation of isolated and purified myelin with pHrodo Green STP Ester Support Protocol 2: Quantification of phagocytic cell number by flow cytometry.
Collapse
Affiliation(s)
- Ariadna Regina Gómez-López
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Manich
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireia Recasens
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Beatriz Almolda
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Berta González
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Bernardo Castellano
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
29
|
Xiao D, Zhang W, Wang Q, Li X, Zhang Y, Rasouli J, Casella G, Ciric B, Curtis M, Rostami A, Zhang GX. CRISPR-mediated rapid generation of neural cell-specific knockout mice facilitates research in neurophysiology and pathology. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:755-764. [PMID: 33738329 PMCID: PMC7940702 DOI: 10.1016/j.omtm.2021.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/14/2021] [Indexed: 01/10/2023]
Abstract
Inducible conditional knockout mice are important tools for studying gene function and disease therapy, but their generation is costly and time-consuming. We introduced clustered regularly interspaced short palindromic repeats (CRISPR) and Cre into an LSL-Cas9 transgene-carrying mouse line by using adeno-associated virus (AAV)-PHP.eB to rapidly knockout gene(s) specifically in central nervous system (CNS) cells of adult mice. NeuN in neurons and GFAP in astrocytes were knocked out 2 weeks after an intravenous injection of vector, with an efficiency comparable to that of inducible Cre-loxP conditional knockout. For functional testing, we generated astrocyte-specific Act1 knockout mice, which exhibited a phenotype similar to mice with Cre-loxP-mediated Act1 knockout, in an animal model of multiple sclerosis (MS), an autoimmune disorder of the CNS. With this novel technique, neural cell-specific knockout can be induced rapidly (few weeks) and cost-effectively. Our study provides a new approach to building inducible conditional knockout mice, which would greatly facilitate research on CNS biology and disease.
Collapse
Affiliation(s)
- Dan Xiao
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Weifeng Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA.,College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Qing Wang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xing Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yuan Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Giacomo Casella
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark Curtis
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
30
|
Brate AA, Boyden AW, Jensen IJ, Badovinac VP, Karandikar NJ. A Functionally Distinct CXCR3 +/IFN-γ +/IL-10 + Subset Defines Disease-Suppressive Myelin-Specific CD8 T Cells. THE JOURNAL OF IMMUNOLOGY 2021; 206:1151-1160. [PMID: 33558376 DOI: 10.4049/jimmunol.2001143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the CNS. We have previously demonstrated that CNS-specific CD8 T cells possess a disease-suppressive function in MS and variations of its animal model, experimental autoimmune encephalomyelitis (EAE), including the highly clinically relevant relapsing-remitting EAE disease course. Regulatory CD8 T cell subsets have been identified in EAE and other autoimmune diseases, but studies vary in defining phenotypic properties of these cells. In relapsing-remitting EAE, PLP178-191 CD8 T cells suppress disease, whereas PLP139-151 CD8 T cells lack this function. In this study, we used this model to delineate the unique phenotypic properties of CNS-specific regulatory PLP178-191 CD8 T cells versus nonregulatory PLP139-151 or OVA323-339 CD8 T cells. Using multiparametric flow cytometric analyses of phenotypic marker expression, we identified a CXCR3+ subpopulation among activated regulatory CD8 T cells, relative to nonregulatory counterparts. This subset exhibited increased degranulation and IFN-γ and IL-10 coproduction. A similar subset was also identified in C57BL/6 mice within autoregulatory PLP178-191 CD8 T cells but not within nonregulatory OVA323-339 CD8 T cells. This disease-suppressing CD8 T cell subpopulation provides better insights into functional regulatory mechanisms, and targeted enhancement of this subset could represent a novel immunotherapeutic approach for MS.
Collapse
Affiliation(s)
- Ashley A Brate
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241.,Iowa City Veterans Affairs Medical Center, Iowa City, IA 52241; and
| | - Alexander W Boyden
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Iowa City Veterans Affairs Medical Center, Iowa City, IA 52241; and
| | - Isaac J Jensen
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241.,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52241
| | - Nitin J Karandikar
- Department of Pathology, University of Iowa Health Care, Iowa City, IA 52241; .,Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241.,Iowa City Veterans Affairs Medical Center, Iowa City, IA 52241; and
| |
Collapse
|
31
|
Tatomir A, Beltrand A, Nguyen V, Boodhoo D, Mekala A, Cudrici C, Badea TC, Muresanu DF, Rus V, Rus H. RGC-32 Regulates Generation of Reactive Astrocytes in Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 11:608294. [PMID: 33569054 PMCID: PMC7868332 DOI: 10.3389/fimmu.2020.608294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022] Open
Abstract
Astrocytes are increasingly recognized as critical contributors to multiple sclerosis pathogenesis. We have previously shown that lack of Response Gene to Complement 32 (RGC-32) alters astrocyte morphology in the spinal cord at the peak of experimental autoimmune encephalomyelitis (EAE), suggesting a role for RGC-32 in astrocyte differentiation. In this study, we analyzed the expression and distribution of astrocytes and astrocyte progenitors by immunohistochemistry in spinal cords of wild-type (WT) and RGC-32-knockout (KO) mice with EAE and of normal adult mice. Our analysis showed that during acute EAE, WT astrocytes had a reactive morphology and increased GFAP expression, whereas RGC-32 KO astrocytes had a morphology similar to that of radial glia and an increased expression of progenitor markers such as vimentin and fatty acid binding protein 7 (FABP7). In control mice, GFAP expression and astrocyte density were also significantly higher in the WT group, whereas the number of vimentin and FABP7-positive radial glia was significantly higher in the RGC-32 KO group. In vitro studies on cultured neonatal astrocytes from WT and RGC-32 KO mice showed that RGC-32 regulates a complex array of molecular networks pertaining to signal transduction, growth factor expression and secretion, and extracellular matrix (ECM) remodeling. Among the most differentially expressed factors were insulin-like growth factor 1 (IGF1), insulin-like growth factor binding proteins (IGFBPs), and connective tissue growth factor (CTGF); their expression was downregulated in RGC-32-depleted astrocytes. The nuclear translocation of STAT3, a transcription factor critical for astrogliogenesis and driving glial scar formation, was also impaired after RGC-32 silencing. Taken together, these data suggest that RGC-32 is an important regulator of astrocyte differentiation during EAE and that in the absence of RGC-32, astrocytes are unable to fully mature and become reactive astrocytes.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Cell Differentiation
