1
|
Takase EO, Yamasaki R, Nagata S, Watanabe M, Masaki K, Yamaguchi H, Kira JI, Takeuchi H, Isobe N. Astroglial connexin 43 is a novel therapeutic target for chronic multiple sclerosis model. Sci Rep 2024; 14:10877. [PMID: 38740862 PMCID: PMC11091090 DOI: 10.1038/s41598-024-61508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
In chronic stages of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalitis (EAE), connexin (Cx)43 gap junction channel proteins are overexpressed because of astrogliosis. To elucidate the role of increased Cx43, the central nervous system (CNS)-permeable Cx blocker INI-0602 was therapeutically administered. C57BL6 mice with chronic EAE initiated by MOG35-55 received INI-0602 (40 mg/kg) or saline intraperitoneally every other day from days post-immunization (dpi) 17-50. Primary astroglia were employed to observe calcein efflux responses. In INI-0602-treated mice, EAE clinical signs improved significantly in the chronic phase, with reduced demyelination and decreased CD3+ T cells, Iba-1+ and F4/80+ microglia/macrophages, and C3+GFAP+ reactive astroglia infiltration in spinal cord lesions. Flow cytometry analysis of CD4+ T cells from CNS tissues revealed significantly reduced Th17 and Th17/Th1 cells (dpi 24) and Th1 cells (dpi 50). Multiplex array of cerebrospinal fluid showed significantly suppressed IL-6 and significantly increased IL-10 on dpi 24 in INI-0602-treated mice, and significantly suppressed IFN-γ and MCP-1 on dpi 50 in the same group. In vitro INI-0602 treatment inhibited ATP-induced calcium propagations of Cx43+/+ astroglial cells to similar levels of those of Cx43-/- cells. Astroglial Cx43 hemichannels represent a novel therapeutic target for chronic EAE and MS.
Collapse
Affiliation(s)
- Ezgi Ozdemir Takase
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroo Yamaguchi
- School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, Fukuoka, Japan
| | - Jun-Ichi Kira
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Ookawa, Japan
- Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, Fukuoka, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
- Department of Neurology, Graduate School of Medicine, International University of Health and Welfare, Narita, Japan.
- Center for Intractable Neurological Diseases and Dementia, International University of Health and Welfare Atami Hospital, Atami, Japan.
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
2
|
Gupta S, Mohindra R, Ramola M, Kanta P, Singla M, Malhotra M, Mehta N, Goyal A, Singh MP. Convergence of inflammatory response: Salivary cytokine dynamics in coronavirus disease 2019 and periodontal disease. J Indian Soc Periodontol 2024; 28:113-121. [PMID: 38988958 PMCID: PMC11232810 DOI: 10.4103/jisp.jisp_508_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
Background Periodontal disease is associated with immune dysregulation, and cytokines released can add on to the coronavirus disease 2019 (COVID-19)-associated cytokine storm, further worsening the related adverse outcomes. Specific studies investigating cytokine levels in COVID-19 patients with periodontal disease are lacking. Examining the correlation between these conditions could aid in categorizing risk categories, determining referrals, and strengthening oral hygiene protocols. The current study sought to evaluate cytokine levels in the saliva of COVID-19-positive patients with and without periodontal disease. Materials and Methods Twenty-six COVID-19-positive patients were subjected to periodontal examination, saliva collection, and assessment of cytokine levels through cytokine bead-based multiplex assay, using fluorescence-encoded beads with flow cytometry (BD FACS LSRFortessa). Eleven cytokines were assessed (interleukin [IL] 2, 4, 6, 10, 17A, and interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-α), chemokine ligand 2 (CCL2/monocyte chemoattractant protein-1), C-X-C motif chemokine ligand (CXCL) 8/IL 8, CXCL 9/monokine-induced gamma interferon [MIG]), and CXCL 10 (chemokine IFN-gamma inducible protein 10 kDa). The cytokine levels of the recruited subjects were also compared graphically with the salivary cytokine levels reported in the literature for health, COVID-19, and periodontal disease alone. Results Out of 26 COVID-19-positive patients, 17 had periodontal disease. Levels of all cytokines were raised in patients with both diseases when compared to values reported in literature for health, periodontal disease alone, or COVID-19 alone. However, there was no statistical difference among the recruited subjects for IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-gamma, TNF-α, CCL2, CXCL 8, and CXCL 10. MIG levels were found to be higher in periodontally healthy, COVID-19-positive subjects (P = 0.01). Conclusions Periodontal disease might contribute to the COVID-19-induced cytokine storm, potentially amplifying its impact.
Collapse
Affiliation(s)
- Shipra Gupta
- Oral Health Sciences Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ritin Mohindra
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Manisha Ramola
- Oral Health Sciences Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Poonam Kanta
- Department of Virology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Mohita Singla
- Oral Health Sciences Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Meenakshi Malhotra
- Department of Virology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Nishant Mehta
- Oral Health Sciences Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashima Goyal
- Oral Health Sciences Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Mini P Singh
- Department of Virology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
3
|
Schnell A, Littman DR, Kuchroo VK. T H17 cell heterogeneity and its role in tissue inflammation. Nat Immunol 2023; 24:19-29. [PMID: 36596896 PMCID: PMC10795475 DOI: 10.1038/s41590-022-01387-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/04/2022] [Indexed: 01/05/2023]
Abstract
Since their discovery almost two decades ago, interleukin-17-producing CD4+ T cells (TH17 cells) have been implicated in the pathogenesis of multiple autoimmune and inflammatory disorders. In addition, TH17 cells have been found to play an important role in tissue homeostasis, especially in the intestinal mucosa. Recently, the use of single-cell technologies, along with fate mapping and various mutant mouse models, has led to substantial progress in the understanding of TH17 cell heterogeneity in tissues and of TH17 cell plasticity leading to alternative T cell states and differing functions. In this Review, we discuss the heterogeneity of TH17 cells and the role of this heterogeneity in diverse functions of TH17 cells from homeostasis to tissue inflammation. In addition, we discuss TH17 cell plasticity and its incorporation into the current understanding of T cell subsets and alternative views on the role of TH17 cells in autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dan R Littman
- Department of Cell Biology and Regenerative Medicine, New York University School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
4
|
Keeler GD, Gaddie CD, Sagadevan AS, Senior KG, Côté I, Rechdan M, Min D, Mahan D, Poma B, Hoffman BE. Induction of antigen-specific tolerance by hepatic AAV immunotherapy regardless of T cell epitope usage or mouse strain background. Mol Ther Methods Clin Dev 2022; 28:177-189. [PMID: 36700122 PMCID: PMC9849872 DOI: 10.1016/j.omtm.2022.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
In vivo induction of antigen (Ag)-specific regulatory T cells (Treg) is considered the holy grail of therapeutic strategies for restoring tolerance in autoimmunity. Unfortunately, in the autoimmune disease multiple sclerosis, an effective and durable therapy targeting the diverse repertoire of emerging Ags without compromising the patient's natural immunity has remained elusive. To address this deficiency, we have developed an Ag-specific adeno-associated virus (AAV) immunotherapy that will restore tolerance in a Treg-dependent manner. Using multiple strains of mice with different genetic and immunological backgrounds, we demonstrate that a liver directed AAV vector expressing a single transgene can prevent experimental autoimmune encephalomyelitis from developing and effectively mitigate pre-existing or established disease that was induced by one or more auto-reactive myelin oligodendrocyte glycoprotein-derived peptides. Overall, the results suggests that AAV can efficiently restore Ag-specific immune tolerance to an immunogenic protein that is neither restricted by the major histocompatibility complex haplotype, nor by the specific antigenic epitope(s) presented. These findings may pave the way for developing a comprehensive Ag-specific immunotherapy that does not require prior knowledge of the specific immunogenic epitopes and that may prove to be universally applicable to all MS patients, and adaptable for other autoimmune diseases.
Collapse
Affiliation(s)
- Geoffrey D. Keeler
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Cristina D. Gaddie
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Addelynn S. Sagadevan
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA,Genetics Institute, University of Florida, Gainesville, FL 32610, USA
| | - Kevin G. Senior
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Isabelle Côté
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Michaela Rechdan
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Daniel Min
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - David Mahan
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Bianca Poma
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brad E. Hoffman
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA,Genetics Institute, University of Florida, Gainesville, FL 32610, USA,Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA,Corresponding author: Brad E. Hoffman, PhD, University of Florida, 2033 Mowry Road Office-207, Gainesville, FL 32610, USA.
| |
Collapse
|
5
|
Yarim GF, Yarim M, Sozmen M, Gokceoglu A, Ertekin A, Kabak YB, Karaca E. Nobiletin attenuates inflammation via modulating proinflammatory and antiinflammatory cytokine expressions in an autoimmune encephalomyelitis mouse model. Fitoterapia 2021; 156:105099. [PMID: 34896483 DOI: 10.1016/j.fitote.2021.105099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 11/17/2022]
Abstract
The aim of this study is to investigate the potential preventive and therapeutic effects of nobiletin by evaluating the expression of cytokines associated with inflammatory reactions in an autoimmune encephalomyelitis mouse model. A total of 60 male C57BL/6 mice aged between 8 and 10 weeks were used. Mice were divided into six groups (n = 10 mice per group): control, EAE, low-prophylaxis, high-prophylaxis, low-treatment and high-treatment. Experimental autoimmune encephalomyelitis (EAE) was induced by myelin oligodendrocyte glycoprotein (MOG) and pertussis toxin. Nobiletin was administered in low (25 mg/kg) and high (50 mg/kg) doses, intraperitoneally. The prophylactic and therapeutic effects of nobiletin on brain tissue and spinal cord were evaluated by expression of interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), interferon gamma (IFNγ), IL-6, IL-10 and transforming growth factor-beta (TGF-β) using immunohistochemistry and real-time polymerase chain reaction (RT-PCR). Prophylactic and therapeutic use of nobiletin inhibited EAE-induced increase of TNF-α, IL-1β and IL-6 activities to alleviate inflammatory response in brain and spinal cord. Moreover, nobiletin supplement dramatically increased the IL-10, TGF-β and IFNγ expressions in prophylaxis and treatment groups compared with the EAE group in the brain and spinal cord. The results obtained from this study show that prophylactic and therapeutic nobiletin modulates expressions of proinflammatory and antiinflammatory cytokines in brain and spinal cord dose-dependent manner in EAE model. These data demonstrates that nobiletin has a potential to attenuate inflammation in EAE mouse model. These experimental findings need to be supported by clinical studies.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Antioxidants/therapeutic use
- Brain/drug effects
- Brain/immunology
- Brain/pathology
- Cytokines/drug effects
- Cytokines/metabolism
- DNA, Complementary/biosynthesis
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Flavones/pharmacology
- Flavones/therapeutic use
- Immunohistochemistry
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/prevention & control
- Male
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/prevention & control
- RNA/genetics
- RNA/isolation & purification
- Real-Time Polymerase Chain Reaction
- Spinal Cord/drug effects
- Spinal Cord/immunology
- Spinal Cord/pathology
Collapse
Affiliation(s)
- Gul Fatma Yarim
- Department of Biochemistry, Faculty of Veterinary Medicine, Ondokuz Mayis University, Atakum, 55200 Samsun, Turkey.
