51
|
Garabadu D, Singh D. Ocimum basilicum attenuates ethidium bromide-induced cognitive deficits and pre-frontal cortical neuroinflammation, astrogliosis and mitochondrial dysfunction in rats. Metab Brain Dis 2020; 35:483-495. [PMID: 31997265 DOI: 10.1007/s11011-020-00536-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/17/2020] [Indexed: 01/13/2023]
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disorder with clinical symptoms of neuroinflammation and demyelination in the central nervous system. Recently, herbal medicines are clinically effective against MS as the current disease-modifying drugs have limited effectiveness. Hence, the present study evaluated the therapeutic potential of Ocimum basilicum essential oil (OB) in ethidium bromide (EB)-induced cognitive deficits in the male rats. Further, the effect of OB (50, 100 and 200 μL/kg) was evaluated on EB-induced neuroinflammation, astrogliosis and mitochondrial dysfunction in the pre-frontal cortex (PFC) of the animals. The EB was injected through bilateral intracerebroventricular route into hippocampus to induce MS-like manifestations in the rats. OB (100 and 200 μL/kg) and Ursolic acid (UA) significantly reduced the EB-induced cognitive deficits in Morris water maze and Y-maze test paradigms. OB (100 and 200 μL/kg) and UA significantly attenuated the EB-induced neuroinflammation in terms of increase in the levels of pro-inflammatory cytokines (TNF-alpha and IL-6) in the rat PFC. Further, OB (100 and 200 μL/kg) and UA significantly attenuated the EB-induced astrogliosis in terms of increase in the levels of GFAP (Glial fibrillary acidic protein) and Iba-1 (Ionized calcium binding adaptor molecule-1) in the rat PFC. In addition, OB (100 and 200 μL/kg) and UA significantly attenuated the EB-induced decrease in the mitochondrial function, integrity, respiratory control rate and ADP/O in the PFC of the rodents. Moreover, OB (100 and 200 μL/kg) and UA significantly reduced the EB-induced mitochondria-dependent apoptosis in the PFC of the rat. Hence, it can be presumed that OB could be a potential alternative drug candidate in the pharmacotherapy of MS.
Collapse
Affiliation(s)
- Debapriya Garabadu
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India.
| | - Deepanshu Singh
- Division of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| |
Collapse
|
52
|
Xin P, Xu X, Deng C, Liu S, Wang Y, Zhou X, Ma H, Wei D, Sun S. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol 2020; 80:106210. [PMID: 31972425 DOI: 10.1016/j.intimp.2020.106210] [Citation(s) in RCA: 500] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 02/09/2023]
Abstract
The JAK/STAT signaling pathway is an universally expressed intracellular signal transduction pathway and involved in many crucial biological processes, including cell proliferation, differentiation, apoptosis, and immune regulation. It provides a direct mechanism for extracellular factors-regulated gene expression. Current researches on this pathway have been focusing on the inflammatory and neoplastic diseases and related drug. The mechanism of JAK/STAT signaling is relatively simple. However, the biological consequences of the pathway are complicated due to its crosstalk with other signaling pathways. In addition, there is increasing evidence indicates that the persistent activation of JAK/STAT signaling pathway is closely related to many immune and inflammatory diseases, yet the specific mechanism remains unclear. Therefore, it is necessary to study the detailed mechanisms of JAK/STAT signaling in disease formation to provide critical reference for clinical treatments of the diseases. In this review, we focus on the structure of JAKs and STATs, the JAK/STAT signaling pathway and its negative regulators, the associated diseases, and the JAK inhibitors for the clinical therapy.
Collapse
Affiliation(s)
- Ping Xin
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Xiaoyun Xu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Chengjie Deng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shuang Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Youzhi Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuegang Zhou
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Hongxing Ma
- Clinical Laboratory Department, Najing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Najing 211200, China
| | - Donghua Wei
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shiqin Sun
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China.
| |
Collapse
|
53
|
Paudel YN, Angelopoulou E, C BK, Piperi C, Othman I. High mobility group box 1 (HMGB1) protein in Multiple Sclerosis (MS): Mechanisms and therapeutic potential. Life Sci 2019; 238:116924. [DOI: 10.1016/j.lfs.2019.116924] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/27/2019] [Accepted: 09/29/2019] [Indexed: 02/07/2023]
|
54
|
Roads to Formation of Normal Myelin Structure and Pathological Myelin Structure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:257-264. [PMID: 31760649 DOI: 10.1007/978-981-32-9636-7_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Demyelination and axonal damage are responsible for neurological deficits in demyelinating diseases including multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system. However, the pathology of demyelination and axonal damage in MS is not fully understood. While immunologists have accumulated evidence, which is involved in many immunological events in these diseases, neuroscientists and anatomists have also investigated morphological changes of myelin in these diseases. In this chapter, a new concept of demyelination will be described.
Collapse
|
55
|
Increased plasma levels of mitochondrial DNA and pro-inflammatory cytokines in patients with progressive multiple sclerosis. J Neuroimmunol 2019; 338:577107. [PMID: 31726376 DOI: 10.1016/j.jneuroim.2019.577107] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/24/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022]
Abstract
The role of damage-associated molecular patterns in multiple sclerosis (MS) is under investigation. Here, we studied the contribution of circulating high mobility group box protein 1 (HMGB1) and mitochondrial DNA (mtDNA) to neuroinflammation in progressive MS. We measured plasmatic mtDNA, HMGB1 and pro-inflammatory cytokines in 38 secondary progressive (SP) patients, 35 primary progressive (PP) patients and 42 controls. Free mtDNA was higher in SP than PP. Pro-inflammatory cytokines were increased in progressive patients. In PP, tumor necrosis factor-α correlated with MS Severity Score. Thus, in progressive patients, plasmatic mtDNA and pro-inflammatory cytokines likely contribute to the systemic inflammatory status.
Collapse
|
56
|
Tan BL, Norhaizan ME. Effect of High-Fat Diets on Oxidative Stress, Cellular Inflammatory Response and Cognitive Function. Nutrients 2019; 11:nu11112579. [PMID: 31731503 PMCID: PMC6893649 DOI: 10.3390/nu11112579] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 12/20/2022] Open
Abstract
Cognitive dysfunction is linked to chronic low-grade inflammatory stress that contributes to cell-mediated immunity in creating an oxidative environment. Food is a vitally important energy source; it affects brain function and provides direct energy. Several studies have indicated that high-fat consumption causes overproduction of circulating free fatty acids and systemic inflammation. Immune cells, free fatty acids, and circulating cytokines reach the hypothalamus and initiate local inflammation through processes such as microglial proliferation. Therefore, the role of high-fat diet (HFD) in promoting oxidative stress and neurodegeneration is worthy of further discussion. Of particular interest in this article, we highlight the associations and molecular mechanisms of HFD in the modulation of inflammation and cognitive deficits. Taken together, a better understanding of the role of oxidative stress in cognitive impairment following HFD consumption would provide a useful approach for the prevention of cognitive dysfunction.
Collapse
Affiliation(s)
- Bee Ling Tan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
| | - Mohd Esa Norhaizan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia;
- Research Centre of Excellent, Nutrition and Non-Communicable Diseases (NNCD), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
- Correspondence: ; Tel.: +603-8947-2427
| |
Collapse
|
57
|
Rudolph J, Meinke C, Voss M, Guttek K, Kliche S, Reinhold D, Schraven B, Reinhold A. Immune Cell-Type Specific Ablation of Adapter Protein ADAP Differentially Modulates EAE. Front Immunol 2019; 10:2343. [PMID: 31632410 PMCID: PMC6779796 DOI: 10.3389/fimmu.2019.02343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/17/2019] [Indexed: 01/03/2023] Open
Abstract
The cytosolic adhesion and degranulation-promoting adapter protein ADAP is expressed in various hematopoietic cells including T cells, NK cells, myeloid cells, and platelets but absent in mature B cells. The role of ADAP in T cell activation, proliferation and integrin activation is well-accepted. We previously demonstrated that conventional ADAP knockout mice show a significantly attenuated course of experimental autoimmune encephalomyelitis (EAE). To dissect the impact of different ADAP expressing cell populations on the reduced EAE severity, here, we generated lineage-specific conditional knockout mice. ADAP was deleted in T cells, myeloid cells, NK cells and platelets, respectively. Specific loss of ADAP was confirmed on the protein level. Detailed immunophenotyping was performed to assess the consequence of deletion of ADAP with regard to the maturation and distribution of immune cells in primary and secondary lymphoid organs. The analysis showed equivalent results as for conventional ADAP knockout mice: impaired thymocyte development in ADAPfl/fl Lck-Cre mice, normal NK cell and myeloid cell distribution in ADAPfl/fl NKp46-Cre mice and ADAPfl/fl LysM-Cre mice, respectively as well as thrombocytopenia in ADAPfl/fl PF4-Cre mice. Active EAE was induced in these animals by immunization with the myelin oligodendrocyte glycoprotein35−55 peptide. The clinical course of EAE was significantly milder in mice with loss of ADAP in T cells, myeloid cells and NK cells compared to ADAP-sufficient control littermates. Surprisingly, specific deletion of ADAP in platelets resulted in a more exacerbated disease. These data show that T cell-independent as well as T cell-dependent mechanisms are responsible for the complex phenotype observed in conventional ADAP knockout mice.
Collapse
Affiliation(s)
- Jochen Rudolph
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Clara Meinke
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Martin Voss
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Karina Guttek
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Stefanie Kliche
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Dirk Reinhold
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Burkhart Schraven
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| | - Annegret Reinhold
- Institute for Molecular and Clinical Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Health Campus Immunology, Infectiology and Inflammation, Magdeburg, Germany
| |
Collapse
|
58
|
Baaklini CS, Rawji KS, Duncan GJ, Ho MFS, Plemel JR. Central Nervous System Remyelination: Roles of Glia and Innate Immune Cells. Front Mol Neurosci 2019; 12:225. [PMID: 31616249 PMCID: PMC6764409 DOI: 10.3389/fnmol.2019.00225] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/04/2019] [Indexed: 12/31/2022] Open
Abstract
In diseases such as multiple sclerosis (MS), inflammation can injure the myelin sheath that surrounds axons, a process known as demyelination. The spontaneous regeneration of myelin, called remyelination, is associated with restoration of function and prevention of axonal degeneration. Boosting remyelination with therapeutic intervention is a promising new approach that is currently being tested in several clinical trials. The endogenous regulation of remyelination is highly dependent on the immune response. In this review article, we highlight the cell biology of remyelination and its regulation by innate immune cells. For the purpose of this review, we discuss the roles of microglia, and also astrocytes and oligodendrocyte progenitor cells (OPCs) as they are being increasingly recognized to have immune cell functions.