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fatty Acid-Binding Protein 7/metabolism
- Female
- Glial Fibrillary Acidic Protein/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Rats, Sprague-Dawley
- Signal Transduction
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Vimentin/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Austin Beltrand
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Armugam Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Cornelia Cudrici
- Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tudor C. Badea
- Retinal Circuit Development and Genetics Unit, Neurobiology Neurodegeneration & Repair Laboratory (N-NRL), National Eye Institute, Bethesda, MD, United States
| | - Dafin F. Muresanu
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, United States
| |
Collapse
|
32
|
Faber H, Kurtoic D, Krishnamoorthy G, Weber P, Pütz B, Müller-Myhsok B, Weber F, Andlauer TFM. Gene Expression in Spontaneous Experimental Autoimmune Encephalomyelitis Is Linked to Human Multiple Sclerosis Risk Genes. Front Immunol 2020; 11:2165. [PMID: 33072080 PMCID: PMC7531036 DOI: 10.3389/fimmu.2020.02165] [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: 05/21/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Recent genome-wide association studies have identified over 230 genetic risk loci for multiple sclerosis. Current experimental autoimmune encephalomyelitis (EAE) models requiring active induction of disease may not be optimally suited for the characterization of the function of these genes. We have thus used gene expression profiling to study whether spontaneous opticospinal EAE (OSE) or MOG-induced EAE mirrors the genetic contribution to the pathogenesis of multiple sclerosis more faithfully. To this end, we compared gene expression in OSE and MOG EAE models and analyzed the relationship of both models to human multiple sclerosis risk genes and T helper cell biology. We observed stronger gene expression changes and an involvement of more pathways of the adaptive immune system in OSE than MOG EAE. Furthermore, we demonstrated a more extensive enrichment of human MS risk genes among transcripts differentially expressed in OSE than was the case for MOG EAE. Transcripts differentially expressed only in diseased OSE mice but not in MOG EAE were significantly enriched for T helper cell-specific transcripts. These transcripts are part of immune-regulatory pathways. The activation of the adaptive immune system and the enrichment of both human multiple sclerosis risk genes and T helper cell-specific transcripts were also observed in OSE mice showing only mild disease signs. These expression changes may, therefore, be indicative of processes at disease onset. In summary, more human multiple sclerosis risk genes were differentially expressed in OSE than was observed for MOG EAE, especially in TH1 cells. When studying the functional role of multiple sclerosis risk genes and pathways during disease onset and their interactions with the environment, spontaneous OSE may thus show advantages over MOG-induced EAE.
Collapse
Affiliation(s)
- Hans Faber
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | | | - Peter Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Frank Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
33
|
DeOca KB, Moorman CD, Garcia BL, Mannie MD. Low-Zone IL-2 Signaling: Fusion Proteins Containing Linked CD25 and IL-2 Domains Sustain Tolerogenic Vaccination in vivo and Promote Dominance of FOXP3 + Tregs in vitro. Front Immunol 2020; 11:541619. [PMID: 33072087 PMCID: PMC7538601 DOI: 10.3389/fimmu.2020.541619] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
Low-zone IL-2 signaling is key to understanding how CD4+ CD25high FOXP3+ regulatory T cells (Tregs) exhibit dominance and overgrow conventional effector T cells (Tcons) that typically express lower levels of the IL-2 receptor alpha chain (i.e., CD25). Thus, modalities such as low-dose IL-2 or IL-2/anti-IL-2 antibody complexes have been advanced in the clinic to selectively expand Treg populations as a treatment for chronic inflammatory autoimmune diseases. However, more effective reagents that efficiently lock IL-2 signaling into a low signaling mode are needed to validate and exploit the low-zone IL-2 signaling niche of Tregs. This study focuses on CD25-IL2 and IL2-CD25 fusion proteins (FPs) that were approximately 32 and 320-fold less potent than IL-2. These FPs exhibited transient binding to transmembrane CD25 on human embryonic kidney (HEK) cells, had partially occluded IL-2 binding sites, and formed higher order multimeric conformers that limited the availability of bioactive IL-2. These FPs exhibited broad bell-shaped concentration ranges that favored dominant Treg outgrowth during continuous culture and were used to derive essentially pure long-term Treg monocultures (∼98% Treg purity). FP-induced Tregs had canonical Treg suppressive activity in that these Tregs suppressed antigen-specific proliferative responses of naïve CD4+ T cells. The in vivo administration of CD25-IL2/Alum elicited robust increases in circulating Tregs and selectively augmented CD25 expression on Tregs but not on Tcons. A single injection of a Myelin Oligodendrocyte Glycoprotein (MOG35-55)-specific tolerogenic vaccine elicited high levels of circulating MOG-specific Tregs in vivo that waned after 2–3 weeks, whereas boosting with CD25-IL2/Alum maintained MOG-specific CD25high Tregs throughout the 30-day observation period. However, these FPs did not antagonize free monomeric IL-2 and lacked therapeutic efficacy in experimental autoimmune encephalomyelitis (EAE). In conclusion, these data reveal that CD25-IL2 FPs can be used to select essentially pure long-term lines of FOXP3+ CD25high Tregs. This study also shows that CD25-IL2 FPs can be administered in vivo in synergy with tolerogenic vaccination to maintain high circulating levels of antigen-specific Tregs. Because tolerogenic vaccination and Treg-based adoptive immunotherapy are limited by gradual waning of Tregs, these FPs have potential utility in sustaining tolerogenic Treg responses in vivo.