| | - Murat Yarim
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Atakum, 55200 Samsun, Turkey
| | - Mahmut Sozmen
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Atakum, 55200 Samsun, Turkey
| | - Ayris Gokceoglu
- Department of Biochemistry, Faculty of Veterinary Medicine, Ondokuz Mayis University, Atakum, 55200 Samsun, Turkey
| | - Ali Ertekin
- Department of Biochemistry, Faculty of Veterinary Medicine, Ondokuz Mayis University, Atakum, 55200 Samsun, Turkey
| | - Yonca Betil Kabak
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Atakum, 55200 Samsun, Turkey
| | - Efe Karaca
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Atakum, 55200 Samsun, Turkey
| |
Collapse
|
6
|
Aboushanab SA, Khedr SM, Gette IF, Danilova IG, Kolberg NA, Ravishankar GA, Ambati RR, Kovaleva EG. Isoflavones derived from plant raw materials: bioavailability, anti-cancer, anti-aging potentials, and microbiome modulation. Crit Rev Food Sci Nutr 2021; 63:261-287. [PMID: 34251921 DOI: 10.1080/10408398.2021.1946006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Isoflavones are secondary metabolites that represent the most abundant category of plant polyphenols. Dietary soy, kudzu, and red clover contain primarily genistein, daidzein, glycitein, puerarin, formononetin, and biochanin A. The structural similarity of these compounds to β-estradiol has demonstrated protection against age-related and hormone-dependent diseases in both genders. Demonstrative shreds of evidence confirmed the fundamental health benefits of the consumption of these isoflavones. These relevant activities are complex and largely driven by the source, active ingredients, dose, and administration period of the bioactive compounds. However, the preclinical and clinical studies of these compounds are greatly variable, controversial, and still with no consensus due to the non-standardized research protocols. In addition, absorption, distribution, metabolism, and excretion studies, and the safety profile of isoflavones have been far limited. This highlights a major gap in understanding the potentially critical role of these isoflavones as prospective replacement therapy. Our general review exclusively focuses attention on the crucial role of isoflavones derived from these plant materials and critically highlights their bioavailability, possible anticancer, antiaging potentials, and microbiome modulation. Despite their fundamental health benefits, plant isoflavones reveal prospective therapeutic effects that worth further standardized analysis.
Collapse
Affiliation(s)
- Saied A Aboushanab
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia
| | - Shaimaa M Khedr
- Pharmaceutical and Fermentation Industries Development Center (PFIDC), City of Scientific Research and Technological Applications, SRTA-City, Alexandria, Egypt
| | - Irina F Gette
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia.,Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Irina G Danilova
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia.,Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Natalia A Kolberg
- Integrated Laboratory Complex, Ural State University of Economics, Yekaterinburg, Russia
| | - Gokare A Ravishankar
- C. D. Sagar Centre for Life Sciences, Dayananda Sagar College of Engineering, Dayananda Sagar Institutions, Bangalore, Karnataka, India
| | - Ranga Rao Ambati
- Department of Biotechnology, Vignan's Foundation of Science, Technology and Research, Guntur, Andhra Pradesh, India
| | - Elena G Kovaleva
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia
| |
Collapse
|
7
|
Wang G, Zhang H, Sun J, Zhang Y, He F, Zou J. Cyclosporin A impairs neurogenesis and cognitive abilities in brain development via the IFN-γ-Shh-BDNF pathway. Int Immunopharmacol 2021; 96:107744. [PMID: 33993101 DOI: 10.1016/j.intimp.2021.107744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 01/07/2023]
Abstract
A wealth of evidence indicate that the peripheral immune activation alters brain development. However, it is still largely unclear whether and how peripheral immunosuppression affects neurodevelopment. Here, we found that the immunosuppressant cyclosporin A (CsA) decreased the number of BrdU+, BrdU+/DCX+, BrdU+/NeuN + cells in the hippocampus, impaired learning and memory and inhibited protein levels of the shh signaling pathway, including Shh, Smo and Gli1. However, the shh pathway receptor agonist SAG could block the impairment of cognitive ability and the decrease of hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) level induced by CsA. We also found that CsA decreased the level of interferon-gamma (IFN-γ), while up-regulation of IFN-γ altered the inhibitory effect of the shh signaling pathway and the decrease of BDNF induced by CsA. Collectively, these data indicate that peripheral CsA impairs neurogenesis and cognition in brain development through downregulating the IFN-γ-Shh-BDNF pathway. The present study guides us to correctly apply immunomodulatory drugs in early life and suggests that the IFN-γ-Shh-BDNF pathway may represent a novel protective target for neurodevelopment under the condition of immunosuppression.
Collapse
Affiliation(s)
- Ge Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200032 Shanghai, People's Republic of China
| | - Hongyang Zhang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiancong Sun
- Department of Radiation Oncology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yuwei Zhang
- Department of Anatomy, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Fen He
- Department of Radiation Oncology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
8
|
Wagner CA, Roqué PJ, Goverman JM. Pathogenic T cell cytokines in multiple sclerosis. J Exp Med 2020; 217:jem.20190460. [PMID: 31611252 PMCID: PMC7037255 DOI: 10.1084/jem.20190460] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/21/2019] [Accepted: 09/11/2019] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the central nervous system that is believed to have an autoimmune etiology. As MS is the most common nontraumatic disease that causes disability in young adults, extensive research has been devoted to identifying therapeutic targets. In this review, we discuss the current understanding derived from studies of patients with MS and animal models of how specific cytokines produced by autoreactive CD4 T cells contribute to the pathogenesis of MS. Defining the roles of these cytokines will lead to a better understanding of the potential of cytokine-based therapies for patients with MS.
Collapse
Affiliation(s)
| | - Pamela J Roqué
- Department of Immunology, University of Washington, Seattle, WA
| | - Joan M Goverman
- Department of Immunology, University of Washington, Seattle, WA
| |
Collapse
|
9
|
Zheng C, Chen J, Chu F, Zhu J, Jin T. Inflammatory Role of TLR-MyD88 Signaling in Multiple Sclerosis. Front Mol Neurosci 2020; 12:314. [PMID: 31998072 PMCID: PMC6965019 DOI: 10.3389/fnmol.2019.00314] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a neuro-autoimmune and neurodegenerative disorder leading to chronic inflammation, demyelination, axonal, and neuronal loss in the central nervous system (CNS). Despite intense research efforts, the pathogenesis of MS still remains unclear. Toll-like receptors (TLRs) are a family of type I transmembrane receptors that play a crucial role in the innate immune response. Myeloid differentiation factor 88 (MyD88) is the adaptor of major TLRs. It has been widely considered that the TLR-MyD88 signaling pathway plays an important role in the occurrence and development of autoimmune disease. Data have revealed that the TLR-MyD88 signaling may be involved in the pathogenesis of MS and experimental autoimmune encephalomyelitis (EAE), an animal model for MS, by regulating the antigen presentation of dendritic cells, the integrity of blood-brain barrier (BBB), and the activation of T cells and B cells. Here, we summarize the role of TLRs and MyD88 in MS and discuss the possible therapies that are based on these molecules.
Collapse
Affiliation(s)
- Chao Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fengna Chu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Yang MG, Sun L, Han J, Zheng C, Liang H, Zhu J, Jin T. Biological characteristics of transcription factor RelB in different immune cell types: implications for the treatment of multiple sclerosis. Mol Brain 2019; 12:115. [PMID: 31881915 PMCID: PMC6935142 DOI: 10.1186/s13041-019-0532-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/04/2019] [Indexed: 12/22/2022] Open
Abstract
Transcription factor RelB is a member of the nuclear factror-kappa B (NF-κB) family, which plays a crucial role in mediating immune responses. Plenty of studies have demonstrated that RelB actively contributes to lymphoid organ development, dendritic cells maturation and function and T cells differentiation, as well as B cell development and survival. RelB deficiency may cause a variety of immunological disorders in both mice and humans. Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system which involves a board of immune cell populations. Thereby, RelB may exert an impact on MS by modulating the functions of dendritic cells and the differentiation of T cells and B cells. Despite intensive research, the role of RelB in MS and its animal model, experimental autoimmune encephalomyelitis, is still unclear. Herein, we give an overview of the biological characters of RelB, summarize the updated knowledge regarding the role of RelB in different cell types that contribute to MS pathogenesis and discuss the potential RelB-targeted therapeutic implications for MS.
Collapse
Affiliation(s)
- Meng-Ge Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.,Present address: Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Chao Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Hudong Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| |
Collapse
|
11
|
Abstract
Helicobacter pylori is a Gram-negative bacterium that infects the gastric epithelia of its human host. Everyone who is colonized with these pathogenic bacteria can develop gastric inflammation, termed gastritis. Additionally, a small proportion of colonized people develop more adverse outcomes, including gastric ulcer disease, gastric adenocarcinoma, or gastric mucosa-associated lymphoid tissue lymphoma. The development of these adverse outcomes is dependent on the establishment of a chronic inflammatory response. The development and control of this chronic inflammatory response are significantly impacted by CD4+ T helper cell activity. Noteworthy, T helper 17 (Th17) cells, a proinflammatory subset of CD4+ T cells, produce several proinflammatory cytokines that activate innate immune cell antimicrobial activity, drive a pathogenic immune response, regulate B cell responses, and participate in wound healing. Therefore, this review was written to take an intricate look at the involvement of Th17 cells and their affiliated cytokines (interleukin-17A [IL-17A], IL-17F, IL-21, IL-22, and IL-26) in regulating the immune response to H. pylori colonization and carcinogenesis.
Collapse
|
12
|
Sarmirova S, Borsanyiova M, Benkoova B, Pospisilova M, Arumugam R, Berakova K, Gomolcak P, Reddy J, Bopegamage S. Pancreas of coxsackievirus-infected dams and their challenged pups: A complex issue. Virulence 2019; 10:207-221. [PMID: 30829107 PMCID: PMC6550550 DOI: 10.1080/21505594.2019.1589364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Enteroviral infections are frequent, often asymptomatic in humans and during gravidity. The present study is an extension of our previous investigations where we had shown pancreatitis in challenged pups of CVB4-E2-infected dams. Present investigation describes the effect of gestational infection with this virus on the pancreas of both dams and their challenged pups. Gravid CD1 outbred mice were orally infected with CVB4-E2 virus at different gestation times. Pups were challenged orally with the same virus after 25 days of birth. Organs were collected at selected intervals postinfection (p.i.), and replicating virus and viral-RNA copies were analyzed. Additional readouts included histopathology and immunohistochemical (IHC) analysis for localization and identification of Ly6G+ cells (neutrophils), CD11b+ cells (macrophages), and viral protein in pancreatic tissue sections of the infected dams and their challenged pups. Our results show the presence of replicating virus in the pancreas of infected dams and their challenged pups, with inflammation leading to chronic necrotizing pancreatitis and atrophy of pancreatic acini of the dams and their offspring. IHC analysis of the infiltrating cells showed pronounced Ly6G+ neutrophils in dams only, whereas CD11b+ macrophages were present in tissues of both, the pups and the dams. Time of infection during gravidity as well as the p.i. intervals when mice were sacrificed influenced the pancreatic pathophysiology in both groups. We conclude that coxsackievirus infection during pregnancy is a risk factor for chronic affliction of the exocrine tissue and could affect endocrine pancreas in the mother and child.