Collapse
Affiliation(s)
- Charbel S. Baaklini
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Khalil S. Rawji
- Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Greg J. Duncan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, United States
| | - Madelene F. S. Ho
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
59
|
Chronic inflammation in multiple sclerosis - seeing what was always there. Nat Rev Neurol 2019; 15:582-593. [PMID: 31420598 DOI: 10.1038/s41582-019-0240-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 12/18/2022]
Abstract
Activation of innate immune cells and other compartmentalized inflammatory cells in the brains and spinal cords of people with relapsing-remitting multiple sclerosis (MS) and progressive MS has been well described histopathologically. However, conventional clinical MRI is largely insensitive to this inflammatory activity. The past two decades have seen the introduction of quantitative dynamic MRI scanning with contrast agents that are sensitive to the reduction in blood-brain barrier integrity associated with inflammation and to the trafficking of inflammatory myeloid cells. New MRI imaging sequences provide improved contrast for better detection of grey matter lesions. Quantitative lesion volume measures and magnetic resonance susceptibility imaging are sensitive to the activity of macrophages in the rims of white matter lesions. PET and magnetic resonance spectroscopy methods can also be used to detect contributions from innate immune activation in the brain and spinal cord. Some of these advanced research imaging methods for visualization of chronic inflammation are practical for relatively routine clinical applications. Observations made with the use of these techniques suggest ways of stratifying patients with MS to improve their care. The imaging methods also provide new tools to support the development of therapies for chronic inflammation in MS.
Collapse
|
60
|
Grassi S, Mauri L, Prioni S, Cabitta L, Sonnino S, Prinetti A, Giussani P. Sphingosine 1-Phosphate Receptors and Metabolic Enzymes as Druggable Targets for Brain Diseases. Front Pharmacol 2019; 10:807. [PMID: 31427962 PMCID: PMC6689979 DOI: 10.3389/fphar.2019.00807] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022] Open
Abstract
The central nervous system is characterized by a high content of sphingolipids and by a high diversity in terms of different structures. Stage- and cell-specific sphingolipid metabolism and expression are crucial for brain development and maintenance toward adult age. On the other hand, deep dysregulation of sphingolipid metabolism, leading to altered sphingolipid pattern, is associated with the majority of neurological and neurodegenerative diseases, even those totally lacking a common etiological background. Thus, sphingolipid metabolism has always been regarded as a promising pharmacological target for the treatment of brain disorders. However, any therapeutic hypothesis applied to complex amphipathic sphingolipids, components of cellular membranes, has so far failed probably because of the high regional complexity and specificity of the different biological roles of these structures. Simpler sphingosine-based lipids, including ceramide and sphingosine 1-phosphate, are important regulators of brain homeostasis, and, thanks to the relative simplicity of their metabolic network, they seem a feasible druggable target for the treatment of brain diseases. The enzymes involved in the control of the levels of bioactive sphingoids, as well as the receptors engaged by these molecules, have increasingly allured pharmacologists and clinicians, and eventually fingolimod, a functional antagonist of sphingosine 1-phosphate receptors with immunomodulatory properties, was approved for the therapy of relapsing-remitting multiple sclerosis. Considering the importance of neuroinflammation in many other brain diseases, we would expect an extension of the use of such analogs for the treatment of other ailments in the future. Nevertheless, many aspects other than neuroinflammation are regulated by bioactive sphingoids in healthy brain and dysregulated in brain disease. In this review, we are addressing the multifaceted possibility to address the metabolism and biology of bioactive sphingosine 1-phosphate as novel targets for the development of therapeutic paradigms and the discovery of new drugs.
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Livia Cabitta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
61
|
Sadler KE, Lewis TR, Waltz TB, Besharse JC, Stucky CL. Peripheral nerve pathology in sickle cell disease mice. Pain Rep 2019; 4:e765. [PMID: 31579856 PMCID: PMC6728004 DOI: 10.1097/pr9.0000000000000765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/02/2019] [Accepted: 05/19/2019] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION Many patients with sickle cell disease (SCD) suffer from chronic pain, which is often described as neuropathic in nature. Although vascular and inflammatory pathology undoubtedly contribute to the SCD pain experience, the nociceptive signals that ultimately drive symptoms are detected and transmitted by peripheral sensory neurons. To date, no systematic histological examination of peripheral nerves has been completed in patients or mouse models of SCD to diagnose disease-related neuropathy. OBJECTIVES In this brief report, we compared peripheral nerve morphology in tissues obtained from Berkeley transgenic SCD mice and control animals. METHODS Sciatic nerves were visualized using light and transmission electron microscopy. Myelin basic protein expression was assessed through Western blot. Blood-nerve barrier permeability was measured using Evan's blue plasma extravasation. RESULTS Peripheral fibers from SCD mice have thinner myelin sheaths than control mice and widespread myelin instability as evidenced by myelin sheath infolding and unwrapping. Deficits are also observed in nonmyelinating Schwann cell structures; Remak bundles from SCD nerves contain fewer C fibers, some of which are not fully ensheathed by the corresponding Schwann cell. Increased blood-nerve barrier permeability and expression of myelin basic protein are noted in SCD tissue. CONCLUSIONS These data are the first to characterize Berkeley SCD mice as a naturally occurring model of peripheral neuropathy. Widespread myelin instability is observed in nerves from SCD mice. This pathology may be explained by increased permeability of the blood-nerve barrier and, thus, increased access to circulating demyelinating agents at the level of primary sensory afferents.
Collapse
Affiliation(s)
- Katelyn E. Sadler
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tylor R. Lewis
- Department of Ophthalmology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tyler B. Waltz
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Joseph C. Besharse
- Department of Ophthalmology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
62
|
Sucksdorff M, Tuisku J, Matilainen M, Vuorimaa A, Smith S, Keitilä J, Rokka J, Parkkola R, Nylund M, Rinne J, Rissanen E, Airas L. Natalizumab treatment reduces microglial activation in the white matter of the MS brain. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e574. [PMID: 31355310 PMCID: PMC6624093 DOI: 10.1212/nxi.0000000000000574] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/02/2019] [Indexed: 01/31/2023]
Abstract
Objective To evaluate whether natalizumab treatment reduces microglial activation in MS. Methods We measured microglial activation using the 18-kDa translocator protein (TSPO)-binding radioligand [11C]PK11195 and PET imaging in 10 patients with MS before and after 1 year treatment with natalizumab. Microglial activation was evaluated as the distribution volume ratio (DVR) of the specifically bound radioligand in brain white and gray matter regions of interest. MRI and disability measurements were performed for comparison. Evaluation was performed identically with 11 age- and sex-matched patients with MS who had no MS therapy. Results Natalizumab treatment reduced microglial activation in the normal-appearing white matter (NAWM; baseline DVR vs DVR after 1 year of treatment 1.25 vs 1.22, p = 0.014, Wilcoxon) and at the rim of chronic lesions (baseline DVR vs DVR after 1 year of treatment 1.24 vs 1.18, p = 0.014). In patients with MS with no treatment, there was an increase in microglial activation at the rim of chronic lesions (1.23 vs 1.27, p = 0.045). No alteration was observed in microglial activation in gray matter areas. In the untreated patient group, higher microglial activation at baseline was associated with more rapid disability progression during an average of 4 years of follow-up. Conclusions TSPO-PET imaging can be used as a tool to assess longitudinal changes in microglial activation in the NAWM and in the perilesional areas in the MS brain in vivo. Natalizumab treatment reduces the diffuse compartmentalized CNS inflammation related to brain resident innate immune cells.
Collapse
Affiliation(s)
- Marcus Sucksdorff
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Markus Matilainen
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Anna Vuorimaa
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Sarah Smith
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Joonas Keitilä
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Johanna Rokka
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Riitta Parkkola
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Marjo Nylund
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Juha Rinne
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Eero Rissanen
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| | - Laura Airas
- Turku PET Centre (M.S., J.T., M.M., A.V., S.S., J.K., J. Rokka, M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; Division of Clinical Neurosciences (M.S., M.N., J. Rinne, E.R., L.A.), Turku University Hospital and University of Turku; and Department of Radiology (R.P.), University Hospital and University of Turku, Finland
| |
Collapse
|
63
|
Mack SJ, Udell J, Cohen F, Osoegawa K, Hawbecker SK, Noonan DA, Ladner MB, Goodridge D, Trachtenberg EA, Oksenberg JR, Erlich HA. High resolution HLA analysis reveals independent class I haplotypes and amino-acid motifs protective for multiple sclerosis. Genes Immun 2019; 20:308-326. [PMID: 29307888 PMCID: PMC6035897 DOI: 10.1038/s41435-017-0006-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/31/2017] [Accepted: 08/11/2017] [Indexed: 11/24/2022]
Abstract
We investigated association between HLA class I and class II alleles and haplotypes, and KIR loci and their HLA class I ligands, with multiple sclerosis (MS) in 412 European American MS patients and 419 ethnically matched controls, using next-generation sequencing. The DRB1*15:01~DQB1*06:02 haplotype was highly predisposing (odds ratio (OR) = 3.98; 95% confidence interval (CI) = 3-5.31; p-value (p) = 2.22E-16), as was DRB1*03:01~DQB1*02:01 (OR = 1.63; CI = 1.19-2.24; p = 1.41E-03). Hardy-Weinberg (HW) analysis in MS patients revealed a significant DRB1*03:01~DQB1*02:01 homozyote excess (15 observed; 8.6 expected; p = 0.016). The OR for this genotype (5.27; CI = 1.47-28.52; p = 0.0036) suggests a recessive MS risk model. Controls displayed no HW deviations. The C*03:04~B*40:01 haplotype (OR = 0.27; CI = 0.14-0.51; p = 6.76E-06) was highly protective for MS, especially in haplotypes with A*02:01 (OR = 0.15; CI = 0.04-0.45; p = 6.51E-05). By itself, A*02:01 is moderately protective, (OR = 0.69; CI = 0.54-0.87; p = 1.46E-03), and haplotypes of A*02:01 with the HLA-B Thr80 Bw4 variant (Bw4T) more so (OR = 0.53; CI = 0.35-0.78; p = 7.55E-04). Protective associations with the Bw4 KIR ligand resulted from linkage disequilibrium (LD) with DRB1*15:01, but the Bw4T variant was protective (OR = 0.64; CI = 0.49-0.82; p = 3.37-04) independent of LD with DRB1*15:01. The Bw4I variant was not associated with MS. Overall, we find specific class I HLA polymorphisms to be protective for MS, independent of the strong predisposition conferred by DRB1*15:01.