Collapse
Affiliation(s)
- Kayla B DeOca
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Cody D Moorman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Brandon L Garcia
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
34
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
35
|
Bapat SP, Liang Y, Zheng Y. Characterization of Immune Cells from Adipose Tissue. ACTA ACUST UNITED AC 2020; 126:e86. [PMID: 31483101 DOI: 10.1002/cpim.86] [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] [Indexed: 12/21/2022]
Abstract
Adipose tissue (AT) serves a crucial role in maintaining organismal metabolic homeostasis. Studies have demonstrated that AT is populated with a diverse array of immune cells that coordinate and regulate AT function. This adipo-immune system is highly dynamic, reflecting the physiologic state of the organism (e.g., obese, lean, aged, or young) as well as the constant physiologic remodeling of AT associated with the daily rhythms of fasting and feeding. Many of the adaptive and maladaptive functional changes of AT are regulated by changes in the quantity and quality of distinct sets of AT-resident immune cells. Here we present protocols to assess the dynamic state of the immune system within AT by constructing censuses of adipose-resident immune cells (macrophages, dendritic cells, neutrophils, eosinophils, NK cells, innate lymphocytes, T cells, and B cells, etc.) based on flow cytometry, which we term adipo-immune profiles (AIPs). Constructing AIPs can be an integral part of assessment for AT health and function. This article describes the protocols to generate such AIPs. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Sagar P Bapat
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Yuqiong Liang
- Nomis Foundation Laboratories of Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California
| | - Ye Zheng
- Nomis Foundation Laboratories of Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
36
|
Safari H, Kelley WJ, Saito E, Kaczorowski N, Carethers L, Shea LD, Eniola-Adefeso O. Neutrophils preferentially phagocytose elongated particles-An opportunity for selective targeting in acute inflammatory diseases. SCIENCE ADVANCES 2020; 6:eaba1474. [PMID: 32577517 PMCID: PMC7286665 DOI: 10.1126/sciadv.aba1474] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/14/2020] [Indexed: 05/18/2023]
Abstract
Polymeric particles have recently been used to modulate the behavior of immune cells in the treatment of various inflammatory conditions. However, there is little understanding of how physical particle parameters affect their specific interaction with different leukocyte subtypes. While particle shape is known to be a crucial factor in their phagocytosis by macrophages, where elongated particles are reported to experience reduced uptake, it remains unclear how shape influences phagocytosis by circulating phagocytes, including neutrophils that are the most abundant leukocyte in human blood. In this study, we investigated the phagocytosis of rod-shaped polymeric particles by human neutrophils relative to other leukocytes. In contrast to macrophages and other mononuclear phagocytes, neutrophils were found to exhibit increased internalization of rods in ex vivo and in vivo experimentation. This result suggests that alteration of particle shape can be used to selectively target neutrophils in inflammatory pathologies where these cells play a substantial role.
Collapse
Affiliation(s)
- Hanieh Safari
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - William J. Kelley
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas Kaczorowski
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lauren Carethers
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
37
|
Regulatory B Cells Normalize CNS Myeloid Cell Content in a Mouse Model of Multiple Sclerosis and Promote Oligodendrogenesis and Remyelination. J Neurosci 2020; 40:5105-5115. [PMID: 32430295 DOI: 10.1523/jneurosci.2840-19.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/03/2020] [Accepted: 05/13/2020] [Indexed: 01/13/2023] Open
Abstract
The unmet medical need of patients with multiple sclerosis (MS) is the inexorable loss of CNS myelin and latterly neurons leading to permanent neurologic disability. Solicitation of endogenous oligodendrocytes progenitor cells, the precursor of oligodendrocytes, to remyelinate axons may abort the onset of disability. In female mice with experimental autoimmune encephalomyelitis (EAE), a murine model of MS, adoptive transfer of IL-10+ regulatory B cells (Bregs) has been shown to reverse EAE by promoting the expansion of peripheral and CNS-infiltrating IL-10+ T cells. Here, we examined whether Bregs treatment and its bystander effect on regulatory T cells are associated with CNS repair as reflected by oligodendrogenesis and remyelination. We have found that transfusion of Bregs reverses established clinical EAE and that clinical improvement is associated with a significant increase in spinal cord remyelination as reflected by g-ratio analysis within the thoracic and lumbar spine. We further observed in the spinal cords of EAE Bregs-treated mice that CNS resident CD11b/CD45intLy6C- microglia, and infiltrating CD11b+/CD45high monocytes/macrophages content reverts to normal and polarize to a M2-like CD206+ phenotype. Concurrently, there was a substantial increase in neo-oligodendrogenesis as manifest by an increase in CD45-/low CNS cells expressing A2B5, an early marker in oligodendrocytes progenitor cell differentiation as well as GalC+/O1+ premyelinating and myelin basic protein+/myelin oligodendrocyte glycoprotein+ mature oligodendrocytes with reciprocal downregulation of paired related homeobox protein 1. These results demonstrate that the clinical benefit of Bregs is associated with normalization of CNS immune milieu and concurrent activation of oligodendrocyte progenitor cells with subsequent remyelination.SIGNIFICANCE STATEMENT In multiple sclerosis patients, demyelination progresses with aging and disease course, leading to irreversible disability. In this study, we have discovered, using a mouse model of multiple sclerosis, that the transfusion of autologous regulatory B cells (Bregs) is able to ameliorate, cure, and sustain the durable remission of the disease. We show that the adoptive transfer of Bregs dramatically decreased the frequency of myeloid-derived cells, both infiltrating monocytes/macrophages and resident microglia, and converted their phenotype to an immunosuppressive-like phenotype. Moreover, we showed that CNS oligodendrocyte progenitor cells are activated following Bregs treatment and differentiate into myelinating oligodendrocytes, which results in neo-oligodendrogenesis and remyelination of spinal cords.