Collapse
Affiliation(s)
- Sona Sarmirova
- a Enterovirus Laboratory, Institute of Microbiology , Faculty of Medicine, Slovak Medical University , Bratislava , Slovak Republic
| | - Maria Borsanyiova
- a Enterovirus Laboratory, Institute of Microbiology , Faculty of Medicine, Slovak Medical University , Bratislava , Slovak Republic
| | - Brigita Benkoova
- a Enterovirus Laboratory, Institute of Microbiology , Faculty of Medicine, Slovak Medical University , Bratislava , Slovak Republic
| | - Michaela Pospisilova
- a Enterovirus Laboratory, Institute of Microbiology , Faculty of Medicine, Slovak Medical University , Bratislava , Slovak Republic
| | - Rajkumar Arumugam
- b School of Veterinary Medicine and Biomedical Sciences , University of Nebraska-Lincoln , Lincoln , NE , USA
| | | | - Pavol Gomolcak
- d Immunohistochemical Laboratory, Medical Laboratory Pathology and Cytology , Cytopathos, s.r.o , Bratislava , Slovak Republic
| | - Jay Reddy
- b School of Veterinary Medicine and Biomedical Sciences , University of Nebraska-Lincoln , Lincoln , NE , USA
| | - Shubhada Bopegamage
- a Enterovirus Laboratory, Institute of Microbiology , Faculty of Medicine, Slovak Medical University , Bratislava , Slovak Republic
| |
Collapse
|
13
|
Katsel P, Haroutunian V. Is Alzheimer disease a failure of mobilizing immune defense? Lessons from cognitively fit oldest-old. DIALOGUES IN CLINICAL NEUROSCIENCE 2019. [PMID: 31607776 PMCID: PMC6780355 DOI: 10.31887/dcns.2019.21.1/vharoutunian] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Multifaceted evidence supports the hypothesis that inflammatory-immune mechanisms contribute to Alzheimer disease (AD) neuropathology and genetic association of several immune specific genes (TREM2, CR1, and CD33) suggests that maladaptive immune responses may be pivotal drivers of AD pathogenesis. We reviewed microglia-related data from postmortem AD studies and examined supporting evidence from AD animal models to answer the following questions: i) What is the temporal sequence of immune activation in AD progression and what is its impact on cognition? ii) Are there discordant, “primed”, microglia responses in AD vs successful cognitive aging? iii) Does central nervous system (CNS) repair in aging depend on recruitment of the elements of cellular adaptive immune response such as effector T cells, and can the recruitment of systemic immune cells ameliorate AD neuropathology? iv) How effective are the immune-system-based therapeutic approaches currently employed for the treatment of AD?
Collapse
Affiliation(s)
- Pavel Katsel
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vahram Haroutunian
- Department of Neuroscience, The Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
14
|
Mahmoodi M, Amiri H, Ayoobi F, Rahmani M, Taghipour Z, Ghavamabadi RT, Jafarzadeh A, Sankian M. Carvacrol ameliorates experimental autoimmune encephalomyelitis through modulating pro- and anti-inflammatory cytokines. Life Sci 2019; 219:257-263. [DOI: 10.1016/j.lfs.2018.11.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 11/17/2022]
|
15
|
The Diversity of Encephalitogenic CD4+ T Cells in Multiple Sclerosis and Its Animal Models. J Clin Med 2019; 8:jcm8010120. [PMID: 30669462 PMCID: PMC6352150 DOI: 10.3390/jcm8010120] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 02/06/2023] Open
Abstract
Autoreactive CD4+ T cells, which target antigens in central nervous system (CNS) myelin, are widely believed to play a critical role in the pathogenesis of multiple sclerosis (MS) in concert with other immune effectors. This theory is supported by data from animal model experiments, genome-wide association studies, and immune profiles of individuals with MS. Furthermore, disease modifying agents that target lymphocytes significantly reduce the rate of MS clinical exacerbations. However, the properties of myelin-reactive CD4+ T cells that are critical for their pathogenic activities are not understood completely. This article reviews the literature on encephalitogenic CD4+ T cells, with an emphasis on T-helper (Th) lineage and cytokine production. An increased understanding of the spectrum of encephalitogenic T cells and how they differ from protective subsets is necessary for the development of the next generation of more effective and safer immunomodulatory therapies customized for individuals with MS and related disorders.
Collapse
|
16
|
Savarin C, Bergmann CC. Fine Tuning the Cytokine Storm by IFN and IL-10 Following Neurotropic Coronavirus Encephalomyelitis. Front Immunol 2018; 9:3022. [PMID: 30619363 PMCID: PMC6306494 DOI: 10.3389/fimmu.2018.03022] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
The central nervous system (CNS) is vulnerable to several viral infections including herpes viruses, arboviruses and HIV to name a few. While a rapid and effective immune response is essential to limit viral spread and mortality, this anti-viral response needs to be tightly regulated in order to limit immune mediated tissue damage. This balance between effective virus control with limited pathology is especially important due to the highly specialized functions and limited regenerative capacity of neurons, which can be targets of direct virus cytolysis or bystander damage. CNS infection with the neurotropic strain of mouse hepatitis virus (MHV) induces an acute encephalomyelitis associated with focal areas of demyelination, which is sustained during viral persistence. Both innate and adaptive immune cells work in coordination to control virus replication. While type I interferons are essential to limit virus spread associated with early mortality, perforin, and interferon-γ promote further virus clearance in astrocytes/microglia and oligodendrocytes, respectively. Effective control of virus replication is nonetheless associated with tissue damage, characterized by demyelinating lesions. Interestingly, the anti-inflammatory cytokine IL-10 limits expansion of tissue lesions during chronic infection without affecting viral persistence. Thus, effective coordination of pro- and anti-inflammatory cytokines is essential during MHV induced encephalomyelitis in order to protect the host against viral infection at a limited cost.
Collapse
Affiliation(s)
- Carine Savarin
- Department of Neuroscience, Cleveland Clinic Foundation, Lerner Research Institute, Cleveland, OH, United States
| | - Cornelia C Bergmann
- Department of Neuroscience, Cleveland Clinic Foundation, Lerner Research Institute, Cleveland, OH, United States
| |
Collapse
|
17
|
Meyer Zu Horste G, Przybylski D, Schramm MA, Wang C, Schnell A, Lee Y, Sobel R, Regev A, Kuchroo VK. Fas Promotes T Helper 17 Cell Differentiation and Inhibits T Helper 1 Cell Development by Binding and Sequestering Transcription Factor STAT1. Immunity 2018; 48:556-569.e7. [PMID: 29562202 DOI: 10.1016/j.immuni.2018.03.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/28/2018] [Accepted: 03/02/2018] [Indexed: 12/21/2022]
Abstract
The death receptor Fas removes activated lymphocytes through apoptosis. Previous transcriptional profiling predicted that Fas positively regulates interleukin-17 (IL-17)-producing T helper 17 (Th17) cells. Here, we demonstrate that Fas promoted the generation and stability of Th17 cells and prevented their differentiation into Th1 cells. Mice with T-cell- and Th17-cell-specific deletion of Fas were protected from induced autoimmunity, and Th17 cell differentiation and stability were impaired. Fas-deficient Th17 cells instead developed a Th1-cell-like transcriptional profile, which a new algorithm predicted to depend on STAT1. Experimentally, Fas indeed bound and sequestered STAT1, and Fas deficiency enhanced IL-6-induced STAT1 activation and nuclear translocation, whereas deficiency of STAT1 reversed the transcriptional changes induced by Fas deficiency. Thus, our computational and experimental approach identified Fas as a regulator of the Th17-to-Th1 cell balance by controlling the availability of opposing STAT1 and STAT3 to have a direct impact on autoimmunity.
Collapse
Affiliation(s)
- Gerd Meyer Zu Horste
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Department of Neurology, University Hospital Münster, Münster, Germany
| | | | - Markus A Schramm
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Chao Wang
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Youjin Lee
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Raymond Sobel
- Palo Alto Veteran's Administration Health Care System and Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA; Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
18
|
Liu HJ, Chen G, Chen L, Zhou M, Xiong XZ, Meng ZJ, Sun SW, Tao XN. Cytokine-induced alterations of BAMBI mediate the reciprocal regulation of human Th17/Treg cells in response to cigarette smoke extract. Int J Mol Med 2018; 42:3404-3414. [PMID: 30320351 PMCID: PMC6202106 DOI: 10.3892/ijmm.2018.3919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 10/01/2018] [Indexed: 11/06/2022] Open
Abstract
In CD4+ T helper (Th) cells, transforming growth factor β (TGF‑β) is indispensable for the induction of both regulatory T (Treg) and interleukin‑17‑producing effector T helper (Th17) cells. Although BMP and activin membrane‑bound inhibitor (BAMBI) is part of a rheostat‑like mechanism for the regulation of TGF‑β signalling and autoimmune arthritis in mouse models, the underlying activity of BAMBI on the human Th17/Treg cell axis, particularly during exposure to cigarette smoke, remains to be elucidated. The present study aimed to further characterize BAMBI expression in human CD4+ cells, as well as immune imbalance during activation and cigarette smoke exposure. Results from the present study indicated that exposure to cigarette smoke extract partially suppressed Treg differentiation and promoted Th17 cell generation under stimulation by anti‑CD3/28 antibodies and TGF‑β1. Additionally, exposure to cigarette smoke induced an inhibition of phosphorylated‑Smad2/Smad3, which may have arisen from a concomitant enhancement of BAMBI expression. In conclusion, human BAMBI may function as a molecular switch to control TGF‑β signalling strength and the Th17/Treg cell balance, which may be used not only as a biomarker but also as a target of new treatment strategies for maintaining immune tolerance and for the treatment of smoking‑induced immune disorders.
Collapse
Affiliation(s)
- Hong-Ju Liu
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Gang Chen
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Long Chen
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Mei Zhou
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xian-Zhi Xiong
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhao-Ji Meng
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Sheng-Wen Sun
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiao-Nan Tao
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
19
|
Peer S, Cappellano G, Hermann-Kleiter N, Albrecht-Schgoer K, Hinterleitner R, Baier G, Gruber T. Regulation of Lymphatic GM-CSF Expression by the E3 Ubiquitin Ligase Cbl-b. Front Immunol 2018; 9:2311. [PMID: 30349541 PMCID: PMC6186797 DOI: 10.3389/fimmu.2018.02311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022] Open
Abstract
Genome-wide association studies as well as lymphatic expression analyses have linked both Cbl-b and GM-CSF to human multiple sclerosis as well as other autoimmune diseases. Both Cbl-b and GM-CSF have been shown to play a prominent role in the development of murine encephalomyelitis; however, no functional connection between the two has yet been established. In this study, we show that Cblb knockout mice demonstrated significantly exacerbated severity of experimental autoimmune encephalomyelitis (EAE), augmented T cell infiltration into the central nervous system (CNS) and strongly increased production of GM-CSF in T cells in vitro and in vivo.GM-CSF neutralization demonstrated that the increased susceptibility of Cblb−/− mice to EAE was dependent on GM-CSF. Mechanistically, p50 binding to the GM-CSF promoter and the IL-3/GM-CSF enhancer element “CNSa” was strongly increased in nuclear extracts from Cbl-b-deficient T cells. This study suggests that Cbl-b limits autoimmunity by preventing the pathogenic effects of GM-CSF overproduction in T cells.