Collapse
Affiliation(s)
- Steven J Mack
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA.
| | - Julia Udell
- University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Franziska Cohen
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Kazutoyo Osoegawa
- Histocompatibility, Immunogenetics & Disease Profiling Laboratory, Stanford Blood Center, Palo Alto, CA, USA
| | - Sharon K Hawbecker
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - David A Noonan
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Martha B Ladner
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | | | | | - Jorge R Oksenberg
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Henry A Erlich
- Center for Genetics, Children's Hospital Oakland Research Institute, Oakland, CA, USA
| |
Collapse
|
64
|
Abdelhak A, Hottenrott T, Morenas-Rodríguez E, Suárez-Calvet M, Zettl UK, Haass C, Meuth SG, Rauer S, Otto M, Tumani H, Huss A. Glial Activation Markers in CSF and Serum From Patients With Primary Progressive Multiple Sclerosis: Potential of Serum GFAP as Disease Severity Marker? Front Neurol 2019; 10:280. [PMID: 30972011 PMCID: PMC6443875 DOI: 10.3389/fneur.2019.00280] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
Background: In progressive multiple sclerosis (MS), glial activation is thought to be a relevant mechanism of disability progression. Therefore, in vivo assessment of the glial cell activity is, in the emerging treatment era of primary progressive MS (PPMS), more important than ever. Objectives: To test the association of cerebrospinal fluid (CSF) and serum markers of glial activation in PPMS patients; including glial fibrillary acidic protein (GFAP), chitinase-3-like protein 1 (CHI3L1), soluble variant of triggering receptor expressed on myeloid cells 2 (sTREM2), and marker of neuroaxonal damage (Neurofilament light chain, NfL) as well as clinical severity. Methods: CSF and serum samples from PPMS patients were collected in the MS-centers at Universities of Freiburg (n = 49), Ulm (n = 27), Muenster (n = 11), and Rostock (n = 6). sTREM2 and CHI3L1 levels were measured using the previously reported ELISA assays, while NfL and GFAP were measured using SIMOA assays. Clinical data included age, gender, disease duration, treatment status, and Expanded Disability Status Scale (EDSS). Results: 93 CSF samples and 71 matching serum samples were analyzed. The median age of patients was 49 years and disease duration 4.5 years. GFAPserum correlated with EDSS after correction for age (β = 0.3, p = 0.001). Furthermore, EDSS was higher in patients with a GFAPserum level ≥ 151.7 pg/ml compared to patients with GFAPserum below this cut-off (5.5 vs. 4.0, p = 0.009). Other markers did not correlate with the clinical severity. Moreover, we found a correlation between NfLCSF and GFAPCSF, sTREM2 and CHI3L1 (ρ = 0.4 for GFAPCSF and sTREM2, ρ = 0.3 for CHI3L1, p < 0.01 for sTREM2 and CHI3L1 and <0.001 for GFAPCSF). CHI3L1 did not correlate with GFAPCSF but with sTREM2 (ρ = 0.4, p < 0.01). Discussion: The correlation between the glial activation markers in CSF with the markers of neuroaxonal demise supports the notion of the glial involvement in PPMS. The positive correlation between GFAPCSF with disease duration and GFAPserum with the clinical severity of the disease may highlight a particular role of the astrocytes in PPMS and mark the potential of GFAPserum as a disease severity marker.
Collapse
Affiliation(s)
- Ahmed Abdelhak
- Department of Neurology, University Hospital of Tuebingen, Tuebingen, Germany.,Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Tilman Hottenrott
- Department of Neurology, University Hospital of Freiburg, Freiburg, Germany
| | - Estrella Morenas-Rodríguez
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,BioMedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Uwe K Zettl
- Neuroimmunological Section, Department of Neurology, University Hospital of Rostock, Rostock, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,BioMedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sven G Meuth
- Department of Neurology, University Hospital of Muenster, Münster, Germany
| | - Sebastian Rauer
- Department of Neurology, University Hospital of Freiburg, Freiburg, Germany
| | - Markus Otto
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, University Hospital of Ulm, Ulm, Germany.,Specialty Hospital Dietenbronn, Schwendi, Germany
| | - André Huss
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
65
|
Rocco P, Eberini I, Musazzi UM, Franzè S, Minghetti P. Glatiramer acetate: A complex drug beyond biologics. Eur J Pharm Sci 2019; 133:8-14. [PMID: 30902653 DOI: 10.1016/j.ejps.2019.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/06/2019] [Accepted: 03/15/2019] [Indexed: 01/18/2023]
Abstract
Complex drugs may be either biological, if the active ingredients are derived from a biological source, or non-biological, if obtained by chemical synthesis. In both cases, their quality depends considerably on the manufacturing process. In the case of Non Biological Complex Drugs (NBCDs), complexity may arise either from the active substance, as in the case of glatiramer acetate, or from other sources, such as the formulation, as in the case of liposomes. In this paper, the case of glatiramer acetate (GA) - a NBCD relevant for clinical and economic reasons - is considered and the differences between US and EU regulatory approaches to GA marketing authorization are highlighted. Indeed, though US and EU regulatory agencies have chosen a generic approach integrated with additional data the implementation is different in the two jurisdictions. In the US, the additional data required are listed in a product specific guideline and copies of Copaxone® have been approved as generics. In the EU, instead regulatory agencies followed a hybrid approach requiring an additional comparative study, and interchangeability policies and substitution schemes have been left to national agencies.
Collapse
Affiliation(s)
- Paolo Rocco
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via G. Colombo, 71, 20133 Milan, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via G. Balzaretti, 9, 20133 Milan, Italy
| | - Umberto M Musazzi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via G. Colombo, 71, 20133 Milan, Italy
| | - Silvia Franzè
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via G. Colombo, 71, 20133 Milan, Italy
| | - Paola Minghetti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via G. Colombo, 71, 20133 Milan, Italy.
| |
Collapse
|
66
|
Human Remyelination Promoting Antibody Stimulates Astrocytes Proliferation Through Modulation of the Sphingolipid Rheostat in Primary Rat Mixed Glial Cultures. Neurochem Res 2018; 44:1460-1474. [PMID: 30569280 DOI: 10.1007/s11064-018-2701-x] [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: 08/04/2018] [Revised: 11/22/2018] [Accepted: 12/12/2018] [Indexed: 01/17/2023]
Abstract
Remyelination promoting human IgMs effectively increase the number of myelinated axons in animal models of multiple sclerosis. Hence, they ultimately stimulate myelin production by oligodendrocytes (OLs); however, their exact mechanism of action remains to be elucidated, and in particular, it remains unclear whether they are directly targeting OLs, or their action is mediated by effects on other cell types. We assessed the effect of remyelination promoting antibody rHIgM22 on the proliferative response and on the ceramide/sphingosine 1-phosphate rheostat in mixed glial cell cultures (MGCs). rHIgM22 treatment caused a time-dependent increase in PDGFαR protein in MGCs. Forty-eight hours of treatment with rHIgM22 induced a dose-dependent proliferative response (evaluated as total cell number and as EdU(+) cell number) in MGCs. When the proliferation response of MGCs to rHIgM22 was analyzed as a function of the cell types, the most significant proliferative response was associated with GLAST(+) cells, i.e., astrocytes. In many cell types, the balance between different sphingolipid mediators (the "sphingolipid rheostat"), in particular ceramide and sphingosine 1-phosphate, is critical in determining the cell fate. rHIgM22 treatment in MGCs induced a moderate but significant inhibition of total acidic sphingomyelinase activity (measured in vitro on cell lysates), the main enzyme responsible for the stimulus-mediated production of ceramide, when treatment was performed in serum containing medium, but no significant differences were observed when antibody treatment was performed in the absence of serum. Moreover, rHIgM22 treatment, either in the presence or in absence of serum, had no effects on ceramide levels. On the other hand, rHIgM22 treatment for 24 h induced increased production and release of sphingosine 1-phosphate in the extracellular milieu of MGC. Release of sphingosine 1-phosphate upon rHIgM22 treatment was strongly reduced by a selective inhibitor of PDGFαR. Increased sphingosine 1-phosphate production does not seem to be mediated by regulation of the biosynthetic enzymes, sphingosine kinase 1 and 2, since protein levels of these enzymes and phosphorylation of sphingosine kinase 1 were unchanged upon rHIgM22 treatment. Instead, we observed a significant reduction in the levels of sphingosine 1-phosphate lyase 1, one of the key catabolic enzymes. Remarkably, rHIgM22 treatment under the same experimental conditions did not induce changes in the production and/or release of sphingosine 1-phosphate in pure astrocyte cultures. Taken together, these data suggest that rHIgM22 indirectly influences the proliferation of astrocytes in MGCs, by affecting the ceramide/sphingosine 1-phosphate balance. The specific cell population directly targeted by rHIgM22 remains to be identified, however our study unveils another aspect of the complexity of rHIgM22-induced remyelinating effect.
Collapse
|
67
|
Yin C, Wu C, Du X, Fang Y, Pu J, Wu J, Tang L, Zhao W, Weng Y, Guo X, Chen G, Wang Z. PRL2 Controls Phagocyte Bactericidal Activity by Sensing and Regulating ROS. Front Immunol 2018; 9:2609. [PMID: 30483267 PMCID: PMC6244668 DOI: 10.3389/fimmu.2018.02609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/23/2018] [Indexed: 01/06/2023] Open
Abstract
Although it is well-recognized that inflammation enhances leukocyte bactericidal activity, the underlying mechanisms are not clear. Here we report that PRL2 is sensitive to oxidative stress at inflamed sites. Reduced PRL2 in phagocytes causes increased respiratory burst activity and enhances phagocyte bactericidal activity. PRL2 (Phosphatase Regenerating Liver 2) is highly expressed in resting immune cells, but is markedly downregulated by inflammation. in vitro experiments showed that PRL2 was sensitive to hydrogen peroxide (H2O2), a common damage signal at inflamed sites. In response to infection, PRL2 knockout (KO) phagocytes were hyper activated, produced more reactive oxygen species (ROS) and exhibited enhanced bactericidal activity. Mice with PRL2 deficiency in the myeloid cell compartment were resistant to lethal listeria infection and cleared the bacteria more rapidly and effectively. Moreover, in vitro experiments demonstrated that PRL2 binds to GTPase Rac and regulates ROS production. Rac GTPases were more active in PRL2 (KO) phagocytes than in wild type cells after bacterium infection. Our findings indicate that PRL2 senses ROS at inflamed sites and regulates ROS production in phagocytes. This positive feedback mechanism promotes bactericidal activity of phagocytes and may play an important role in innate anti-bacterial immunity.