Collapse
|
38
|
Assessing the anterior visual pathway in optic neuritis: recent experimental and clinical aspects. Curr Opin Neurol 2020; 32:346-357. [PMID: 30694926 DOI: 10.1097/wco.0000000000000675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) and related autoimmune disorders of the central nervous system such as neuromyelitis optica spectrum disorders (NMOSD) are characterized by chronic disability resulting from autoimmune neuroinflammation, with demyelination, astrocyte damage, impaired axonal transmission and neuroaxonal loss. Novel therapeutics stopping or reversing the progression of disability are still urgently warranted. This review addresses research on optic neuritis in preclinical experimental models and their translation to clinical trials. RECENT FINDINGS Optic neuritis can be used as paradigm for an MS relapse which can serve to evaluate the efficacy of novel therapeutics in clinical trials with a reasonable duration and cohort size. The advantage is the linear structure of the visual pathway allowing the assessment of visual function and retinal structure as highly sensitive outcome parameters. Experimental autoimmune encephalomyelitis is an inducible, inflammatory and demyelinating central nervous system disease extensively used as animal model of MS. Optic neuritis is part of the clinicopathological manifestations in a number of experimental autoimmune encephalomyelitis models. These have gained increasing interest for studies evaluating neuroprotective and/or remyelinating substances as longitudinal, visual and retinal readouts have become available. SUMMARY Translation of preclinical experiments, evaluating neuroprotective or remyelinating therapeutics to clinical studies is challenging. In-vivo readouts like optical coherence tomography, offers the possibility to transfer experimental study designs to clinical optic neuritis trials.
Collapse
|
39
|
Amarilla AA, Santos-Junior NN, Figueiredo ML, Luiz JPM, Fumagalli MJ, Colón DF, Lippi V, Alfonso HL, Lima-Junior DS, Trabuco AC, Spinieli RL, Desidera AC, Leite-Panissi CRA, Lauretti F, Mendoza SES, Silva CLA, Rego EM, Galvao-Lima LJ, Bassi GS, Penharvel Martíns SLB, Manrique WG, Alves-Filho JC, Cunha FQ, Peng NYG, Modhiran N, Setoh YX, Khromykh AA, Figueiredo LTM, Aquino VH. CCR2 Plays a Protective Role in Rocio Virus-Induced Encephalitis by Promoting Macrophage Infiltration Into the Brain. J Infect Dis 2020; 219:2015-2025. [PMID: 30715407 PMCID: PMC7107438 DOI: 10.1093/infdis/jiz029] [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: 01/10/2019] [Accepted: 01/24/2019] [Indexed: 12/15/2022] Open
Abstract
Rocio virus (ROCV) is a highly neuropathogenic mosquito-transmitted flavivirus responsible for an unprecedented outbreak of human encephalitis during 1975–1976 in Sao Paulo State, Brazil. Previous studies have shown an increased number of inflammatory macrophages in the central nervous system (CNS) of ROCV-infected mice, implying a role for macrophages in the pathogenesis of ROCV. Here, we show that ROCV infection results in increased expression of CCL2 in the blood and in infiltration of macrophages into the brain. Moreover, we show, using CCR2 knockout mice, that CCR2 expression is essential for macrophage infiltration in the brain during ROCV infection and that the lack of CCR2 results in increased disease severity and mortality. Thus, our findings show the protective role of CCR2-mediated infiltration of macrophages in the brain during ROCV infection.