Collapse
Affiliation(s)
- Sebastian Peer
- Division of Translational Cell Genetics, Department for Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Giuseppe Cappellano
- Division of Translational Cell Genetics, Department for Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Division of Translational Cell Genetics, Department for Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Karin Albrecht-Schgoer
- Division of Translational Cell Genetics, Department for Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinhard Hinterleitner
- Division of Translational Cell Genetics, Department for Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Gottfried Baier
- Division of Translational Cell Genetics, Department for Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Gruber
- Division of Translational Cell Genetics, Department for Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
20
|
Qi F, Zuo Z, Hu S, Xia Y, Song D, Kong J, Yang Y, Wu Y, Wang X, Yang J, Hu D, Yuan Q, Zou J, Guo K, Xu J, Yao Z. An enriched environment restores hepatitis B vaccination-mediated impairments in synaptic function through IFN-γ/Arginase1 signaling. Brain Behav Immun 2018; 71:116-132. [PMID: 29627530 DOI: 10.1016/j.bbi.2018.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/06/2018] [Accepted: 04/04/2018] [Indexed: 12/28/2022] Open
Abstract
Activation of the neonatal immune system may contribute to deficits in neuronal plasticity. We have reported that neonatal vaccination with a hepatitis B vaccine (HBV) transiently impairs mood status and spatial memory involving a systemic T helper (Th) 2 bias and M1 microglial activation. Here, an EE induced microglial anti-inflammatory M2 polarization, as evidenced by selectively enhanced expression of the Arginase1 gene (Arg-1) in the hippocampus. Interestingly, knock-down of the Arg-1 gene prevented the effects of EE on restoring the dendritic spine density. Moreover, levels of the Th1-derived cytokine IFN-gamma (IFN-γ) were elevated in the choroid plexus (CP), which is the interface between the brain and the periphery. IFN-γ-blocking antibodies blunted the protective effects of an EE on spine density and LTP. Furthermore, levels of complement proteins C1q and C3 were elevated, and this elevation was associated with synapse loss induced by the HBV, whereas an EE reversed the effects of the HBV. Similarly, blockade of C1q activation clearly prevented synaptic pruning by microglia, LTP inhibition and memory deficits in hepatitis B-vaccinated mice. Together, the EE-induced increase in IFN-γ levels in the CP may disrupt systemic immunosuppression related to HBV via an IFN-γ/Arg-1/complement-dependent pathway.
Collapse
Affiliation(s)
- Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Saisai Hu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Yucen Xia
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, 232 Waihuandong Road, Guangzhou 510006, PR China
| | - Dan Song
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Jiechen Kong
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Yang Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Yingying Wu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Dandan Hu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Department of Neurology, Guangzhou Women and Children's Hospital, #9, Jinsui Road, Guangzhou 510623, PR China
| | - Qunfang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Kaihua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou 510080, PR China.
| |
Collapse
|
21
|
Ramakrishna C, Cantin EM. IFNγ inhibits G-CSF induced neutrophil expansion and invasion of the CNS to prevent viral encephalitis. PLoS Pathog 2018; 14:e1006822. [PMID: 29352287 PMCID: PMC5792029 DOI: 10.1371/journal.ppat.1006822] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/31/2018] [Accepted: 12/16/2017] [Indexed: 12/12/2022] Open
Abstract
Emergency hematopoiesis facilitates the rapid expansion of inflammatory immune cells in response to infections by pathogens, a process that must be carefully regulated to prevent potentially life threatening inflammatory responses. Here, we describe a novel regulatory role for the cytokine IFNγ that is critical for preventing fatal encephalitis after viral infection. HSV1 encephalitis (HSE) is triggered by the invasion of the brainstem by inflammatory monocytes and neutrophils. In mice lacking IFNγ (GKO), we observed unrestrained increases in G-CSF levels but not in GM-CSF or IL-17. This resulted in uncontrolled expansion and infiltration of apoptosis-resistant, degranulating neutrophils into the brainstem, causing fatal HSE in GKO but not WT mice. Excessive G-CSF in GKO mice also induced granulocyte derived suppressor cells, which inhibited T-cell proliferation and function, including production of the anti-inflammatory cytokine IL-10. Unexpectedly, we found that IFNγ suppressed G-CSF signaling by increasing SOCS3 expression in neutrophils, resulting in apoptosis. Depletion of G-CSF, but not GM-CSF, in GKO mice induced neutrophil apoptosis and reinstated IL-10 secretion by T cells, which restored their ability to limit innate inflammatory responses resulting in protection from HSE. Our studies reveals a novel, complex interplay among IFNγ, G-CSF and IL-10, which highlights the opposing roles of G-CSF and IFNγ in regulation of innate inflammatory responses in a murine viral encephalitis model and reveals G-CSF as a potential therapeutic target. Thus, the antagonistic G-CSF-IFNγ interactions emerge as a key regulatory node in control of CNS inflammatory responses to virus infection.
Collapse
Affiliation(s)
- Chandran Ramakrishna
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California, United States of America
- * E-mail: (CR); (EMC)
| | - Edouard M. Cantin
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California, United States of America
- * E-mail: (CR); (EMC)
| |
Collapse
|
22
|
Ghafelehbashi H, Pahlevan Kakhki M, Kular L, Moghbelinejad S, Ghafelehbashi SH. Decreased Expression of IFNG-AS1
,IFNG
and IL-1B
Inflammatory Genes in Medicated Schizophrenia and Bipolar Patients. Scand J Immunol 2017; 86:479-485. [DOI: 10.1111/sji.12620] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/09/2017] [Indexed: 01/22/2023]
Affiliation(s)
- H. Ghafelehbashi
- Cellular and Molecular Research Center; Qazvin University of Medical Sciences; Qazvin Iran
| | - M. Pahlevan Kakhki
- Department of Genetics; Faculty of Biological Sciences; Tarbiat Modares University; Tehran Iran
| | - L. Kular
- Department of Clinical Neuroscience; Center for Molecular Medicine; Karolinska Institute; Karolinska University Hospital; Stockholm Sweden
| | - S. Moghbelinejad
- Cellular and Molecular Research Center; Qazvin University of Medical Sciences; Qazvin Iran
- Department of Psychiatry; Qazvin University of Medical Sciences; Qazvin Iran
| | - S. H. Ghafelehbashi
- Cellular and Molecular Research Center; Qazvin University of Medical Sciences; Qazvin Iran
- Department of Psychiatry; Qazvin University of Medical Sciences; Qazvin Iran
| |
Collapse
|
23
|
Minchenberg SB, Massa PT. The control of oligodendrocyte bioenergetics by interferon-gamma (IFN-γ) and Src homology region 2 domain-containing phosphatase-1 (SHP-1). J Neuroimmunol 2017; 331:46-57. [PMID: 29113698 DOI: 10.1016/j.jneuroim.2017.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/17/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
Glycolysis and mitochondrial respiration are essential for oligodendrocyte metabolism in both the developing and adult CNS. Based on recent reports on the effects of the proinflammatory cytokine IFN-γ on metabolism and on oligodendrocytes, we addressed whether IFN-γ may affect oligodendrocyte bioenergetics in ways relevant to CNS disease. Oligodendrocytes of mice treated with IFN-γ showed significant reductions in aerobic glycolysis and mitochondrial respiration. As expected, IFN-γ treatment led to the induction of STAT1 in oligodendrocytes indicating active signaling into these cells. To determine the direct effects of IFN-γ on oligodendrocyte metabolism, cultured oligodendrocytes were treated with IFN-γ in vitro, which resulted in suppression of glycolysis similar to oligodendrocytes of animals treated with IFN-γ in vivo. Mice lacking SHP-1, a key regulator of IFN-γ and STAT1 signaling in CNS glia, had high constitutive levels of STAT1 and decreased aerobic glycolysis and mitochondrial respiration rates relative to wild type mouse oligodendrocytes. Together, these data show that IFN-γ and SHP-1 control oligodendrocyte bioenergetics in ways that may relate to the role of this cytokine in CNS disease.
Collapse
Affiliation(s)
- Scott B Minchenberg
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, United States
| | - Paul T Massa
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, United States; Department of Neurology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, United States.
| |
Collapse
|
24
|
Khakzad MR, Ganji A, Ariabod V, Farahani I. Artemisinin therapeutic efficacy in the experimental model of multiple sclerosis. Immunopharmacol Immunotoxicol 2017; 39:348-353. [DOI: 10.1080/08923973.2017.1379087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mohammad Reza Khakzad
- Innovative Medical Research Center, Department of Immunology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
- Allergy Research Center, Mashhad, University of Medical Sciences, Mashhad, Iran
| | - Ali Ganji
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
- Department of Microbiology and Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Vahid Ariabod
- Department of Pathology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Iman Farahani
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
- Department of Microbiology and Immunology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
25
|
Casserly CS, Nantes JC, Whittaker Hawkins RF, Vallières L. Neutrophil perversion in demyelinating autoimmune diseases: Mechanisms to medicine. Autoimmun Rev 2017; 16:294-307. [DOI: 10.1016/j.autrev.2017.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
|
26
|
Abstract
TGF-β is an anti-inflammatory cytokine whose signaling is negatively controlled by Smad7. Previously, we established a role for Smad7 in the generation of autoreactive T cells; however, the function of Smad7 in dendritic cells (DCs) remains elusive. Here, we demonstrate that DC-specific Smad7 deficiency resulted in elevated expression of the transcription factors Batf3 and IRF8, leading to increased frequencies of CD8+CD103+ DCs in the spleen. Furthermore, Smad7-deficient DCs expressed higher levels of indoleamine 2,3-dioxygenase (IDO), an enzyme associated with tolerance induction. Mice devoid of Smad7 specifically in DCs are resistant to the development of experimental autoimmune encephalomyelitis (EAE) as a result of an increase of protective regulatory T cells (Tregs) and reduction of encephalitogenic effector T cells in the central nervous system. In agreement, inhibition of IDO activity or depletion of Tregs restored disease susceptibility. Intriguingly, when Smad7-deficient DCs also lacked the IFN-γ receptor, the mice regained susceptibility to EAE, demonstrating that IFN-γ signaling in DCs mediates their tolerogenic function. Our data indicate that Smad7 expression governs splenic DC subset differentiation and is critical for the promotion of their efficient function in immunity.