Collapse
Affiliation(s)
- Cennan Yin
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chenyun Wu
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xinyue Du
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Fang
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Juebiao Pu
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianhua Wu
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lili Tang
- Department of Basic Medicine, Guangxi Medical University, Nanning, China
| | - Wei Zhao
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yongqiang Weng
- Department of General Surgery, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaokui Guo
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guangjie Chen
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaojun Wang
- Department of Immunology and Microbiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
68
|
Perriot S, Mathias A, Perriard G, Canales M, Jonkmans N, Merienne N, Meunier C, El Kassar L, Perrier AL, Laplaud DA, Schluep M, Déglon N, Du Pasquier R. Human Induced Pluripotent Stem Cell-Derived Astrocytes Are Differentially Activated by Multiple Sclerosis-Associated Cytokines. Stem Cell Reports 2018; 11:1199-1210. [PMID: 30409508 PMCID: PMC6234919 DOI: 10.1016/j.stemcr.2018.09.015] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 02/08/2023] Open
Abstract
Recent studies highlighted the importance of astrocytes in neuroinflammatory diseases, interacting closely with other CNS cells but also with the immune system. However, due to the difficulty in obtaining human astrocytes, their role in these pathologies is still poorly characterized. Here, we develop a serum-free protocol to differentiate human induced pluripotent stem cells (hiPSCs) into astrocytes. Gene expression and functional assays show that our protocol consistently yields a highly enriched population of resting mature astrocytes across the 13 hiPSC lines differentiated. Using this model, we first highlight the importance of serum-free media for astrocyte culture to generate resting astrocytes. Second, we assess the astrocytic response to IL-1β, TNF-α, and IL-6, all cytokines important in neuroinflammation, such as multiple sclerosis. Our study reveals very specific profiles of reactive astrocytes depending on the triggering stimulus. This model provides ideal conditions for in-depth and unbiased characterization of astrocyte reactivity in neuroinflammatory conditions.
Collapse
Affiliation(s)
- Sylvain Perriot
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Amandine Mathias
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Guillaume Perriard
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Mathieu Canales
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Nils Jonkmans
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Nicolas Merienne
- Laboratory of Neurotherapies and NeuroModulation, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Cécile Meunier
- Laboratory of Neurotherapies and NeuroModulation, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Lina El Kassar
- Institute for Stem Cell Therapy and Exploration of Monogenic Diseases (I-Stem), Corbeil-Essonnes, France
| | - Anselme L Perrier
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR861, I-Stem, AFM, Corbeil-Essonnes, France
| | - David-Axel Laplaud
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Myriam Schluep
- Service of Neurology, Department of Clinical Neurosciences, CHUV, CHUV BH-10/131, 46, rue du Bugnon, Lausanne 1011, Switzerland
| | - Nicole Déglon
- Laboratory of Neurotherapies and NeuroModulation, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Renaud Du Pasquier
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Service of Neurology, Department of Clinical Neurosciences, CHUV, CHUV BH-10/131, 46, rue du Bugnon, Lausanne 1011, Switzerland.
| |
Collapse
|
69
|
Manich G, Recasens M, Valente T, Almolda B, González B, Castellano B. Role of the CD200-CD200R Axis During Homeostasis and Neuroinflammation. Neuroscience 2018; 405:118-136. [PMID: 30367946 DOI: 10.1016/j.neuroscience.2018.10.030] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/27/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
Microglia are considered to be the resident macrophages of the CNS and main effector of immune brain function. Due to their essential role in the regulation of neuroinflammatory response, microglia constitute an important target for neurological diseases, such as multiple sclerosis, Alzheimer's or Parkinson's disease. The communication between neurons and microglia contributes to a proper maintenance of homeostasis in the CNS. Research developed in the last decade has demonstrated that this interaction is mediated by "Off-signals" - molecules exerting immune inhibition - and "On signals" - molecules triggering immune activation. Among "Off signals", molecular pair CD200 and its CD200R receptor, expressed mainly in the membrane of neurons and microglia, respectively, have centered our attention due to its unexplored and powerful immunoregulatory functions. In this review, we will offer an updated global view of the CD200-CD200R role in the microglia-neuron crosstalk during homeostasis and neuroinflammation. Specifically, the effects of CD200-CD200R in the inhibition of pro-inflammatory microglial activation will be explained, and their involvement in other functions such as homeostasis preservation, tissue repair, and brain aging, among others, will be pointed out. In addition, we will depict the effects of CD200-CD200R uncoupling in the etiopathogenesis of autoimmune and neurodegenerative diseases. Finally, we will explore how to translate the scientific evidence of CD200-CD200R interaction into possible clinical therapeutic strategies to tackle neuroinflammatory CNS diseases.
Collapse
Affiliation(s)
- Gemma Manich
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience. Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Mireia Recasens
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience. Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Tony Valente
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience. Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Beatriz Almolda
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience. Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain.
| | - Berta González
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience. Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Bernardo Castellano
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience. Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| |
Collapse
|
70
|
Bozic I, Tesovic K, Laketa D, Adzic M, Jakovljevic M, Bjelobaba I, Savic D, Nedeljkovic N, Pekovic S, Lavrnja I. Voltage Gated Potassium Channel Kv1.3 Is Upregulated on Activated Astrocytes in Experimental Autoimmune Encephalomyelitis. Neurochem Res 2018; 43:1020-1034. [PMID: 29574670 DOI: 10.1007/s11064-018-2509-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 12/13/2022]
Abstract
Kv1.3 is a voltage gated potassium channel that has been implicated in pathophysiology of multiple sclerosis (MS). In the present study we investigated temporal and cellular expression pattern of this channel in the lumbar part of spinal cords of animals with experimental autoimmune encephalomyelitis (EAE), animal model of MS. EAE was actively induced in female Dark Agouti rats. Expression of Kv1.3 was analyzed at different time points of disease progression, at the onset, peak and end of EAE. We here show that Kv1.3 increased by several folds at the peak of EAE at both gene and protein level. Double immunofluorescence analyses demonstrated localization of Kv1.3 on activated microglia, macrophages, and reactive astrocytes around inflammatory lesions. In vitro experiments showed that pharmacological block of Kv1.3 in activated astrocytes suppresses the expression of proinflammatory mediators, suggesting a role of this channel in inflammation. Our results support the hypothesis that Kv1.3 may be a therapeutic target of interest for MS and add astrocytes to the list of cells whose activation would be suppressed by inhibiting Kv1.3 in inflammatory conditions.
Collapse
Affiliation(s)
- Iva Bozic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia.
| | - Katarina Tesovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Danijela Laketa
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Adzic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Jakovljevic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Ivana Bjelobaba
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Danijela Savic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Nadezda Nedeljkovic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Sanja Pekovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| | - Irena Lavrnja
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Blvd Despota Stefana 142, 11060, Belgrade, Serbia
| |
Collapse
|
71
|
Elieh-Ali-Komi D, Cao Y. Role of Mast Cells in the Pathogenesis of Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. Clin Rev Allergy Immunol 2018; 52:436-445. [PMID: 28025778 DOI: 10.1007/s12016-016-8595-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune disorder of the central nervous system (CNS), characterized by recurrent episodes of inflammatory demyelination and consequent axonal deterioration. The hallmark of the disease is the demyelinated plaque, a hypocellular area characterized by formation of astrocytic scars and infiltration of mononuclear cells. Recent studies have revealed that both innate and adaptive immune cells contribute to the pathogenesis of MS and its experimental autoimmune encephalomyelitis (EAE) model. Here, we review the current understanding of the role of mast cells in the pathogenesis of MS and EAE. Mast cells may act at the early stage that promote demyelination through interactions among mast cells, neurons, and other immune cells to mediate neuroinflammation. Studies from EAE model suggest that mast cells regulate adaptive autoimmune responses, present myelin antigens to T cells, disrupt the blood-brain barrier, and permit the entry of inflammatory cells and mediators into the CNS. Depletion or limiting mast cells could be a new promising therapeutic target for MS and EAE.
Collapse
Affiliation(s)
- Daniel Elieh-Ali-Komi
- Immunology Research Center, Department of Immunology, and Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yonghao Cao
- Center for Synthetic Biology Engineering Research, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China. .,Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
72
|
O'Loughlin E, Madore C, Lassmann H, Butovsky O. Microglial Phenotypes and Functions in Multiple Sclerosis. Cold Spring Harb Perspect Med 2018; 8:8/2/a028993. [PMID: 29419406 DOI: 10.1101/cshperspect.a028993] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microglia are the resident immune cells that constantly survey the central nervous system. They can adapt to their environment and respond to injury or insult by altering their morphology, phenotype, and functions. It has long been debated whether microglial activation is detrimental or beneficial in multiple sclerosis (MS). Recently, the two opposing yet connected roles of microglial activation have been described with the aid of novel microglial markers, RNA profiling, and in vivo models. In this review, microglial phenotypes and functions in the context of MS will be discussed with evidence from both human pathological studies, in vitro and in vivo models. Microglial functional diversity-phagocytosis, antigen presentation, immunomodulation, support, and repair-will also be examined in detail. In addition, this review discusses the emerging evidence for microglia-related targets as biomarkers and therapeutic targets for MS.
Collapse
Affiliation(s)
- Elaine O'Loughlin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Charlotte Madore
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
73
|
Bove RM. Why monkeys do not get multiple sclerosis (spontaneously): An evolutionary approach. EVOLUTION MEDICINE AND PUBLIC HEALTH 2018; 2018:43-59. [PMID: 29492266 PMCID: PMC5824939 DOI: 10.1093/emph/eoy002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022]
Abstract
The goal of this review is to apply an evolutionary lens to understanding the origins of multiple sclerosis (MS), integrating three broad observations. First, only humans are known to develop MS spontaneously. Second, humans have evolved large brains, with characteristically large amounts of metabolically costly myelin. This myelin is generated over long periods of neurologic development—and peak MS onset coincides with the end of myelination. Third, over the past century there has been a disproportionate increase in the rate of MS in young women of childbearing age, paralleling increasing westernization and urbanization, indicating sexually specific susceptibility in response to changing exposures. From these three observations about MS, a life history approach leads us to hypothesize that MS arises in humans from disruption of the normal homeostatic mechanisms of myelin production and maintenance, during our uniquely long myelination period. This review will highlight under-explored areas of homeostasis in brain development, that are likely to shed new light on the origins of MS and to raise further questions about the interactions between our ancestral genes and modern environments.