Collapse
Affiliation(s)
- Alberto A Amarilla
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil.,Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | | | - Mario Luis Figueiredo
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Joao Paulo Mesquita Luiz
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | | | - David F Colón
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Veronica Lippi
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Helda Liz Alfonso
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Djalma S Lima-Junior
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, Ribeirao Preto, SP, Brazil
| | - Amanda C Trabuco
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Richard L Spinieli
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Amanda C Desidera
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Christie R A Leite-Panissi
- Department of Psychology, School of Philosophy, Science and Literature of Ribeirao Preto of the University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | - Silvia Elena Sánchez Mendoza
- Division of Hematology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil.,Division of Clinical Oncology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil
| | | | - Eduardo Magalhaes Rego
- Division of Hematology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil.,Division of Clinical Oncology, Department of Internal Medicine, Ribeirao Preto, SP, Brazil
| | - Leonardo J Galvao-Lima
- Department of Immunology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Gabriel S Bassi
- Department of Pharmacology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sandra L B Penharvel Martíns
- Department of Surgery and Anatomy, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Wilson Gomez Manrique
- Animal Health Laboratory, Veterinary Medicine Course, Federal University of Rondonia - UNIR, Rolim de Moura, RO, Brazil
| | - José Carlos Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Fernando Q Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto, SP, Brazil
| | - Nias Y G Peng
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Naphak Modhiran
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Yin Xiang Setoh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Alexander A Khromykh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Luiz T M Figueiredo
- Virology Research Center, Ribeirao Preto, SP, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Victor H Aquino
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
40
|
Musella A, Fresegna D, Rizzo FR, Gentile A, De Vito F, Caioli S, Guadalupi L, Bruno A, Dolcetti E, Buttari F, Bullitta S, Vanni V, Centonze D, Mandolesi G. 'Prototypical' proinflammatory cytokine (IL-1) in multiple sclerosis: role in pathogenesis and therapeutic targeting. Expert Opin Ther Targets 2020; 24:37-46. [PMID: 31899994 DOI: 10.1080/14728222.2020.1709823] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: It has been recognized for about 20 years that interleukin (IL)-1 signaling is implicated in Multiple Sclerosis (MS), a disabling, chronic, inflammatory and neurodegenerative disease of the central nervous system (CNS). Only recently, multifaceted roles of IL-1 emerged in MS pathophysiology as a result of both clinical and preclinical studies. Notably, drugs that directly target the IL-1 system have not been tested so far in MS.Areas covered: Recent studies in animal models, together with the development of ex vivo chimeric MS models, have disclosed a critical role for IL-1 not only at the peripheral level but also within the CNS. In the present review, we highlight the IL-1-dependent neuropathological aspects of MS, by providing an overview of the cells of the immune and CNS systems that respond to IL-1 signaling, and by emphasizing the subsequent effects on the CNS, from demyelinating processes, to synaptopathy, and excitotoxicity.Expert opinion: Drugs that act on the IL-1 system show a therapeutic potential in several autoinflammatory diseases and preclinical studies have highlighted the effects of these compounds in MS. We will discuss why anti-IL-1 therapies in MS have been neglected to date.
Collapse
Affiliation(s)
- Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele University, Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Francesca Romana Rizzo
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | - Silvia Caioli
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Antonio Bruno
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Ettore Dolcetti
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Silvia Bullitta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Diego Centonze
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, Rome, Italy.,Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele University, Rome, Italy
| |
Collapse
|
41
|
Therapeutic intervention in relapsing autoimmune demyelinating disease through induction of myelin-specific regulatory CD8 T cell responses. J Transl Autoimmun 2019; 2. [PMID: 32161909 PMCID: PMC7065686 DOI: 10.1016/j.jtauto.2019.100010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). We have shown that CNS-specific CD8 T cells (CNS-CD8) possess a disease suppressive function in MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Previous studies have focused on the role of these cells predominantly in chronic models of disease, but the majority of MS patients present with a relapsing-remitting disease course. In this study, we evaluated the therapeutic role of CD8 T cells in the context of relapsing-remitting disease (RR-EAE), using SJL mice. We found that PLP178-191- and MBP84-104-CD8 ameliorated disease severity in an antigen-specific manner. In contrast, PLP139-151-CD8 did not suppress disease. PLP178-191-CD8 were able to reduce the number of relapses even when transferred during ongoing disease. We further ascertained that the suppressive subset of CD8 T cells was contained within the CD25 + CD8 T cell compartment post-in vitro activation with PLP178-191. Using Listeria monocytogenes (LM) encoding CNS antigens to preferentially prime suppressive CD8 T cells in vivo, we show that LM infection induced disease suppressive CD8 T cells that protected and treated PLP178-191 disease. Importantly, a combination of PLP178-191-CD8 transfer boosted by LM-PLP175-194 infection effectively treated ongoing disease induced by a non-cognate peptide (PLP139-151), indicating that this approach could be effective even in the context of epitope spreading. These data support a potential immunotherapeutic strategy using CD8 transfer and/or LM vaccination to boost disease regulatory CD8 T cells. Adoptive transfer of CD8 T cells ameliorates RR-EAE in an antigen specific manner . Listeria monocytogenes (LM) can be used to prime disease-ameliorating CD8 T cells in RR-EAE . Activated CD25 + CD8 T cells preferentially harbor the disease-suppressive activity . Relapses in RR-EAE can be curbed using a dual strategy of CD8 T cell transfer and LM boosting .