Collapse
|
27
|
Marin‐Bañasco C, Benabdellah K, Melero‐Jerez C, Oliver B, Pinto‐Medel MJ, Hurtado‐Guerrero I, de Castro F, Clemente D, Fernández O, Martin F, Leyva L, Suardíaz M. Gene therapy with mesenchymal stem cells expressing IFN-ß ameliorates neuroinflammation in experimental models of multiple sclerosis. Br J Pharmacol 2017; 174:238-253. [PMID: 27882538 PMCID: PMC5241389 DOI: 10.1111/bph.13674] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Recombinant IFN-ß is one of the first-line treatments in multiple sclerosis (MS), despite its lack of efficacy in some patients. In this context, mesenchymal stem cells (MSCs) represent a promising therapeutic alternative due to their immunomodulatory properties and multipotency. Moreover, by taking advantage of their pathotropism, these cells can be genetically modified to be used as carriers for delivering or secreting therapeutic drugs into injured tissues. Here, we report the therapeutic effect of systemic delivery of adipose-derived MSCs (AdMSCs), transduced with the IFN-β gene, into mice with experimental autoimmune encephalomyelitis (EAE). EXPERIMENTAL APPROACH Relapsing-remitting and chronic progressive EAE were induced in mice. Cells were injected i.v. Disease severity, inflammation and tissue damage were assessed clinically, by flow cytometry of spleens and histopathological evaluation of the CNS respectively. KEY RESULTS Genetic engineering did not modify the biological characteristics of these AdMSCs (morphology, growth rate, immunophenotype and multipotency). Furthermore, the transduction of IFN-ß to AdMSCs maintained and, in some cases, enhanced the functional properties of AdMSCs by ameliorating the symptoms of MS in EAE models and by decreasing indications of peripheral and central neuro-inflammation. CONCLUSION AND IMPLICATIONS Gene therapy was found to be more effective than cell therapy in ameliorating several clinical parameters in both EAE models, presumably due to the continuous expression of IFN-β. Furthermore, it has significant advantages over AdMSC therapy, and also over systemic IFN-ß treatment, by providing long-term expression of the cytokine at therapeutic concentrations and reducing the frequency of injections, while minimizing dose-limiting side effects.
Collapse
MESH Headings
- Adipose Tissue/cytology
- Animals
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Flow Cytometry
- Genetic Therapy/methods
- Interferon-beta/genetics
- Mesenchymal Stem Cell Transplantation/methods
- Mesenchymal Stem Cells/cytology
- Mice
- Mice, Inbred C57BL
- Multiple Sclerosis, Chronic Progressive/genetics
- Multiple Sclerosis, Chronic Progressive/physiopathology
- Multiple Sclerosis, Chronic Progressive/therapy
- Multiple Sclerosis, Relapsing-Remitting/genetics
- Multiple Sclerosis, Relapsing-Remitting/physiopathology
- Multiple Sclerosis, Relapsing-Remitting/therapy
- Severity of Illness Index
Collapse
Affiliation(s)
- C Marin‐Bañasco
- Unidad de Gestión Clínica Inter‐centros de Neurociencias, Laboratorio de Investigación y Servicio de Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaUniversidad de MálagaSpain
| | - K Benabdellah
- Cell and Gene Therapy Group, Genomic Medicine DepartmentGENYO, Centre for Genomics and Oncological Research, Pfizer‐University of Granada‐Andalusian Regional GovernmentGranadaSpain
| | - C Melero‐Jerez
- Grupo de Neurobiología del Desarrollo‐GNDeInstituto Cajal‐CSIC28002MadridSpain
- Grupo de Neuroinmuno‐ReparaciónHospital Nacional de Parapléjicos45071ToledoSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| | - B Oliver
- Unidad de Gestión Clínica Inter‐centros de Neurociencias, Laboratorio de Investigación y Servicio de Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaUniversidad de MálagaSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| | - M J Pinto‐Medel
- Unidad de Gestión Clínica Inter‐centros de Neurociencias, Laboratorio de Investigación y Servicio de Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaUniversidad de MálagaSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| | - I Hurtado‐Guerrero
- Unidad de Gestión Clínica Inter‐centros de Neurociencias, Laboratorio de Investigación y Servicio de Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaUniversidad de MálagaSpain
| | - F de Castro
- Grupo de Neurobiología del Desarrollo‐GNDeInstituto Cajal‐CSIC28002MadridSpain
- Grupo de Neurobiología del Desarrollo‐GNDeHospital Nacional de Parapléjicos45071ToledoSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| | - D Clemente
- Grupo de Neuroinmuno‐ReparaciónHospital Nacional de Parapléjicos45071ToledoSpain
- Grupo de Neurobiología del Desarrollo‐GNDeHospital Nacional de Parapléjicos45071ToledoSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| | - O Fernández
- Unidad de Gestión Clínica Inter‐centros de Neurociencias, Laboratorio de Investigación y Servicio de Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaUniversidad de MálagaSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| | - F Martin
- Cell and Gene Therapy Group, Genomic Medicine DepartmentGENYO, Centre for Genomics and Oncological Research, Pfizer‐University of Granada‐Andalusian Regional GovernmentGranadaSpain
| | - L Leyva
- Unidad de Gestión Clínica Inter‐centros de Neurociencias, Laboratorio de Investigación y Servicio de Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaUniversidad de MálagaSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| | - M Suardíaz
- Unidad de Gestión Clínica Inter‐centros de Neurociencias, Laboratorio de Investigación y Servicio de Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de MálagaUniversidad de MálagaSpain
- Red Española de Esclerosis Múltiple (REEM)BarcelonaSpain
| |
Collapse
|
28
|
McGray AJR, Bramson J. Adaptive Resistance to Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:213-227. [PMID: 29275474 DOI: 10.1007/978-3-319-67577-0_14] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Immunosuppressive mechanisms within the tumor microenvironment have emerged as a major impediment to cancer immunotherapy. While a broad range of secreted factors, receptors/ligands, and cell populations have been described that contribute to the immunosuppression, the involvement of these processes in immune evasion by tumors is typically considered to be an intrinsic property of the tumor. Evidence is now emerging that the processes underlying immune suppression within the tumor are, in fact, triggered by immune attack and reflect a dynamic interplay between the tumor and the host's immune system. The term adaptive resistance has been coined to describe the induction of immune suppressive pathways in the tumor following active attack on the tumor. Adaptive resistance is a scalable process where the magnitude of immune suppression matches the magnitude of the immune attack; the net balance between suppression and attack determines the durability of the anti-tumor response and tumor outcome. In this chapter, we will examine the data supporting adaptive resistance and the opposing roles of T cells in simultaneously promoting both anti-tumor immunity and immune suppression within the tumor microenvironment. The clinical implications of adaptive resistance in the design and application of immunotherapeutic strategies is also discussed.
Collapse
Affiliation(s)
- A J Robert McGray
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Jonathan Bramson
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
29
|
Recombinant soluble IFN receptor (sIFNAR2) exhibits intrinsic therapeutic efficacy in a murine model of Multiple Sclerosis. Neuropharmacology 2016; 110:480-492. [DOI: 10.1016/j.neuropharm.2016.07.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 07/19/2016] [Accepted: 07/20/2016] [Indexed: 11/23/2022]
|
30
|
Renner K, Hellerbrand S, Hermann F, Riedhammer C, Talke Y, Schiechl G, Rodriguez Gomez M, Kutzi S, Halbritter D, Goebel N, Brühl H, Weissert R, Mack M. IL-3 promotes the development of experimental autoimmune encephalitis. JCI Insight 2016; 1:e87157. [PMID: 27734026 DOI: 10.1172/jci.insight.87157] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Little is known about the role of IL-3 in multiple sclerosis (MS) in humans and in experimental autoimmune encephalomyelitis (EAE). Using myelin oligodendrocyte glycoprotein (MOG) peptide-induced EAE, we show that CD4+ T cells are the main source of IL-3 and that cerebral IL-3 expression correlates with the influx of T cells into the brain. Blockade of IL-3 with monoclonal antibodies, analysis of IL-3 deficient mice, and adoptive transfer of leukocytes demonstrate that IL-3 plays an important role for development of clinical symptoms of EAE, for migration of leukocytes into the brain, and for cerebral expression of adhesion molecules and chemokines. In contrast, injection of recombinant IL-3 exacerbates EAE symptoms and cerebral inflammation. In patients with relapsing-remitting MS (RRMS), IL-3 expression by T cells is markedly upregulated during episodes of relapse. Our data indicate that IL-3 plays an important role in EAE and may represent a new target for treatment of MS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hilke Brühl
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Robert Weissert
- Department of Neurology.,Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Matthias Mack
- Department of Internal Medicine II - Nephrology.,Regensburg Center for Interventional Immunology, Regensburg, Germany
| |
Collapse
|
31
|
Baxter VK, Griffin DE. Interferon gamma modulation of disease manifestation and the local antibody response to alphavirus encephalomyelitis. J Gen Virol 2016; 97:2908-2925. [PMID: 27667782 DOI: 10.1099/jgv.0.000613] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Infection of mice with Sindbis virus (SINV) produces encephalomyelitis and provides a model for examination of the central nervous system (CNS) immune response to alphavirus infection. Clearance of infectious virus is accomplished through a cooperative effort between SINV-specific antibody and IFN-γ, but the regulatory interactions are poorly understood. To determine the effects of IFN-γ on clinical disease and the antiviral immune response, C57BL/6 mice lacking IFN-γ (Ifng-/-) or IFN-γ receptor (Ifngr1-/-) were studied in comparison to WT mice. Maximum production of Ifng mRNA and IFN-γ protein in the CNS of WT and Ifngr1-/- mice occurred 5-7 days after infection, with higher levels of IFN-γ in Ifngr1-/- mice. Onset of clinical disease was earlier in mice with impaired IFN-γ signalling, although Ifngr1-/- mice recovered more rapidly. Ifng-/- and Ifngr1-/- mice maintained body weight better than WT mice, associated with better food intake and lower brain levels of inflammatory cytokines. Clearance of infectious virus from the spinal cords was slower, and CNS, but not serum, levels of SINV-specific IgM, IgG2a and IgG2b were lower in Ifngr1-/- and Ifng-/- mice compared to WT mice. Decreased CNS antiviral antibody was associated with lower expression of mRNAs for B-cell attracting chemokines CXCL9, CXCL10 and CXCL13 and fewer B cells in the CNS. Therefore, IFN-γ signalling increases levels of CNS pro-inflammatory cytokines, leading to clinical disease, but synergistically clears virus with SINV-specific antibody at least in part by increasing chemokine production important for infiltration of antibody-secreting B cells into the CNS.