Collapse
Affiliation(s)
- Riley M Bove
- Department of Neurology, UCSF, San Francisco, CA, USA
| |
Collapse
|
74
|
Veroni C, Serafini B, Rosicarelli B, Fagnani C, Aloisi F. Transcriptional profile and Epstein-Barr virus infection status of laser-cut immune infiltrates from the brain of patients with progressive multiple sclerosis. J Neuroinflammation 2018; 15:18. [PMID: 29338732 PMCID: PMC5771146 DOI: 10.1186/s12974-017-1049-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/21/2017] [Indexed: 01/01/2023] Open
Abstract
Background It is debated whether multiple sclerosis (MS) might result from an immunopathological response toward an active Epstein-Barr virus (EBV) infection brought into the central nervous system (CNS) by immigrating B cells. Based on this model, a relationship should exist between the local immune milieu and EBV infection status in the MS brain. To test this hypothesis, we analyzed expression of viral and cellular genes in brain-infiltrating immune cells. Methods Twenty-three postmortem snap-frozen brain tissue blocks from 11 patients with progressive MS were selected based on good RNA quality and prominent immune cell infiltration. White matter perivascular and intrameningeal immune infiltrates, including B cell follicle-like structures, were isolated from brain sections using laser capture microdissection. Enhanced PCR-based methods were used to investigate expression of 75 immune-related genes and 6 EBV genes associated with latent and lytic infection. Data were analyzed using univariate and multivariate statistical methods. Results Genes related to T cell activation, cytotoxic cell-mediated (or type 1) immunity, B cell growth and differentiation, pathogen recognition, myeloid cell function, type I interferon pathway activation, and leukocyte recruitment were found expressed at different levels in most or all MS brain immune infiltrates. EBV genes were detected in brain samples from 9 of 11 MS patients with expression patterns suggestive of in situ activation of latent infection and, less frequently, entry into the lytic cycle. Comparison of data obtained in meningeal and white matter infiltrates revealed higher expression of genes related to interferonγ production, B cell differentiation, cell proliferation, lipid antigen presentation, and T cell and myeloid cell recruitment, as well as more widespread EBV infection in the meningeal samples. Multivariate analysis grouped genes expressed in meningeal and white matter immune infiltrates into artificial factors that were characterized primarily by genes involved in type 1 immunity effector mechanisms and type I interferon pathway activation. Conclusion These results confirm profound in situ EBV deregulation and suggest orchestration of local antiviral function in the MS brain, lending support to a model of MS pathogenesis that involves EBV as possible antigenic stimulus of the persistent immune response in the central nervous system. Electronic supplementary material The online version of this article (10.1186/s12974-017-1049-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caterina Veroni
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Barbara Serafini
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Barbara Rosicarelli
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Corrado Fagnani
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
75
|
Liu Y, Gibson SA, Benveniste EN, Qin H. Opportunities for Translation from the Bench: Therapeutic Intervention of the JAK/STAT Pathway in Neuroinflammatory Diseases. Crit Rev Immunol 2018; 35:505-27. [PMID: 27279046 DOI: 10.1615/critrevimmunol.2016015517] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathogenic CD4+ T cells and myeloid cells play critical roles in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. These immune cells secrete aberrantly high levels of pro-inflammatory cytokines that pathogenically bridge the innate and adaptive immune systems and damage neurons and oligodendrocytes. These cytokines include interleukin-2 (IL-2), IL-6, IL-12, IL-21, IL-23, granulocyte macrophage-colony stimulating factor (GM-CSF), and interferon-γ (IFN-γ). It is, therefore, not surprising that both the dysregulated expression of these cytokines and the subsequent activation of their downstream signaling cascades is a common feature in MS/EAE. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is utilized by numerous cytokines for signal transduction and is essential for the development and regulation of immune responses. Unbridled activation of the JAK/STAT pathway by pro-inflammatory cytokines has been demonstrated to be critically involved in the pathogenesis of MS/EAE. In this review, we discuss recent advancements in our understanding of the involvement of the JAK/STAT signaling pathway in the pathogenesis of MS/EAE, with a particular focus on therapeutic approaches to target the JAK/STAT pathway.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294; Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sara A Gibson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| |
Collapse
|
76
|
White MPJ, Webster G, Leonard F, La Flamme AC. Innate IFN-γ ameliorates experimental autoimmune encephalomyelitis and promotes myeloid expansion and PDL-1 expression. Sci Rep 2018; 8:259. [PMID: 29321652 PMCID: PMC5762891 DOI: 10.1038/s41598-017-18543-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/13/2017] [Indexed: 11/26/2022] Open
Abstract
The innate immune system plays a central role in the immune-mediated pathology of multiple sclerosis, and is a therapeutic target for progressive disease. Recently, it has been demonstrated that MIS416, a novel immunomodulatory microparticle that activates NOD-2 and TLR-9-signaling, has disease-modifying activity in multiple sclerosis models. This activity is dependent on innate IFN-γ; however, the precise immune regulatory mechanisms amplified by MIS416 have not previously been determined. Using the experimental autoimmune encephalomyelitis model, MIS416 treatment was associated with IFN-γ–dependant expansion of Treg number and increased suppressive function; however, these cells did not account for disease reduction. Additionally, MIS416 treatment stimulated increased nitric oxide production that was IFN-γ–dependant but dispensable for protection. Finally, MIS416-mediated protection was shown to correlate with IFN-γ–dependant expansion of PDL-1-expressing peripheral myeloid cells, a subset of which was found to be selectively recruited to the brain. This central nervous system trafficking was independent of neuro-inflammatory signals as it occurred in MIS416-treated healthy mice. Together, these findings provide insight into regulatory myeloid cell activities amplified by MIS416-mediated NOD-2 and TLR-9 signalling and highlight the potential importance of these cells in accessing the brain where they may act locally and contribute to the control of neuroinflammation.
Collapse
Affiliation(s)
- Madeleine P J White
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Gill Webster
- Innate Immunotherapeutics, Auckland, New Zealand
| | - Faith Leonard
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.,Innate Immunotherapeutics, Auckland, New Zealand
| | - Anne Camille La Flamme
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand. .,Malaghan Institute of Medical Research, Wellington, New Zealand.
| |
Collapse
|
77
|
Toll-Like Receptor 2-Mediated Glial Cell Activation in a Mouse Model of Cuprizone-Induced Demyelination. Mol Neurobiol 2017; 55:6237-6249. [PMID: 29288338 DOI: 10.1007/s12035-017-0838-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/12/2017] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS) is a chronic degenerative disease of the central nervous system that is characterized by myelin abnormalities, oligodendrocyte pathology, and concomitant glia activation. The factors triggering gliosis and demyelination are currently not well characterized. New findings suggest an important role of the innate immune response in the initiation and progression of active demyelinating lesions. Especially during progressive disease, aberrant glia activation rather than the invasion of peripheral immune cells is accountable for progressive neuronal injury. The innate immune response can be induced by pathogen-associated or danger-associated molecular patterns, which are identified by pattern recognition receptors (PRRs), including the Toll-like receptors (TLRs). In this study, we used the cuprizone model in mice to investigate the expression of TLR2 during the course of cuprizone-induced demyelination. In addition, we used TLR2-deficient mice to analyze the functional role of TLR2 activation during cuprizone-induced demyelination and reactive gliosis. We show a significantly increased expression of TLR2 in the corpus callosum and hippocampus of cuprizone-intoxicated mice. The absence of receptor signaling in TLR2-deficient mice resulted in less severe reactive astrogliosis in the corpus callosum and cortex. In addition, microglia activation was ameliorated in the corpus callosum of TLR2-deficient mice, but augmented in the cortex compared to wild-type littermates. Extent of demyelination and loss of mature oligodendrocytes was comparable in both genotypes. These results suggest that the TLR2 orchestrates glia activation during gray and white matter demyelination in the presence of an intact blood-brain barrier. Future studies now have to address the underlying mechanisms of the region-specific TLR2-mediated glia activation.
Collapse
|
78
|
Dunham J, van de Vis R, Bauer J, Wubben J, van Driel N, Laman JD, ‘t Hart BA, Kap YS. Severe oxidative stress in an acute inflammatory demyelinating model in the rhesus monkey. PLoS One 2017; 12:e0188013. [PMID: 29136024 PMCID: PMC5685592 DOI: 10.1371/journal.pone.0188013] [Citation(s) in RCA: 13] [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: 05/17/2017] [Accepted: 10/29/2017] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress is increasingly implicated as a co-factor of tissue injury in inflammatory/demyelinating disorders of the central nervous system (CNS), such as multiple sclerosis (MS). While rodent experimental autoimmune encephalomyelitis (EAE) models diverge from human demyelinating disorders with respect to limited oxidative injury, we observed that in a non-human primate (NHP) model for MS, namely EAE in the common marmoset, key pathological features of the disease were recapitulated, including oxidative tissue injury. Here, we investigated the presence of oxidative injury in another NHP EAE model, i.e. in rhesus macaques, which yields an acute demyelinating disease, which may more closely resemble acute disseminated encephalomyelitis (ADEM) than MS. Rhesus monkey EAE diverges from marmoset EAE by abundant neutrophil recruitment into the CNS and destructive injury to white matter. This difference prompted us to investigate to which extent the oxidative pathway features elicited in MS and marmoset EAE are reflected in the acute rhesus monkey EAE model. The rhesus EAE brain was characterized by widespread demyelination and active lesions containing numerous phagocytic cells and to a lesser extent T cells. We observed induction of the oxidative stress pathway, including injury, with a predilection of p22phox expression in neutrophils and macrophages/microglia. In addition, changes in iron were observed. These results indicate that pathogenic mechanisms in the rhesus EAE model may differ from the marmoset EAE and MS brain due to the neutrophil involvement, but may in the end lead to similar induction of oxidative stress and injury.
Collapse
Affiliation(s)
- Jordon Dunham
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
- University Groningen, University Medical Center, Department of Neuroscience, Groningen, The Netherlands
| | - Reinofke van de Vis
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Jan Bauer
- Department Neuroimmunology, Brain Research Institute, Medical University, Vienna, Austria
| | - Jacqueline Wubben
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Nikki van Driel
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Jon D. Laman
- University Groningen, University Medical Center, Department of Neuroscience, Groningen, The Netherlands
| | - Bert A. ‘t Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
- University Groningen, University Medical Center, Department of Neuroscience, Groningen, The Netherlands
| | - Yolanda S. Kap
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
- * E-mail:
| |
Collapse
|
79
|
Modulation of Astrocyte Activity by Cannabidiol, a Nonpsychoactive Cannabinoid. Int J Mol Sci 2017; 18:ijms18081669. [PMID: 28788104 PMCID: PMC5578059 DOI: 10.3390/ijms18081669] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 07/26/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
The astrocytes have gained in recent decades an enormous interest as a potential target for neurotherapies, due to their essential and pleiotropic roles in brain physiology and pathology. Their precise regulation is still far from understood, although several candidate molecules/systems arise as promising targets for astrocyte-mediated neuroregulation and/or neuroprotection. The cannabinoid system and its ligands have been shown to interact and affect activities of astrocytes. Cannabidiol (CBD) is the main non-psychotomimetic cannabinoid derived from Cannabis. CBD is devoid of direct CB1 and CB2 receptor activity, but exerts a number of important effects in the brain. Here, we attempt to sum up the current findings on the effects of CBD on astrocyte activity, and in this way on central nervous system (CNS) functions, across various tested models and neuropathologies. The collected data shows that increased astrocyte activity is suppressed in the presence of CBD in models of ischemia, Alzheimer-like and Multiple-Sclerosis-like neurodegenerations, sciatic nerve injury, epilepsy, and schizophrenia. Moreover, CBD has been shown to decrease proinflammatory functions and signaling in astrocytes.