Collapse
|
42
|
Alrashdi B, Dawod B, Schampel A, Tacke S, Kuerten S, Marshall JS, Côté PD. Nav1.6 promotes inflammation and neuronal degeneration in a mouse model of multiple sclerosis. J Neuroinflammation 2019; 16:215. [PMID: 31722722 PMCID: PMC6852902 DOI: 10.1186/s12974-019-1622-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In multiple sclerosis (MS) and in the experimental autoimmune encephalomyelitis (EAE) model of MS, the Nav1.6 voltage-gated sodium (Nav) channel isoform has been implicated as a primary contributor to axonal degeneration. Following demyelination Nav1.6, which is normally co-localized with the Na+/Ca2+ exchanger (NCX) at the nodes of Ranvier, associates with β-APP, a marker of neural injury. The persistent influx of sodium through Nav1.6 is believed to reverse the function of NCX, resulting in an increased influx of damaging Ca2+ ions. However, direct evidence for the role of Nav1.6 in axonal degeneration is lacking. METHODS In mice floxed for Scn8a, the gene that encodes the α subunit of Nav1.6, subjected to EAE we examined the effect of eliminating Nav1.6 from retinal ganglion cells (RGC) in one eye using an AAV vector harboring Cre and GFP, while using the contralateral either injected with AAV vector harboring GFP alone or non-targeted eye as control. RESULTS In retinas, the expression of Rbpms, a marker for retinal ganglion cells, was found to be inversely correlated to the expression of Scn8a. Furthermore, the gene expression of the pro-inflammatory cytokines Il6 (IL-6) and Ifng (IFN-γ), and of the reactive gliosis marker Gfap (GFAP) were found to be reduced in targeted retinas. Optic nerves from targeted eyes were shown to have reduced macrophage infiltration and improved axonal health. CONCLUSION Taken together, our results are consistent with Nav1.6 promoting inflammation and contributing to axonal degeneration following demyelination.
Collapse
Affiliation(s)
- Barakat Alrashdi
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Biology, Al-Jouf University, Sakaka, Saudi Arabia
| | - Bassel Dawod
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Andrea Schampel
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine Tacke
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Patrice D Côté
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
43
|
Rudolph J, Meinke C, Voss M, Guttek K, Kliche S, Reinhold D, Schraven B, Reinhold A. Immune Cell-Type Specific Ablation of Adapter Protein ADAP Differentially Modulates EAE. Front Immunol 2019; 10:2343. [PMID: 31632410 PMCID: PMC6779796 DOI: 10.3389/fimmu.2019.02343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/17/2019] [Indexed: 01/03/2023] Open
Abstract
The cytosolic adhesion and degranulation-promoting adapter protein ADAP is expressed in various hematopoietic cells including T cells, NK cells, myeloid cells, and platelets but absent in mature B cells. The role of ADAP in T cell activation, proliferation and integrin activation is well-accepted. We previously demonstrated that conventional ADAP knockout mice show a significantly attenuated course of experimental autoimmune encephalomyelitis (EAE). To dissect the impact of different ADAP expressing cell populations on the reduced EAE severity, here, we generated lineage-specific conditional knockout mice. ADAP was deleted in T cells, myeloid cells, NK cells and platelets, respectively. Specific loss of ADAP was confirmed on the protein level. Detailed immunophenotyping was performed to assess the consequence of deletion of ADAP with regard to the maturation and distribution of immune cells in primary and secondary lymphoid organs. The analysis showed equivalent results as for conventional ADAP knockout mice: impaired thymocyte development in ADAPfl/fl Lck-Cre mice, normal NK cell and myeloid cell distribution in ADAPfl/fl NKp46-Cre mice and ADAPfl/fl LysM-Cre mice, respectively as well as thrombocytopenia in ADAPfl/fl PF4-Cre mice. Active EAE was induced in these animals by immunization with the myelin oligodendrocyte glycoprotein35−55 peptide. The clinical course of EAE was significantly milder in mice with loss of ADAP in T cells, myeloid cells and NK cells compared to ADAP-sufficient control littermates. Surprisingly, specific deletion of ADAP in platelets resulted in a more exacerbated disease. These data show that T cell-independent as well as T cell-dependent mechanisms are responsible for the complex phenotype observed in conventional ADAP knockout mice.
Collapse
Affiliation(s)
- Jochen Rudolph
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Clara Meinke
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Martin Voss
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Karina Guttek
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Stefanie Kliche
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Dirk Reinhold
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Burkhart Schraven
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Annegret Reinhold
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| |
Collapse
|
44
|
Saito E, Kuo R, Kramer KR, Gohel N, Giles DA, Moore BB, Miller SD, Shea LD. Design of biodegradable nanoparticles to modulate phenotypes of antigen-presenting cells for antigen-specific treatment of autoimmune disease. Biomaterials 2019; 222:119432. [PMID: 31480002 DOI: 10.1016/j.biomaterials.2019.119432] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/18/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Current therapeutic options for autoimmune diseases, such as multiple sclerosis (MS), often require lifelong treatment with immunosuppressive drugs, yet strategies for antigen-specific immunomodulation are emerging. Biodegradable particles loaded with disease-specific antigen, either alone or with immunomodulators, have been reported to ameliorate disease. Herein, we hypothesized that the carrier could impact polarization of the immune cells that associate with particles and the subsequent disease progression. Single injection of three polymeric carriers, 50:50 poly (DL-lactide-co-glycolide) (PLG) with two molecular weights (Low, High) and poly (DL-lactide) (PLA), loaded with the disease-specific antigen, proteolipid protein (PLP139-151), were investigated for the ability to attenuate clinical scores in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. At a low particle dose, mice treated with PLA-based particles had significantly lower clinical scores at the chronic stage of the disease over 200 days post immunization, while neither PLG-based particles nor OVA control particles reduced the clinical scores. Compared to PLG-based particles, PLA-based particles were largely associated with Kupffer cells and liver sinusoidal endothelial cells, which had a reduced co-stimulatory molecule expression that correlated with a reduction of CD4+ T-cell populations in the central nervous system. Delivery of PLA-based particles encapsulated with higher levels of PLP139-151 at a reduced dose were able to completely ameliorate EAE over 200 days along with inhibition of Th1 and Th17 polarization. Collectively, our study demonstrates that the carrier properties and antigen loading determine phenotypes of immune cells in the peripheral organs, influencing the amelioration of both acute and chronic stages of autoimmunity.