Collapse
Affiliation(s)
- Victoria K Baxter
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
32
|
High-fat diet-induced brain region-specific phenotypic spectrum of CNS resident microglia. Acta Neuropathol 2016; 132:361-75. [PMID: 27393312 PMCID: PMC4992033 DOI: 10.1007/s00401-016-1595-4] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/17/2022]
Abstract
Diets high in fat (HFD) are known to cause an immune response in the periphery as well as the central nervous system. In peripheral adipose tissue, this immune response is primarily mediated by macrophages that are recruited to the tissue. Similarly, reactivity of microglia, the innate immune cells of the brain, has been shown to occur in the hypothalamus of mice fed a high-fat diet. To characterize the nature of the microglial response to diets high in fat in a temporal fashion, we studied the phenotypic spectrum of hypothalamic microglia of mice fed high-fat diet for 3 days and 8 weeks by assessing their tissue reaction and inflammatory signature. While we observed a significant increase in Iba1+ myeloid cells and a reaction of GFAP+ astrocytes in the hypothalamus after 8 weeks of HFD feeding, we found the hypothalamic myeloid cell reaction to be limited to endogenous microglia and not mediated by infiltrating myeloid cells. Moreover, obese humans were found to present with signs of hypothalamic gliosis and exacerbated microglia dystrophy, suggesting a targeted microglia response to diet in humans as well. Notably, the glial reaction occurring in the mouse hypothalamus was not accompanied by an increase in pro-inflammatory cytokines, but rather by an anti-inflammatory reaction. Gene expression analyses of isolated microglia not only confirmed this observation, but also revealed a downregulation of microglia genes important for sensing signals in the microenvironment. Finally, we demonstrate that long-term exposure of microglia to HFD in vivo does not impair the cell’s ability to respond to additional stimuli, like lipopolysaccharide. Taken together, our findings support the notion that microglia react to diets high in fat in a region-specific manner in rodents as well as in humans; however, this response changes over time as it is not exclusively pro-inflammatory nor does exposure to HFD prime microglia in the hypothalamus.
Collapse
|
33
|
Korniotis S, Gras C, Letscher H, Montandon R, Mégret J, Siegert S, Ezine S, Fallon PG, Luther SA, Fillatreau S, Zavala F. Treatment of ongoing autoimmune encephalomyelitis with activated B-cell progenitors maturing into regulatory B cells. Nat Commun 2016; 7:12134. [PMID: 27396388 PMCID: PMC4942579 DOI: 10.1038/ncomms12134] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 06/02/2016] [Indexed: 12/24/2022] Open
Abstract
The influence of signals perceived by immature B cells during their development in bone marrow on their subsequent functions as mature cells are poorly defined. Here, we show that bone marrow cells transiently stimulated in vivo or in vitro through the Toll-like receptor 9 generate proB cells (CpG-proBs) that interrupt experimental autoimmune encephalomyelitis (EAE) when transferred at the onset of clinical symptoms. Protection requires differentiation of CpG-proBs into mature B cells that home to reactive lymph nodes, where they trap T cells by releasing the CCR7 ligand, CCL19, and to inflamed central nervous system, where they locally limit immunopathogenesis through interleukin-10 production, thereby cooperatively inhibiting ongoing EAE. These data demonstrate that a transient inflammation at the environment, where proB cells develop, is sufficient to confer regulatory functions onto their mature B-cell progeny. In addition, these properties of CpG-proBs open interesting perspectives for cell therapy of autoimmune diseases. Evidence of how functional Bregs develop in vivo has been lacking. Here the authors show that proB cells exposed in vivo to CpG differentiate into distinct Breg subsets that inhibit autoimmunity by arresting T cells in the lymph nodes via CCL19 and by producing IL-10 at the site of immunopathology.
Collapse
Affiliation(s)
- Sarantis Korniotis
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Christophe Gras
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Hélène Letscher
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Ruddy Montandon
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Jérôme Mégret
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Structure Fédérative de Recherche Necker, INSERM US 24, CNRS UMS 3633, Paris 75014, France
| | - Stefanie Siegert
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Sophie Ezine
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| | - Padraic G Fallon
- Department of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Simon Fillatreau
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Assistance Publique-Hopitaux de Paris (AP-HP), Hopital Necker Enfants Malades, Paris 75015, France.,Deutsches Rheuma-Forschungszentrum, a Leibniz Institute, Chariteplatz 1, Berlin 10117, Germany
| | - Flora Zavala
- Institut Necker Enfants Malades, Immunology, Infectiology and Haematology Department, Inserm U1151, CNRS UMR 8253, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Site Necker, 14 rue Maria Helena Vieira da Silva, CS 61431, Paris 75014, France
| |
Collapse
|
34
|
A(H1N1) vaccination recruits T lymphocytes to the choroid plexus for the promotion of hippocampal neurogenesis and working memory in pregnant mice. Brain Behav Immun 2016; 53:72-83. [PMID: 26576725 DOI: 10.1016/j.bbi.2015.11.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 12/20/2022] Open
Abstract
We previously demonstrated that A(H1N1) influenza vaccine (AIV) promoted hippocampal neurogenesis and working memory in pregnant mice. However, the underlying mechanism of flu vaccination in neurogenesis and memory has remained unclear. In this study, we found that T lymphocytes were recruited from the periphery to the choroid plexus (CP) of the lateral and third (3rd) ventricles in pregnant mice vaccinated with AIV (Pre+AIV). Intracerebroventricular delivery of anti-TCR antibodies markedly decreased neurogenesis and the working memory of the Pre+AIV mice. Similarly, intravenous delivery of anti-CD4 antibodies to the periphery also down-regulated neurogenesis. Furthermore, AIV vaccination caused microglia to skew toward an M2-like phenotype (increased Arginase-1 and Ym1 mRNA levels), and elevated levels of brain-derived growth factor (BDNF) and insulin-like growth factor-1 (IGF-1) were found in the hippocampus, whereas these effects were offset by anti-TCR antibody treatment. Additionally, in the CP, the expression level of adhesion molecules and chemokines, which assist leukocytes in permeating into the brain, were also elevated after AIV vaccination of pregnant mice. Collectively, the results suggested that the infiltrative T lymphocytes in the CP contribute to the increase in hippocampal neurogenesis and working memory caused by flu vaccination, involving activation of the brain's CP, M2 microglial polarization and neurotrophic factor expression.
Collapse
|
35
|
Sato F, Omura S, Jaffe S, Tsunoda I. Role of CD4+ T Cells in the Pathophysiology of Multiple Sclerosis. MULTIPLE SCLEROSIS 2016. [PMCID: PMC7150304 DOI: 10.1016/b978-0-12-800763-1.00004-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Although the precise etiology of MS remains unclear, CD4+ T cells have been proposed to play not only effector but also regulatory roles in MS. CD4+ T cells can be divided into four subsets: pro-inflammatory helper T (Th) 1 and Th17 cells, anti-inflammatory Th2 cells and regulatory T cells (Tregs). The roles of CD4+ T cells in MS have been clarified by either “loss-of-function” or “gain-of-function” methods, which have been carried out mainly in autoimmune and viral models of MS: experimental autoimmune encephalomyelitis and Theiler's murine encephalomyelitis virus infection, respectively. Observations in MS patients were consistent with the mechanisms found in the MS models, that is, increased pro-inflammatory Th1 and Th17 activity is associated with disease exacerbation, while anti-inflammatory Th2 cells and Tregs appear to play a protective role.
Collapse
|
36
|
Abstract
Neurotropic strains of the mouse hepatitis virus (MHV) cause a range of diseases in infected mice ranging from mild encephalitis with clearance of the virus followed by demyelination to rapidly fatal encephalitis. This chapter discusses the structure, life cycle, transmission, and pathology of neurotropic coronaviruses, as well as the immune response to coronavirus infection. Mice infected with neurotropic strains of MHV have provided useful systems in which to study processes of virus- and immune-mediated demyelination and virus clearance and/or persistence in the CNS, and the mechanisms of virus evasion of the immune system.
Collapse
|
37
|
Xie C, Ciric B, Yu S, Zhang GX, Rostami A. IL-12Rβ2 has a protective role in relapsing-remitting experimental autoimmune encephalomyelitis. J Neuroimmunol 2015; 291:59-69. [PMID: 26857496 DOI: 10.1016/j.jneuroim.2015.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 12/10/2015] [Accepted: 12/17/2015] [Indexed: 12/11/2022]
Abstract
IL-12Rβ2 is a common receptor subunit of heterodimeric receptors for IL-12 and IL-35, two cytokines that are implicated in immunopathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. We evaluated the role of IL-12Rβ2 in relapsing-remitting EAE (RR-EAE). IL-12Rβ2-deficient SJL/J mice developed markedly more severe clinical EAE, and had greater mortality and more severe relapses compared with wild-type controls. IL-12Rβ2-deficient EAE mice also had more infiltrating mononuclear cells in the CNS, as well as higher T cell proliferative capacity and decreased IFN-γ production at the periphery. These findings demonstrate a protective role of IL-12Rβ2 in RR-EAE.
Collapse
Affiliation(s)
- Chong Xie
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shuo Yu
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
38
|
Lin H, Song P, Zhao Y, Xue LJ, Liu Y, Chu CQ. Targeting Th17 Cells with Small Molecules and Small Interference RNA. Mediators Inflamm 2015; 2015:290657. [PMID: 26792955 PMCID: PMC4697089 DOI: 10.1155/2015/290657] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/30/2015] [Indexed: 02/05/2023] Open
Abstract
T helper 17 (Th17) cells play a central role in inflammatory and autoimmune diseases via the production of proinflammatory cytokines interleukin- (IL-) 17, IL-17F, and IL-22. Anti-IL-17 monoclonal antibodies show potent efficacy in psoriasis but poor effect in rheumatoid arthritis (RA) and Crohn's disease. Alternative agents targeting Th17 cells may be a better way to inhibit the development and function of Th17 cells than antibodies of blocking a single effector cytokine. Retinoic acid-related orphan receptor gamma t (RORγt) which acts as the master transcription factor of Th17 differentiation has been an attractive pharmacologic target for the treatment of Th17-mediated autoimmune disease. Recent progress in technology of chemical screen and engineering nucleic acid enable two new classes of therapeutics targeting RORγt. Chemical screen technology identified several small molecule specific inhibitors of RORγt from a small molecule library. Systematic evolution of ligands by exponential enrichment (SELEX) technology enabled target specific aptamers to be isolated from a random sequence oligonucleotide library. In this review, we highlight the development and therapeutic potential of small molecules inhibiting Th17 cells by targeting RORγt and aptamer mediated CD4(+) T cell specific delivery of small interference RNA against RORγt gene expression to inhibit pathogenic effector functions of Th17 lineage.
Collapse
Affiliation(s)
- Hui Lin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Pingfang Song
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University and VA Portland Health Care System, Portland, OR 97239, USA
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li-Jia Xue
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University and VA Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
39
|
Arellano G, Ottum PA, Reyes LI, Burgos PI, Naves R. Stage-Specific Role of Interferon-Gamma in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Front Immunol 2015; 6:492. [PMID: 26483787 PMCID: PMC4586507 DOI: 10.3389/fimmu.2015.00492] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022] Open
Abstract
The role of interferon (IFN)-γ in multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), has remained as an enigmatic paradox for more than 30 years. Several studies attribute this cytokine a prominent proinflammatory and pathogenic function in these pathologies. However, accumulating evidence shows that IFN-γ also plays a protective role inducing regulatory cell activity and modulating the effector T cell response. Several innate and adaptive immune cells also develop opposite functions strongly associated with the production of IFN-γ in EAE. Even the suppressive activity of different types of regulatory cells is dependent on IFN-γ. Interestingly, recent data supports a stage-specific participation of IFN-γ in EAE providing a plausible explanation for previous conflicting results. In this review, we will summarize and discuss such literature, emphasizing the protective role of IFN-γ on immune cells. These findings are fundamental to understand the complex role of IFN-γ in the pathogenesis of these diseases and can provide basis for potential stage-specific therapy for MS targeting IFN-γ-signaling or IFN-γ-producing immune cells.