Collapse
|
80
|
Horng S, Therattil A, Moyon S, Gordon A, Kim K, Argaw AT, Hara Y, Mariani JN, Sawai S, Flodby P, Crandall ED, Borok Z, Sofroniew MV, Chapouly C, John GR. Astrocytic tight junctions control inflammatory CNS lesion pathogenesis. J Clin Invest 2017; 127:3136-3151. [PMID: 28737509 DOI: 10.1172/jci91301] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 05/26/2017] [Indexed: 02/06/2023] Open
Abstract
Lesions and neurologic disability in inflammatory CNS diseases such as multiple sclerosis (MS) result from the translocation of leukocytes and humoral factors from the vasculature, first across the endothelial blood-brain barrier (BBB) and then across the astrocytic glia limitans (GL). Factors secreted by reactive astrocytes open the BBB by disrupting endothelial tight junctions (TJs), but the mechanisms that control access across the GL are unknown. Here, we report that in inflammatory lesions, a second barrier composed of reactive astrocyte TJs of claudin 1 (CLDN1), CLDN4, and junctional adhesion molecule A (JAM-A) subunits is induced at the GL. In a human coculture model, CLDN4-deficient astrocytes were unable to control lymphocyte segregation. In models of CNS inflammation and MS, mice with astrocyte-specific Cldn4 deletion displayed exacerbated leukocyte and humoral infiltration, neuropathology, motor disability, and mortality. These findings identify a second inducible barrier to CNS entry at the GL. This barrier may be therapeutically targetable in inflammatory CNS disease.
Collapse
Affiliation(s)
- Sam Horng
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Anthony Therattil
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Sarah Moyon
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alexandra Gordon
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Karla Kim
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Azeb Tadesse Argaw
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Yuko Hara
- Friedman Brain Institute.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - John N Mariani
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Setsu Sawai
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Per Flodby
- Will Rogers Institute Pulmonary Research Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Edward D Crandall
- Will Rogers Institute Pulmonary Research Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zea Borok
- Will Rogers Institute Pulmonary Research Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael V Sofroniew
- Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Candice Chapouly
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| | - Gareth R John
- Friedman Brain Institute.,Corinne Goldsmith Dickinson Center for Multiple Sclerosis.,Department of Neurology, and
| |
Collapse
|
81
|
Boghozian R, McKenzie BA, Saito LB, Mehta N, Branton WG, Lu J, Baker GB, Noorbakhsh F, Power C. Suppressed oligodendrocyte steroidogenesis in multiple sclerosis: Implications for regulation of neuroinflammation. Glia 2017; 65:1590-1606. [PMID: 28707358 DOI: 10.1002/glia.23179] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 05/26/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Neurosteroids are reported to exert anti-inflammatory effects in several neurological disorders. We investigated the expression and actions of the neurosteroid, dehydroepiandrosterone (DHEA), and its more stable 3β-sulphated ester, DHEA-S, in MS and associated experimental models. CNS tissues from patients with MS and animals with experimental autoimmune encephalomyelitis (EAE) displayed reduced DHEA concentrations, accompanied by diminished expression of the DHEA-synthesizing enzyme CYP17A1 in oligodendrocytes (ODCs), in association with increased expression of inflammatory genes including interferon (IFN)-γ and interleukin (IL)-1β. CYP17A1 was expressed variably in different human neural cell types but IFN-γ exposure selectively reduced CYP17A1 detection in ODCs. DHEA-S treatment reduced IL-1β and -6 release from activated human myeloid cells with minimal effect on lymphocyte viability. Animals with EAE receiving DHEA-S treatment showed reduced Il1b and Ifng transcript levels in spinal cord compared to vehicle-treated animals with EAE. DHEA-S treatment also preserved myelin basic protein immunoreactivity and reduced axonal loss in animals with EAE, relative to vehicle-treated EAE animals. Neurobehavioral deficits were reduced in DHEA-S-treated EAE animals compared with vehicle-treated animals with EAE. Thus, CYP17A1 expression in ODCs and its product DHEA were downregulated in the CNS during inflammatory demyelination while DHEA-S provision suppressed neuroinflammation, demyelination, and axonal injury that was evident as improved neurobehavioral performance. These findings indicate that DHEA production is an immunoregulatory pathway within the CNS and its restoration represents a novel treatment approach for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Roobina Boghozian
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada.,Department of Medical Microbiology & Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Brienne A McKenzie
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada
| | - Leina B Saito
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada
| | - Ninad Mehta
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada
| | - William G Branton
- Department of, Medicine, University of Alberta Edmonton, Alberta, Canada
| | - JianQiang Lu
- Department of Laboratory Medicine & Pathology, University of Alberta Edmonton, Alberta, Canada
| | - Glen B Baker
- Depatment of Psychiatry, University of Alberta Edmonton, Alberta, Canada
| | - Farshid Noorbakhsh
- Department of Medical Microbiology & Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Christopher Power
- Department of Medical Microbiology & Immunology, University of Alberta Edmonton, Alberta, Canada.,Department of, Medicine, University of Alberta Edmonton, Alberta, Canada.,Depatment of Psychiatry, University of Alberta Edmonton, Alberta, Canada
| |
Collapse
|
82
|
Wekerle H. Brain Autoimmunity and Intestinal Microbiota: 100 Trillion Game Changers. Trends Immunol 2017; 38:483-497. [DOI: 10.1016/j.it.2017.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 02/17/2017] [Accepted: 03/31/2017] [Indexed: 02/07/2023]
|
83
|
Opazo MC, González PA, Flores BD, Venegas LF, Albornoz EA, Cisternas P, Bohmwald K, Nieto PA, Bueno SM, Kalergis AM, Riedel CA. Gestational Hypothyroxinemia Imprints a Switch in the Capacity of Astrocytes and Microglial Cells of the Offspring to React in Inflammation. Mol Neurobiol 2017; 55:4373-4387. [PMID: 28656482 DOI: 10.1007/s12035-017-0627-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
Hypothyroxinemia (Hpx) is a highly frequent condition characterized by low thyroxine (T4) and normal 3,3',5'-triiodothyronine (T3) and thyroid stimulating hormone (TSH) levels in the blood. Gestational Hpx is closely related to cognitive impairment in the human offspring. In animal models gestational Hpx causes impairment at glutamatergic synapsis, spatial learning, and the susceptibility to suffer strong autoimmune diseases like experimental autoimmune encephalomyelitis (EAE). However, the mechanisms underlying these phenotypes are unknown. On the other hand, it has been shown that astrocytes and microglia affect the outcome of EAE. In fact, the activation of astrocytes and microglia in the central nervous system (CNS) contributes to EAE progression. Thus, in this work, the reactivity of astrocytes and microglia from rats gestated in Hpx was evaluated aiming to understand whether these cells are targets of gestational Hpx. Interestingly, microglia derived from the offspring gestated in Hpx were less reactive compared to microglia derived from offspring gestated in euthyroidism. Instead, astrocytes derived from the offspring gestated in Hpx were significantly more reactive than the astrocytes from the offspring gestated in euthyroidism. This work contributes with novel information regarding the effects of gestational Hpx over astrocytes and microglia in the offspring. It suggests that astrocyte could react strongly to an inflammatory insult inducing neuronal death in the CNS.
Collapse
Affiliation(s)
- María C Opazo
- Laboratorio de Biología Celular y Farmacología, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Laboratorio de Biología Celular y Farmacología, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Betsi D Flores
- Laboratorio de Biología Celular y Farmacología, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis F Venegas
- Laboratorio de Biología Celular y Farmacología, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo A Albornoz
- Laboratorio de Biología Celular y Farmacología, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Cisternas
- Laboratorio de Biología Celular y Farmacología, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela A Nieto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Laboratorio de Biología Celular y Farmacología, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile. .,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
84
|
Gabbi P, Ribeiro LR, Jessié Martins G, Cardoso AS, Haupental F, Rodrigues FS, Machado AK, Sperotto Brum J, Medeiros Frescura Duarte MM, Schetinger MRC, da Cruz IBM, Flávia Furian A, Oliveira MS, Dos Santos ARS, Royes LFF, Fighera MR, de Freitas ML. Methylmalonate Induces Inflammatory and Apoptotic Potential: A Link to Glial Activation and Neurological Dysfunction. J Neuropathol Exp Neurol 2017; 76:160-178. [PMID: 28395089 DOI: 10.1093/jnen/nlw121] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Methylmalonic acid (MMA) accumulates in tissues in methylmalonic acidemia, a heterogeneous group of inherited childhood diseases characterized by neurological dysfunction, oxidative stress and neuroinflammation; it is associated with degeneration of striatal neurons and cerebral cortical atrophy. It is presently unknown, however, whether transient exposure to MMA in the neonatal period is sufficient to trigger inflammatory and apoptotic processes that lead to brain structural damage. Here, newborn mice were given a single intracerebroventricular dose of MMA at 12 hours after birth. Maze testing of 21- and 40-day-old mice showed that MMA-injected animals exhibited deficit in the working memory test but not in the reference test. MMA-injected mice showed increased levels of the reactive oxygen species marker 2',7'-dichlorofluorescein diacetate, tumor necrosis factor, interleukin-1β, caspases 1, 3, and 8, and increased acetylcholinesterase activity in the cortex, hippocampus and striatum. This was associated with increased astrocyte and microglial immunoreactivity in all brain regions. These findings suggest that transient exposure to MMA may alter the redox state and cause neuroinflammatory/apoptotic processes and glial activation during critical periods of brain development. Similar processes may underlie brain dysfunction and cognitive impairment in patients with methylmalonic acidemia.