Collapse
Affiliation(s)
- Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert Kuo
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kevin R Kramer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nishant Gohel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David A Giles
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Bethany B Moore
- Department of Immunology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, 60611, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
45
|
Abstract
SLAMF9 belongs to the conserved lymphocytic activation molecule family (SLAMF). Unlike other SLAMs, which have been extensively studied, the role of SLAMF9 in the immune system remained mostly unexplored. By generating CRISPR/Cas9 SLAMF9 knockout mice, we analyzed the role of this receptor in plasmacytoid dendritic cells (pDCs), which preferentially express the SLAMF9 transcript and protein. These cells display a unique capacity to produce type I IFN and bridge between innate and adaptive immune response. Analysis of pDCs in SLAMF9-/- mice revealed an increase of immature pDCs in the bone marrow and enhanced accumulation of pDCs in the lymph nodes. In the periphery, SLAMF9 deficiency resulted in lower levels of the transcription factor SpiB, elevation of pDC survival, and attenuated IFN-α and TNF-α production. To define the role of SLAMF9 during inflammation, pDCs lacking SLAMF9 were followed during induced experimental autoimmune encephalomyelitis. SLAMF9-/- mice demonstrated attenuated disease and delayed onset, accompanied by a prominent increase of immature pDCs in the lymph node, with a reduced costimulatory potential and enhanced infiltration of pDCs into the central nervous system. These results suggest the crucial role of SLAMF9 in pDC differentiation, homeostasis, and function in the steady state and during experimental autoimmune encephalomyelitis.
Collapse
|
46
|
Yoo IH, Kim MJ, Kim J, Sung JJ, Park ST, Ahn SW. The Anti-Inflammatory Effect of Sulforaphane in Mice with Experimental Autoimmune Encephalomyelitis. J Korean Med Sci 2019; 34:e197. [PMID: 31327180 PMCID: PMC6639507 DOI: 10.3346/jkms.2019.34.e197] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/02/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an immune-associated inflammatory disorder of the central nervous system and results in serious disability. Although many disease-modifying therapy drugs have been developed, these drugs have shown limited clinical efficacy and some adverse effects in previous studies, therefore, there has been reasonable need for less harmful and cost-effective therapeutics. Herein, we tested the anti-inflammatory effect of sulforaphane (SFN) in a mouse model of experimental autoimmune encephalomyelitis (EAE). METHODS The EAE mice were randomly assigned into two experimental groups: the phosphate-buffered saline (PBS)-treated EAE group and SFN-treated EAE group. After EAE mice induction by auto-immunization against the myelin oligodendrocyte glycoprotein peptide, we evaluated EAE symptom scores and biochemical analyses such as infiltration of inflammatory cells and demyelination of the spinal cord. Furthermore, western blotting was performed using the spinal cords of EAE mice. RESULTS In the behavioral study, the SFN-treated EAE mice showed favorable clinical scores compared with PBS-treated EAE mice at the 13th day (1.30 ± 0.15 vs. 1.90 ± 0.18; P = 0.043) and 14th day (1.80 ± 0.13 vs. 2.75 ± 0.17; P = 0.003). Additionally, the biochemical studies revealed that SFN treatment inhibited the inflammatory infiltration, demyelinating injury of the spinal cords, and the up-regulation of inducible nitric oxide synthase in the EAE mice. CONCLUSION The SFN treatment showed anti-inflammatory and anti-oxidative effects in the EAE mice. Conclusively, this study suggests that SFN has neuroprotective effects via anti-inflammatory processing, so it could be a new therapeutic or nutritional supplement for MS.
Collapse
Affiliation(s)
- Il Han Yoo
- Department of Neurology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Myung Jin Kim
- Department of Neurology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jiyoung Kim
- Center for Food and Bioconvergence, College of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Jung Joon Sung
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Taek Park
- Department of Obstetrics and Gynecology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Suk Won Ahn
- Department of Neurology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea.
| |
Collapse
|
47
|
Vaitaitis GM, Yussman MG, Wagner DH. A CD40 targeting peptide prevents severe symptoms in experimental autoimmune encephalomyelitis. J Neuroimmunol 2019; 332:8-15. [PMID: 30925295 PMCID: PMC6535109 DOI: 10.1016/j.jneuroim.2019.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 01/07/2023]
Abstract
CD40/CD154-interaction is critical in the development of Experimental Autoimmune Encephalomyelitis (EAE; mouse model of Multiple Sclerosis). Culprit CD4+CD40+ T cells drive a more severe form of EAE than conventional CD4 T cells. Blocking CD40/CD154-interaction with CD154-antibody prevents or ameliorates disease but had thrombotic complications in clinical trials. We targeted CD40 using a CD154-sequence based peptide. Peptides in human therapeutics demonstrate good safety. A small peptide, KGYY6, ameliorates EAE when given as pretreatment or at first symptoms. KGYY6 binds Th40 and memory T cells, affecting expression of CD69 and IL-10 in the CD4 T cell compartment, ultimately hampering disease development.