Collapse
Affiliation(s)
- Gabriel Arellano
- Immunology Program, Biomedical Sciences Institute, School of Medicine, Universidad de Chile , Santiago , Chile
| | - Payton A Ottum
- Immunology Program, Biomedical Sciences Institute, School of Medicine, Universidad de Chile , Santiago , Chile
| | - Lilian I Reyes
- Faculty of Science, Universidad San Sebastián , Santiago , Chile
| | - Paula I Burgos
- Department of Clinical Immunology and Rheumatology, School of Medicine, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Rodrigo Naves
- Immunology Program, Biomedical Sciences Institute, School of Medicine, Universidad de Chile , Santiago , Chile
| |
Collapse
|
40
|
Miller NM, Wang J, Tan Y, Dittel BN. Anti-inflammatory mechanisms of IFN-γ studied in experimental autoimmune encephalomyelitis reveal neutrophils as a potential target in multiple sclerosis. Front Neurosci 2015; 9:287. [PMID: 26347600 PMCID: PMC4539553 DOI: 10.3389/fnins.2015.00287] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 07/30/2015] [Indexed: 01/01/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) mediated by T helper (h)1 and/or Th17 CD4 T cells that drive inflammatory lesion development along with demyelination and neuronal damage. Defects in immune regulatory mechanisms are thought to play a role in the pathogenesis of MS. While an early clinical trial indicated that IFN-γ administration was detrimental to MS, studies in the mouse model of MS, experimental autoimmune encephalomyelitis (EAE), indicated that IFN-γ exhibits a number of anti-inflammatory properties within the CNS. These mechanisms include inhibition of IL-17 production, induction of regulatory T cells, T cell apoptosis and regulation of chemokine production. Mice deficient in IFN-γ or its receptor were instrumental in deciphering the anti-inflammatory properties of IFN-γ in the CNS. In particular, they revealed that IFN-γ is a major regulator of neutrophil recruitment into the CNS, which by a variety of mechanisms including disruption of the blood-brain-barrier (BBB) and production of reactive oxygen species are thought to contribute to the onset and progression of EAE. Neutrophils were also shown to be instrumental in EAE relapses. To date neutrophils have not been appreciated as a driver of MS, but more recently based largely on strong EAE data this view is being reevaluated by some investigators in the field.
Collapse
Affiliation(s)
- Nichole M Miller
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| | - Jun Wang
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| | - Yanping Tan
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| | - Bonnie N Dittel
- BloodCenter of Wisconsin, Blood Research Institute Milwaukee, WI, USA
| |
Collapse
|
41
|
Lee YY, Yang YP, Huang PI, Li WC, Huang MC, Kao CL, Chen YJ, Chen MT. Exercise suppresses COX-2 pro-inflammatory pathway in vestibular migraine. Brain Res Bull 2015; 116:98-105. [PMID: 26151770 DOI: 10.1016/j.brainresbull.2015.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/19/2015] [Accepted: 06/26/2015] [Indexed: 01/18/2023]
Abstract
Migraine and dizziness are relatively common disorders. Patients with dizziness have a higher incidence of migraines than the general population. The discomfort experienced by these patients is often poorly controlled by medication. However, the pathophysiology of vestibular migraine (VM) remains unclear. We hypothesized that patients with VM would experience remission from symptoms after exercise training and that this effect may be mediated through the suppression of cyclooxygenase-2 (COX-2)-mediated inflammation. Thus, the aim of the present study was to investigate the efficacy and possible anti-inflammatory benefits of exercise in patients with VM. We assessed the level of soluble inflammatory mediators in plasma from VM patients and control subjects. Our analysis of cytokine expression in the patients with VM undergoing exercise treatment revealed a significant reduction in pro-inflammatory cytokines and/or cytotoxic factors, such as tumor necrosis factor-α, interleukins, nitric oxide (NO), inducible NO synthase, and reactive oxygen species. In contrast, we found an increase in the level of anti-inflammatory cytokines after exercise. Moreover, the group undergoing exercise training showed significant symptomatic improvement and demonstrated suppressed antioxidant enzyme activity. To summarize, our data suggest that exercise significantly inhibits COX-2 activity, leading to the suppression of pro-inflammatory cytokines and changes in redox status. These results suggest that there is a molecular link between the central nervous system and the immune system. Furthermore, elucidation of the neurobiological mechanisms underlying VM could potentially lead to the development of novel therapeutic interventions for these patients.
Collapse
Affiliation(s)
- Yi-Yen Lee
- Institute of Clinical Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan; Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan; Faculty of Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan
| | - Yi-Ping Yang
- Institute of Clinical Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan; Faculty of Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan
| | - Pin-I Huang
- Faculty of Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan; Cancer Center, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan
| | - Wen-Cheng Li
- Department of Occupation Medicine, Chang-Gung Memorial Hospital, Taipei, 199, Tung Hwa North Road, Taipei 105, Taiwan
| | - Ming-Chao Huang
- Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan; Faculty of Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan
| | - Chung-Lan Kao
- Faculty of Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan; Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan
| | - Yann-Jang Chen
- Institute of Clinical Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan; Department of Pediatrics, Renai Branch, Taipei City Hospital, No. 10, Sec. 4, Ren'ai Rd., Da'an Dist., Taipei City 106, Taiwan; Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, No.155, Sec.2, Linong Street, Taipei 11221, Taiwan
| | - Ming-Teh Chen
- Faculty of Medicine, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 11221, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No.201, Sec.2, Shih-Pai Road, Taipei 11217, Taiwan.
| |
Collapse
|
42
|
Millward JM, Guo J, Berndt D, Braun J, Sack I, Infante-Duarte C. Tissue structure and inflammatory processes shape viscoelastic properties of the mouse brain. NMR IN BIOMEDICINE 2015; 28:831-839. [PMID: 25963743 DOI: 10.1002/nbm.3319] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 04/02/2015] [Accepted: 04/07/2015] [Indexed: 06/04/2023]
Abstract
Magnetic resonance elastography (MRE) is an imaging method that reveals the mechanical properties of tissue, modelled as a combination of " viscosity" and " elasticity" . We recently showed reduced brain viscoelasticity in multiple sclerosis (MS) patients compared with healthy controls, and in the relapsing-remitting disease model experimental autoimmune encephalomyelitis (EAE). However, the mechanisms by which these intrinsic tissue properties become altered remain unclear. This study investigates whether distinct regions in the mouse brain differ in their native viscoelastic properties, and how these properties are affected during chronic EAE in C57Bl/6 mice and in mice lacking the cytokine interferon-gamma. IFN-γ(-/-) mice exhibit a more severe EAE phenotype, with amplified inflammation in the cerebellum and brain stem. Brain scans were performed in the sagittal plane using a 7 T animal MRI scanner, and the anterior (cerebral) and posterior (cerebellar) regions analyzed separately. MRE investigations were accompanied by contrast-enhanced MRI scans, and by histopathology and gene expression analysis ex vivo. Compared with the cerebrum, the cerebellum in healthy mice has a lower viscoelasticity, i.e. it is intrinsically " softer" . This was seen both in the wild-type mice and the IFNγ(-/-) mice. During chronic EAE, C57Bl/6 mice did not show altered brain viscoelasticity. However, as expected, the IFNγ(-/-) mice showed a more severe EAE phenotype, and these mice did show altered brain elasticity during the course of disease. The magnitude of the elasticity reduction correlated with F4/80 gene expression, a marker for macrophages/microglia in inflamed central nervous system tissue. Together these results demonstrate that MRE is sensitive enough to discriminate between viscoelastic properties in distinct anatomical structures in the mouse brain, and to confirm a further relationship between cellular inflammation and mechanical alterations of the brain. This study underscores the utility of MRE to monitor pathological tissue alterations in vivo.
Collapse
Affiliation(s)
- Jason M Millward
- Institute for Medical Immunology, Charité - Universitätmedizin Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Jing Guo
- Department of Radiology, Charité - Universitätsmedizin Berlin, Germany
| | - Dominique Berndt
- Institute for Medical Immunology, Charité - Universitätmedizin Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Jürgen Braun
- Department of Radiology, Charité - Universitätsmedizin Berlin, Germany
| | - Ingolf Sack
- Department of Radiology, Charité - Universitätsmedizin Berlin, Germany
| | - Carmen Infante-Duarte
- Institute for Medical Immunology, Charité - Universitätmedizin Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
43
|
Kostic M, Stojanovic I, Marjanovic G, Zivkovic N, Cvetanovic A. Deleterious versus protective autoimmunity in multiple sclerosis. Cell Immunol 2015; 296:122-32. [PMID: 25944389 DOI: 10.1016/j.cellimm.2015.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/18/2015] [Accepted: 04/22/2015] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disorder of central nervous system, in which myelin specific CD4(+) T cells have a central role in orchestrating pathological events involved in disease pathogenesis. There is compelling evidence that Th1, Th9 and Th17 cells, separately or in cooperation, could mediate deleterious autoimmune response in MS. However, the phenotype differences between Th cell subpopulations initially employed in MS pathogenesis are mainly reflected in the different patterns of inflammation introduction, which results in the development of characteristic pathological features (blood-brain barrier disruption, demyelination and neurodegeneration), clinically presented with MS symptoms. Although, autoimmunity was traditionally seen as deleterious, some studies indicated that autoimmunity mediated by Th2 cells and T regulatory cells could be protective by nature. The concept of protective autoimmunity in MS pathogenesis is still poorly understood, but could be of great importance in better understanding of MS immunology and therefore, creating better therapeutic strategies.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia.
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Goran Marjanovic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty, University of Nis, Blvd. Dr. Zorana Djindjica 81, 18000 Nis, Serbia
| | - Ana Cvetanovic
- Clinic of Oncology, Clinical Centre, Blvd. Dr. Zorana Djindjica 48, 18000 Nis, Serbia
| |
Collapse
|
44
|
Nallar SC, Kalvakolanu DV. Interferons, signal transduction pathways, and the central nervous system. J Interferon Cytokine Res 2015; 34:559-76. [PMID: 25084173 DOI: 10.1089/jir.2014.0021] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The interferon (IFN) family of cytokines participates in the development of innate and acquired immune defenses against various pathogens and pathogenic stimuli. Discovered originally as a proteinaceous substance secreted from virus-infected cells that afforded immunity to neighboring cells from virus infection, these cytokines are now implicated in various human pathologies, including control of tumor development, cell differentiation, and autoimmunity. It is now believed that the IFN system (IFN genes and the genes induced by them, and the factors that regulate these processes) is a generalized alarm of cellular stress, including DNA damage. IFNs exert both beneficial and deleterious effects on the central nervous system (CNS). Our knowledge of the IFN-regulated processes in the CNS is far from being clear. In this article, we reviewed the current understanding of IFN signal transduction pathways and gene products that might have potential relevance to diseases of the CNS.