Collapse
Affiliation(s)
- Patricia Gabbi
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM
| | - Leandro Rodrigo Ribeiro
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM
| | | | - Alexandra Seide Cardoso
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM
| | - Fernanda Haupental
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM
| | - Fernanda Silva Rodrigues
- Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica
| | - Alencar Kolinski Machado
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde
| | | | | | | | | | - Ana Flávia Furian
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde
| | - Mauro Schneider Oliveira
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde
| | - Adair Roberto Soares Dos Santos
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica.,Universidade Federal de Santa Catarina, Centro, de Programa Pós-graduação em Neurociências, de Ciências Biológicas
| | - Luiz Fernando Freire Royes
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica
| | - Michele Rechia Fighera
- Programa de Pós-Graduação em Farmacologia, Centro, de, Departamento Fisiologia e Farmacologia, de Ciências da Saúde.,Laboratório de Bioquímica do Exercício, Centro, de, Departamento Métodos e Técnicas Desportivas, de Educação Física e Desportos, UFSM.,Centro, de Departamento Neuropsiquiatria; de Ciências da Saúde, UFSM.,de Programa Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica
| | | |
Collapse
|
85
|
Pinoli M, Marino F, Cosentino M. Dopaminergic Regulation of Innate Immunity: a Review. J Neuroimmune Pharmacol 2017; 12:602-623. [PMID: 28578466 DOI: 10.1007/s11481-017-9749-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/28/2017] [Indexed: 12/13/2022]
Abstract
Dopamine (DA) is a neurotransmitter in the central nervous system as well as in peripheral tissues. Emerging evidence however points to DA also as a key transmitter between the nervous system and the immune system as well as a mediator produced and released by immune cells themselves. Dopaminergic pathways have received so far extensive attention in the adaptive branch of the immune system, where they play a role in health and disease such as multiple sclerosis, rheumatoid arthritis, cancer, and Parkinson's disease. Comparatively little is known about DA and the innate immune response, although DA may affect innate immune system cells such as dendritic cells, macrophages, microglia, and neutrophils. The present review aims at providing a complete and exhaustive summary of currently available evidence about DA and innate immunity, and to become a reference for anyone potentially interested in the fields of immunology, neurosciences and pharmacology. A wide array of dopaminergic drugs is used in therapeutics for non-immune indications, such as Parkinson's disease, hyperprolactinemia, shock, hypertension, with a usually favorable therapeutic index, and they might be relatively easily repurposed for immune-mediated disease, thus leading to innovative treatments at low price, with benefit for patients as well as for the healthcare systems.
Collapse
Affiliation(s)
- Monica Pinoli
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varese, VA, Italy
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varese, VA, Italy.
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, Via Ottorino Rossi n. 9, 21100, Varese, VA, Italy
| |
Collapse
|
86
|
Aarts SABM, Seijkens TTP, Kusters PJH, van der Pol SMA, Zarzycka B, Heijnen PDAM, Beckers L, den Toom M, Gijbels MJJ, Boon L, Weber C, de Vries HE, Nicolaes GAF, Dijkstra CD, Kooij G, Lutgens E. Inhibition of CD40-TRAF6 interactions by the small molecule inhibitor 6877002 reduces neuroinflammation. J Neuroinflammation 2017; 14:105. [PMID: 28494768 PMCID: PMC5427621 DOI: 10.1186/s12974-017-0875-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/26/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The influx of leukocytes into the central nervous system (CNS) is a key hallmark of the chronic neuro-inflammatory disease multiple sclerosis (MS). Strategies that aim to inhibit leukocyte migration across the blood-brain barrier (BBB) are therefore regarded as promising therapeutic approaches to combat MS. As the CD40L-CD40 dyad signals via TNF receptor-associated factor 6 (TRAF6) in myeloid cells to induce inflammation and leukocyte trafficking, we explored the hypothesis that specific inhibition of CD40-TRAF6 interactions can ameliorate neuro-inflammation. METHODS Human monocytes were treated with a small molecule inhibitor (SMI) of CD40-TRAF6 interactions (6877002), and migration capacity across human brain endothelial cells was measured. To test the therapeutic potential of the CD40-TRAF6-blocking SMI under neuro-inflammatory conditions in vivo, Lewis rats and C57BL/6J mice were subjected to acute experimental autoimmune encephalomyelitis (EAE) and treated with SMI 6877002 for 6 days (rats) or 3 weeks (mice). RESULTS We here show that a SMI of CD40-TRAF6 interactions (6877002) strongly and dose-dependently reduces trans-endothelial migration of human monocytes. Moreover, upon SMI treatment, monocytes displayed a decreased production of ROS, tumor necrosis factor (TNF), and interleukin (IL)-6, whereas the production of the anti-inflammatory cytokine IL-10 was increased. Disease severity of EAE was reduced upon SMI treatment in rats, but not in mice. However, a significant reduction in monocyte-derived macrophages, but not in T cells, that had infiltrated the CNS was eminent in both models. CONCLUSIONS Together, our results indicate that SMI-mediated inhibition of the CD40-TRAF6 pathway skews human monocytes towards anti-inflammatory cells with reduced trans-endothelial migration capacity, and is able to reduce CNS-infiltrated monocyte-derived macrophages during neuro-inflammation, but minimally ameliorates EAE disease severity. We therefore conclude that SMI-mediated inhibition of the CD40-TRAF6 pathway may represent a beneficial treatment strategy to reduce monocyte recruitment and macrophage activation in the CNS and has the potential to be used as a co-treatment to combat MS.
Collapse
Affiliation(s)
- Suzanne A. B. M. Aarts
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Tom T. P. Seijkens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Pascal J. H. Kusters
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Susanne M. A. van der Pol
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Barbara Zarzycka
- Department of Biochemistry, University of Maastricht, 6200 MD Maastricht, The Netherlands
| | - Priscilla D. A. M. Heijnen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Marion J. J. Gijbels
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
- Department of Pathology and Department of Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Louis Boon
- Bioceros, 3584 CM Utrecht, The Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Pettenkoferstraße 9, 80336 Munich, Germany
| | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Gerry A. F. Nicolaes
- Department of Biochemistry, University of Maastricht, 6200 MD Maastricht, The Netherlands
| | - Christine D. Dijkstra
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Pettenkoferstraße 9, 80336 Munich, Germany
| |
Collapse
|
87
|
Bihler K, Kress E, Esser S, Nyamoya S, Tauber SC, Clarner T, Stope MB, Pufe T, Brandenburg LO. Formyl Peptide Receptor 1-Mediated Glial Cell Activation in a Mouse Model of Cuprizone-Induced Demyelination. J Mol Neurosci 2017; 62:232-243. [PMID: 28466255 DOI: 10.1007/s12031-017-0924-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/17/2017] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is a chronic degenerative disease of the central nervous system that is characterized by myelin abnormalities, oligodendrocyte pathology, and concomitant glia activation. Unclear are the factors triggering gliosis and demyelination. New findings suggest an important role of the innate immune response in the initiation and progression of active demyelinating lesions. The innate immune response is induced by pathogen-associated or danger-associated molecular patterns, which are identified by pattern recognition receptors (PRRs), including the G-protein coupled with formyl peptide receptors (FPRs). Glial cells, the immune cells of the central nervous system, also express the PRRs. In this study, we used the cuprizone mice model to investigate the expression of the FPR1 in the course of cuprizone-induced demyelination In addition, we used FPR1-deficient mice to analyze glial cell activation through immunohistochemistry and real-time RT-PCR in cuprizone model. Our results revealed a significantly increased expression of FPR1 in the cortex of cuprizone-treated mice. FPR1-deficient mice showed a slight but significant decrease of demyelination in the corpus callosum compared to the wild-type mice. Furthermore, FPR1 deficiency resulted in reduced glial cell activation and mRNA expression of microglia/macrophages markers, as well as pro- and anti-inflammatory cytokines in the cortex, compared to wild-type mice after cuprizone-induced demyelination. Combined together, these results suggest that the FPR1 is an important part of the innate immune response in the course of cuprizone-induced demyelination.
Collapse
Affiliation(s)
- Kai Bihler
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Eugenia Kress
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Stefan Esser
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Stella Nyamoya
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Tim Clarner
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Lars-Ove Brandenburg
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
88
|
Nury T, Zarrouk A, Ragot K, Debbabi M, Riedinger JM, Vejux A, Aubourg P, Lizard G. 7-Ketocholesterol is increased in the plasma of X-ALD patients and induces peroxisomal modifications in microglial cells: Potential roles of 7-ketocholesterol in the pathophysiology of X-ALD. J Steroid Biochem Mol Biol 2017; 169:123-136. [PMID: 27041118 DOI: 10.1016/j.jsbmb.2016.03.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/09/2015] [Accepted: 03/31/2016] [Indexed: 01/08/2023]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a genetic disorder induced by a mutation in the ABCD1 gene, which causes the accumulation of very long-chain fatty acids in tissue and plasma. Oxidative stress may be a hallmark of X-ALD. In the plasma of X-ALD patients with different forms of the disease, characterized by high levels of C24:0 and C26:0, we observed the presence of oxidative stress revealed by decreased levels of GSH, α-tocopherol, and docosahexaenoic acid (DHA). We showed that oxidative stress caused the oxidation of cholesterol and linoleic acid, leading to the formation of cholesterol oxide derivatives oxidized at C7 (7-ketocholesterol (7KC), 7β-hydroxycholesterol (7β-OHC), and 7α-hydroxycholesrol (7α-OHC)) and of 9- and 13-hydroxyoctadecadienoic acids (9-HODE, 13-HODE), respectively. High levels of 7KC, 7β-OHC, 7α-OHC, 9-HODE and 13-HODE were found. As 7KC induces oxidative stress, inflammation and cell death, which could play key roles in the development of X-ALD, the impact of 7KC on the peroxisomal status was determined in microglial BV-2 cells. Indeed, environmental stress factors such as 7KC could exacerbate peroxisomal dysfunctions in microglial cells and thus determine the progression of the disease. 7KC induces oxiapoptophagy in BV-2 cells: overproduction of H2O2 and O2-, presence of cleaved caspase-3 and PARP, nuclear condensation and/or fragmentation; elevated [LC3-II/LC3-I] ratio, increased p62 levels. 7KC also induces several peroxisomal modifications: decreased Abcd1, Abcd2, Abcd3, Acox1 and/or Mfp2 mRNA and protein levels, increased catalase activity and decreased Acox1-activity. However, the Pex14 level was unchanged. It is suggested that high levels of 7KC in X-ALD patients could foster generalized peroxisomal dysfunction in microglial cells, which could in turn intensify brain damage.