Collapse
Affiliation(s)
- Gisela M Vaitaitis
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Martin G Yussman
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - David H Wagner
- Webb-Waring Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
48
|
Zhou S, Liu G, Guo J, Kong F, Chen S, Wang Z. Pro-inflammatory Effect of Downregulated CD73 Expression in EAE Astrocytes. Front Cell Neurosci 2019; 13:233. [PMID: 31191254 PMCID: PMC6549520 DOI: 10.3389/fncel.2019.00233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 05/09/2019] [Indexed: 01/31/2023] Open
Abstract
CD73, an ectonucleotidase, participates in the regulation of immune responses by controlling the conversion of extracellular AMP to adenosine. In this study, we investigated whether any type of brain cells, especially neuroglia cells, exhibit altered CD73 expression, localization or activity upon experimental autoimmune uveitis (EAU) induction and whether altered CD73 manipulates the activation of effector T cells that interact with such cell types. First, the amount of cell membrane-exposed CD73 was detected by flow cytometry in various types of brain cells collected from either naïve or EAE mice. Compared to that in astrocytes from naïve control mice, the amount of membrane-bound CD73 was significantly decreased in astrocytes from EAE mice, while no significant differences were detected in other cell types. Thereafter, wild-type and CD73-/- astrocytes were used to study whether CD73 influences the function of inflammatory astrocytes, such as the production of cytokines/chemokines and the activation of effector T cells that interact with astrocytes. The results indicated that the addition of exogenous AMP significantly inhibited cytokine/chemokine production by wild type astrocytes but had no effect on CD73-/- astrocytes and that the effect of AMP was almost completely blocked by the addition of either a CD73 inhibitor (APCP) or an adenosine receptor A1 subtype (ARA1) antagonist (DPCPX). Although the addition of AMP did not affect CD73-/- astrocytes, the addition of adenosine successfully inhibited their cytokine/chemokine production. The antigen-specific interaction of astrocytes with invading CD4 cells caused CD73 downregulation in astrocytes from mice that underwent EAE induction. Collectively, our findings support the conclusion that, upon EAE induction, likely due to an interaction with invading CD4+ cells, astrocytes lose most of their membrane-localized CD73; this inhibits the generation of adenosine in the local microenvironment. As adenosine has anti-inflammatory effects on astrocytes and CNS-infiltrating effector T cells in EAE, the downregulation of CD73 in astrocytes may be considered a pro-inflammatory process for facilitating the pathogenesis of EAE.
Collapse
Affiliation(s)
- Shumin Zhou
- Clinical Laboratory, The 2nd Hospital of Tianjin Medical University, Tianjin, China
| | - Guoping Liu
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| | - Jie Guo
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| | - Fanqiang Kong
- Clinical Laboratory, General Hospital of Tianjin Medical University, Tianjin, China
| | - Song Chen
- Department of Ophthalmology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiyun Wang
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
49
|
Pickens CJ, Christopher MA, Leon MA, Pressnall MM, Johnson SN, Thati S, Sullivan BP, Berkland C. Antigen-Drug Conjugates as a Novel Therapeutic Class for the Treatment of Antigen-Specific Autoimmune Disorders. Mol Pharm 2019; 16:2452-2461. [PMID: 31083955 DOI: 10.1021/acs.molpharmaceut.9b00063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis represents the world's most common cause of neurological disability in young people and is attributed to a loss of immune tolerance toward proteins of the myelin sheath. Typical treatment options for MS patients involve immunomodulatory drugs, which act nonspecifically, resulting in global immunosuppression. The study discussed herein aims to demonstrate the efficacy of antigen-specific immunotherapies involving the conjugation of disease causing autoantigen, PLP139-151, and a potent immunosuppressant, dexamethasone. Antigen-drug conjugates (AgDCs) were formed using copper-catalyzed azide-alkyne cycloaddition chemistry with the inclusion of a hydrolyzable linker to maintain the activity of released dexamethasone. Subcutaneous administration of this antigen-drug conjugates to SJL mice induced with experimental autoimmune encephalomyelitis, protected the mice from a symptom onset throughout the 25 day study, demonstrating enhanced efficacy in comparison to dexamethasone treatment. These results highlight the benefits of co-delivery of autoantigens with immunosuppressant drugs as AgDCs for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Chad J Pickens
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - Martin A Leon
- Department of Chemistry , University of Kansas , 1567 Irving Hill Road , Lawrence , Kansas 66045 , United States
| | - Melissa M Pressnall
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - Stephanie N Johnson
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - Sharadvi Thati
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - Bradley P Sullivan
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - Cory Berkland
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| |
Collapse
|
50
|
Solouki S, August A, Huang W. Non-receptor tyrosine kinase signaling in autoimmunity and therapeutic implications. Pharmacol Ther 2019; 201:39-50. [PMID: 31082431 DOI: 10.1016/j.pharmthera.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/18/2019] [Indexed: 12/17/2022]
Abstract
Autoimmune diseases are characterized by impaired immune tolerance towards self-antigens, leading to enhanced immunity to self by dysfunctional B cells and/or T cells. The activation of these cells is controlled by non-receptor tyrosine kinases (NRTKs), which are critical mediators of antigen receptor and cytokine receptor signaling pathways. NRTKs transduce, amplify and sustain activating signals that contribute to autoimmunity, and are counter-regulated by protein tyrosine phosphatases (PTPs). The function of and interaction between NRTKs and PTPs during the development of autoimmunity could be key points of therapeutic interference against autoimmune diseases. In this review, we summarize the current state of knowledge of the functions of NRTKs and PTPs involved in B cell receptor (BCR), T cell receptor (TCR), and cytokine receptor signaling pathways that contribute to autoimmunity, and discuss their targeting for therapeutic approaches against autoimmune diseases.
Collapse
Affiliation(s)
- Sabrina Solouki
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|