Collapse
Affiliation(s)
- Shreeram C Nallar
- Department of Microbiology & Immunology, Program in Oncology, Greenebaum Cancer Center, University of Maryland School of Medicine , Baltimore, Maryland
| | | |
Collapse
|
45
|
Song J, Wu C, Korpos E, Zhang X, Agrawal SM, Wang Y, Faber C, Schäfers M, Körner H, Opdenakker G, Hallmann R, Sorokin L. Focal MMP-2 and MMP-9 activity at the blood-brain barrier promotes chemokine-induced leukocyte migration. Cell Rep 2015; 10:1040-54. [PMID: 25704809 DOI: 10.1016/j.celrep.2015.01.037] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 12/22/2014] [Accepted: 01/14/2015] [Indexed: 01/25/2023] Open
Abstract
Although chemokines are sufficient for chemotaxis of various cells, increasing evidence exists for their fine-tuning by selective proteolytic processing. Using a model of immune cell chemotaxis into the CNS (experimental autoimmune encephalomyelitis [EAE]) that permits precise localization of immigrating leukocytes at the blood-brain barrier, we show that, whereas chemokines are required for leukocyte migration into the CNS, additional MMP-2/9 activities specifically at the border of the CNS parenchyma strongly enhance this transmigration process. Cytokines derived from infiltrating leukocytes regulate MMP-2/9 activity at the parenchymal border, which in turn promotes astrocyte secretion of chemokines and differentially modulates the activity of different chemokines at the CNS border, thereby promoting leukocyte migration out of the cuff. Hence, cytokines, chemokines, and cytokine-induced MMP-2/9 activity specifically at the inflammatory border collectively act to accelerate leukocyte chemotaxis across the parenchymal border.
Collapse
Affiliation(s)
- Jian Song
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Chuan Wu
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany
| | - Eva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Xueli Zhang
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Smriti M Agrawal
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany
| | - Ying Wang
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Heinrich Körner
- Menzies Institute for Medical Research, Hobart, TAS 7000, Australia
| | - Ghislain Opdenakker
- Department of Microbiology and Immunobiology, Rega Institute for Medical Research, University of Leuven, 3000 Leuven, Belgium
| | - Rupert Hallmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149 Muenster, Germany; Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany.
| |
Collapse
|
46
|
Th17 differentiation and their pro-inflammation function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 841:99-151. [PMID: 25261206 DOI: 10.1007/978-94-017-9487-9_5] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CD4(+) T helper cells are classical but constantly reinterpreted T-cell subset, playing critical roles in a diverse range of inflammatory responses or diseases. Depending on the cytokines they release and the immune responses they mediate, CD4(+) T cells are classically divided into two major cell populations: Th1 and Th2 cells. However, recent studies challenged this Th1/Th2 paradigm by discovering several T-helper cell subsets with specific differentiation program and functions, including Th17 cells, Treg cells, and Tfh cells. In this chapter, we summarize the current understanding and recent progresses on the Th17 lineage differentiation and its effector impacts on variety of inflammatory responses or disease pathogenesis.
Collapse
|
47
|
Sex-Based Differences in Multiple Sclerosis (Part I): Biology of Disease Incidence. Curr Top Behav Neurosci 2015; 26:29-56. [PMID: 25690593 DOI: 10.1007/7854_2015_371] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease that leads to neuron damage and progressive disability. One major feature of multiple sclerosis (MS) is that it affects women three times more often than men. In this chapter, we overview the evidence that the autoimmune component of MS, which predominates in the early stages of this disease, is more robust in women than in men and undergoes a sharp increase with the onset of puberty. In addition, we discuss the common rodent models of MS that have been used to study the sex-based differences in the development of central nervous system (CNS) autoimmunity. We then address the biological underpinnings of this enhanced MS risk in women by first reviewing the autoimmune mechanisms that are thought to lead to the initiation of this disease and then honing in on how these mechanisms differ between the sexes. Finally, we review what is known about the hormonal and genetic basis of these sex differences in CNS autoimmunity.
Collapse
|
48
|
Litteljohn D, Nelson E, Hayley S. IFN-γ differentially modulates memory-related processes under basal and chronic stressor conditions. Front Cell Neurosci 2014; 8:391. [PMID: 25477784 PMCID: PMC4238410 DOI: 10.3389/fncel.2014.00391] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/03/2014] [Indexed: 02/06/2023] Open
Abstract
Cytokines are inflammatory messengers that orchestrate the brain’s response to immunological challenges, as well as possibly even toxic and psychological insults. We previously reported that genetic ablation of the pro-inflammatory cytokine, interferon-gamma (IFN-γ), attenuated some of the corticosteroid, cytokine, and limbic dopaminergic variations induced by 6 weeks of exposure to an unpredictable psychologically relevant stressor. Presently, we sought to determine whether a lack of IFN-γ would likewise modify the impact of chronic stress on hippocampus-dependent memory function and related neurotransmitter and neurotrophin signaling systems. As predicted, chronic stress impaired spatial recognition memory (Y-maze task) in the wild-type animals. In contrast, though the IFN-γ knockouts (KOs) showed memory disturbances in the basal state, under conditions of chronic stress these mice actually exhibited facilitated memory performance. Paralleling these findings, while overall the KOs displayed altered noradrenergic and/or serotonergic activity in the hippocampus and locus coeruleus, norepinephrine utilization in both of these memory-related brain regions was selectively increased among the chronically stressed KOs. However, contrary to our expectations, neither IFN-γ deletion nor chronic stressor exposure significantly affected nucleus accumbens dopaminergic neurotransmission or hippocampal brain-derived neurotrophic factor protein expression. These findings add to a growing body of evidence implicating cytokines in the often differential regulation of neurobehavioral processes in health and disease. Whereas in the basal state IFN-γ appears to be involved in sustaining memory function and the activity of related brain monoamine systems, in the face of ongoing psychologically relevant stress the cytokine may, in fact, act to restrict potentially adaptive central noradrenergic and spatial memory responses.
Collapse
Affiliation(s)
- Darcy Litteljohn
- Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | - Eric Nelson
- Faculty of Medicine, University of Toronto Toronto, ON, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University Ottawa, ON, Canada
| |
Collapse
|
49
|
Optimal attenuation of experimental autoimmune encephalomyelitis by intravenous immunoglobulin requires an intact interleukin-11 receptor. PLoS One 2014; 9:e101947. [PMID: 25078447 PMCID: PMC4117465 DOI: 10.1371/journal.pone.0101947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 06/12/2014] [Indexed: 01/29/2023] Open
Abstract
Background Intravenous immunoglobulin (IVIg) has been used to treat a variety of autoimmune disorders including multiple sclerosis (MS); however its mechanism of action remains elusive. Recent work has shown that interleukin-11 (IL-11) mRNAs are upregulated by IVIg in MS patient T cells. Both IVIg and IL-11 have been shown to ameliorate experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The objective of this study was to determine whether the protective effects of IVIg in EAE occur through an IL-11 and IL-11 receptor (IL-11R)-dependent mechanism. Methods We measured IL-11 in the circulation of mice and IL-11 mRNA expression in various organs after IVIg treatment. We then followed with EAE studies to test the efficacy of IVIg in wild-type (WT) mice and in mice deficient for the IL-11 receptor (IL-11Rα−/−). Furthermore, we evaluated myelin-specific Th1 and Th17 responses and assessed spinal cord inflammation and demyelination in WT and IL-11Rα−/− mice, with and without IVIg treatment. We also examined the direct effects of mouse recombinant IL-11 on the production of IL-17 by lymph node mononuclear cells. Results IVIg treatment induced a dramatic surge (>1000-fold increase) in the levels of IL-11 in the circulation and a prominent increase of IL-11 mRNA expression in the liver. Furthermore, we found that IL-11Rα−/− mice, unlike WT mice, although initially protected, were resistant to full protection by IVIg during EAE and developed disease with a similar incidence and severity as control-treated IL-11Rα−/− mice, despite initially showing protection. We observed that Th17 cytokine production by myelin-reactive T cells in the draining lymph nodes was unaffected by IVIg in IL-11Rα−/− mice, yet was downregulated in WT mice. Finally, IL-11 was shown to directly inhibit IL-17 production of lymph node cells in culture. Conclusion These results implicate IL-11 as an important immune effector of IVIg in the prevention of Th17-mediated autoimmune inflammation during EAE.
Collapse
|
50
|
Aubé B, Lévesque SA, Paré A, Chamma É, Kébir H, Gorina R, Lécuyer MA, Alvarez JI, De Koninck Y, Engelhardt B, Prat A, Côté D, Lacroix S. Neutrophils mediate blood-spinal cord barrier disruption in demyelinating neuroinflammatory diseases. THE JOURNAL OF IMMUNOLOGY 2014; 193:2438-54. [PMID: 25049355 DOI: 10.4049/jimmunol.1400401] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Disruption of the blood-brain and blood-spinal cord barriers (BBB and BSCB, respectively) and immune cell infiltration are early pathophysiological hallmarks of multiple sclerosis (MS), its animal model experimental autoimmune encephalomyelitis (EAE), and neuromyelitis optica (NMO). However, their contribution to disease initiation and development remains unclear. In this study, we induced EAE in lys-eGFP-ki mice and performed single, nonterminal intravital imaging to investigate BSCB permeability simultaneously with the kinetics of GFP(+) myeloid cell infiltration. We observed a loss in BSCB integrity within a day of disease onset, which paralleled the infiltration of GFP(+) cells into the CNS and lasted for ∼4 d. Neutrophils accounted for a significant proportion of the circulating and CNS-infiltrating myeloid cells during the preclinical phase of EAE, and their depletion delayed the onset and reduced the severity of EAE while maintaining BSCB integrity. We also show that neutrophils collected from the blood or bone marrow of EAE mice transmigrate more efficiently than do neutrophils of naive animals in a BBB cell culture model. Moreover, using intravital videomicroscopy, we demonstrate that the IL-1R type 1 governs the firm adhesion of neutrophils to the inflamed spinal cord vasculature. Finally, immunostaining of postmortem CNS material obtained from an acutely ill multiple sclerosis patient and two neuromyelitis optica patients revealed instances of infiltrated neutrophils associated with regions of BBB or BSCB leakage. Taken together, our data provide evidence that neutrophils are involved in the initial events that take place during EAE and that they are intimately linked with the status of the BBB/BSCB.
Collapse
Affiliation(s)
- Benoit Aubé
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada; Centre d'Optique, Photonique et Laser, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Sébastien A Lévesque
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Alexandre Paré
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Émilie Chamma
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada; Centre d'Optique, Photonique et Laser, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Hania Kébir
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Roser Gorina
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Marc-André Lécuyer
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Jorge I Alvarez
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Yves De Koninck
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Alexandre Prat
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Daniel Côté
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada; Centre d'Optique, Photonique et Laser, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Steve Lacroix
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada;
| |
Collapse
|