Collapse
Affiliation(s)
- Thomas Nury
- Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270/Univ. Bourgogne Franche Comté/INSERM, Dijon, France
| | - Amira Zarrouk
- Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270/Univ. Bourgogne Franche Comté/INSERM, Dijon, France; Univ. Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS Nutrition - Functional Food & Vascular Health, Monastir, Tunisia; Univ. Sousse, Faculty of Medicine, Sousse, Tunisia
| | - Kévin Ragot
- SYSMEX, Department of Cytometry, Roissy, France
| | - Meryam Debbabi
- Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270/Univ. Bourgogne Franche Comté/INSERM, Dijon, France; Univ. Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS Nutrition - Functional Food & Vascular Health, Monastir, Tunisia
| | | | - Anne Vejux
- Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270/Univ. Bourgogne Franche Comté/INSERM, Dijon, France
| | - Patrick Aubourg
- INSERM UMR 1169, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Gérard Lizard
- Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270/Univ. Bourgogne Franche Comté/INSERM, Dijon, France.
| |
Collapse
|
89
|
Bellaver B, Dos Santos JP, Leffa DT, Bobermin LD, Roppa PHA, da Silva Torres IL, Gonçalves CA, Souza DO, Quincozes-Santos A. Systemic Inflammation as a Driver of Brain Injury: the Astrocyte as an Emerging Player. Mol Neurobiol 2017; 55:2685-2695. [PMID: 28421541 DOI: 10.1007/s12035-017-0526-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/06/2017] [Indexed: 12/20/2022]
Abstract
Severe systemic inflammation has strong effects on brain functions, promoting permanent neurocognitive dysfunction and high mortality rates. Additionally, hippocampal damage seems to be directly involved in this process and astrocytes play an important role in neuroinflammation and in the neuroimmune response. However, the contribution of the astrocytes to the pathology of acute brain dysfunction is not well understood. Recently, our group established a protocol for obtaining astrocyte cultures from mature brain to allow the characterization of these cells and their functions under pathologic conditions. The present study was designed to characterize astrocyte function after acute systemic inflammation induced by cecal ligation and perforation (CLP). Hippocampal astrocyte cultures from CLP animals presented increased levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, IL-18, and cyclooxygenase-2 and decreased levels of IL-10. This proinflammatory profile was accompanied by an increase in Toll-like receptor (TLR)2 mRNA expression levels and no change either in TLR4 or in vascular endothelial growth factor (VEGF) gene expression. These alterations were associated with increased expressions of p21, nuclear factor kappa B (NFκB), and inducible nitric oxide synthase (iNOS) in astrocytes from CLP animals. The same parameters were also evaluated in whole hippocampal tissue, but differences in this profile were found compared to hippocampal astrocyte cultures from CLP, reflecting an interaction between other central nervous system cell types, which may mask specific astrocytic changes. These results improve our understanding of the mechanisms by which astrocytes react against systemic inflammation, and suggest these cells to be potential targets for therapeutic modulation.
Collapse
Affiliation(s)
- Bruna Bellaver
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| | - João Paulo Dos Santos
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Douglas Teixeira Leffa
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Unidade de Experimentação Animal, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Paola Haack Amaral Roppa
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Iraci Lucena da Silva Torres
- Programa de Pós-Graduação em Medicina: Ciências Médicas, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Unidade de Experimentação Animal, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - André Quincozes-Santos
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
90
|
Debbabi M, Zarrouk A, Bezine M, Meddeb W, Nury T, Badreddine A, Karym EM, Sghaier R, Bretillon L, Guyot S, Samadi M, Cherkaoui-Malki M, Nasser B, Mejri M, Ben-Hammou S, Hammami M, Lizard G. Comparison of the effects of major fatty acids present in the Mediterranean diet (oleic acid, docosahexaenoic acid) and in hydrogenated oils (elaidic acid) on 7-ketocholesterol-induced oxiapoptophagy in microglial BV-2 cells. Chem Phys Lipids 2017; 207:151-170. [PMID: 28408132 DOI: 10.1016/j.chemphyslip.2017.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/05/2017] [Indexed: 01/12/2023]
Abstract
Increased levels of 7-ketocholesterol (7KC), which results mainly from cholesterol auto-oxidation, are often found in the plasma and/or cerebrospinal fluid of patients with neurodegenerative diseases and might contribute to activation of microglial cells involved in neurodegeneration. As major cellular dysfunctions are induced by 7KC, it is important to identify molecules able to impair its side effects. Since consumption of olive and argan oils, and fish is important in the Mediterranean diet, the aim of the study was to determine the ability of oleic acid (OA), a major compound of olive and argan oil, and docosahexaenoic acid (DHA) present in fatty fishes, such as sardines, to attenuate 7KC-induced cytotoxic effects. Since elaidic acid (EA), the trans isomer of OA, can be found in hydrogenated cooking oils and fried foods, its effects on 7KC-induced cytotoxicity were also determined. In murine microglial BV-2 cells, 7KC induces cell growth inhibition, mitochondrial dysfunctions, reactive oxygen species overproduction and lipid peroxidation, increased plasma membrane permeability and fluidity, nuclei condensation and/or fragmentation and caspase-3 activation, which are apoptotic characteristics, and an increased LC3-II/LC3-I ratio, which is a criterion of autophagy. 7KC is therefore a potent inducer of oxiapoptophagy (OXIdation+APOPTOsis+autoPHAGY) on BV-2 cells. OA and EA, but not DHA, also favor the accumulation of lipid droplets revealed with Masson's trichrome, Oil Red O, and Nile Red staining. The cytotoxicity of 7KC was strongly attenuated by OA and DHA. Protective effects were also observed with EA. However, 7KC-induced caspase-3 activation was less attenuated with EA. Different effects of OA and EA on autophagy were also observed. In addition, EA (but not OA) increased plasma membrane fluidity, and only OA (but not EA) was able to prevent the 7KC-induced increase in plasma membrane fluidity. Thus, in BV-2 microglial cells, the principal fatty acids of the Mediterranean diet (OA, DHA) were able to attenuate the major toxic effects of 7KC, thus reinforcing the interest of natural compounds present in the Mediterranean diet to prevent the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Meryam Debbabi
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia
| | - Amira Zarrouk
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia; Univ Sousse, Faculty of Medicine, Sousse, Tunisia
| | - Maryem Bezine
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Tunis El Manar - Pasteur Institut, Lab. 'Venoms & Therapeutic Biomolecules', Tunis, Tunisia
| | - Wiem Meddeb
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Carthage, Faculty of Sciences, Bizerte, Tunisia
| | - Thomas Nury
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France
| | - Asmaa Badreddine
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Lab of 'Biochemistry of Neuroscience', Univ. Hassan I, Settat, Morocco
| | - El Mostafa Karym
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Lab of 'Biochemistry of Neuroscience', Univ. Hassan I, Settat, Morocco
| | - Randa Sghaier
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France; Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia; Univ Sousse, Faculty of Medicine, Sousse, Tunisia
| | - Lionel Bretillon
- Eye & Nutrition Research Group, CSGA, UMR 1324 INRA, 6265 CNRS, Univ. Bourgogne Franche-Comté, Dijon, France
| | | | - Mohammad Samadi
- LCPMC-A2, ICPM, Département de Chimie, Université de Lorraine, Metz, France
| | - Mustapha Cherkaoui-Malki
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France
| | - Boubker Nasser
- Lab of 'Biochemistry of Neuroscience', Univ. Hassan I, Settat, Morocco
| | - Mondher Mejri
- Univ Carthage, Faculty of Sciences, Bizerte, Tunisia
| | - Sofien Ben-Hammou
- Department of Neurology, University Hospital Sahloul, 4000 Sousse, Tunisia
| | - Mohamed Hammami
- Univ Monastir, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir, Tunisia
| | - Gérard Lizard
- Univ Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Inserm, Dijon, France.
| |
Collapse
|
91
|
Bianchini E, De Biasi S, Simone AM, Ferraro D, Sola P, Cossarizza A, Pinti M. Invariant natural killer T cells and mucosal-associated invariant T cells in multiple sclerosis. Immunol Lett 2017; 183:1-7. [PMID: 28119072 DOI: 10.1016/j.imlet.2017.01.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is a chronic progressive inflammatory demyelinating disorder of the central nervous system, and in several countries is a leading cause of permanent neurological disability in young adults, particularly women. MS is considered an autoimmune disease, caused by an aberrant immune response to environmental triggers in genetically susceptible subjects. However, the contribution of the innate or of the adaptive immune system to the development and progression of the disease has not yet been fully elucidated. Innate-like T lymphocytes are unconventional T cells that bridge the innate and adaptive arms of the immune system, because they use a T cell receptor to sense external ligands, but behave like innate cells when they rapidly respond to stimuli. These cells could play an important role in the pathogenesis of MS. Here, we focus on invariant natural killer T (iNKT) cells and mucosal-associated invariant T (MAIT) cells, and we review the current knowledge on their biology and possible involvement in MS. Although several studies have evaluated the frequency and functions of iNKT and MAIT cells both in MS patients and in experimental mouse models, contradictory observations have been reported, and it is not clear whether they exert a protective or a pro-inflammatory and harmful role. A better understanding of how immune cells are involved in MS, and of their interactions could be of great interest for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Elena Bianchini
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Sara De Biasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Anna Maria Simone
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, Nuovo Ospedale Civile Sant'Agostino Estense, Via P. Giardini 1355, 41126 Modena, Italy
| | - Diana Ferraro
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, Nuovo Ospedale Civile Sant'Agostino Estense, Via P. Giardini 1355, 41126 Modena, Italy
| | - Patrizia Sola
- Neurology Unit, Department of Biomedical, Metabolic and Neurosciences, Nuovo Ospedale Civile Sant'Agostino Estense, Via P. Giardini 1355, 41126 Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences of Children and Adults, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy.
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| |
Collapse
|
92
|
Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS) characterized by loss of motor and sensory function that results from immune-mediated inflammation, demyelination, and subsequent axonal damage. Clinically, most MS patients experience recurrent episodes (relapses) of neurological impairment, but in most cases (60–80%) the course of the disease eventually becomes chronic and progressive, leading to cumulative motor, sensory, and visual disability, and cognitive deficits. The course of the disease is largely unpredictable and its clinical presentation is variable, but its predilection for certain parts of the CNS, which includes the optic nerves, the brain stem, cerebellum, and cervical spinal cord, provides a characteristic constellation of signs and symptoms. Several variants of MS have been nowadays defined with variable immunopathogenesis, course and prognosis. Many new treatments targeting the immune system have shown efficacy in preventing the relapses of MS and have been introduced to its management during the last decade.
Collapse
|
93
|
Cheng KY, Liu Y, Han YG, Li JK, Jia JL, Chen B, Yao ZX, Nie L, Cheng L. Follistatin-like protein 1 suppressed pro-inflammatory cytokines expression during neuroinflammation induced by lipopolysaccharide. J Mol Histol 2016; 48:63-72. [PMID: 27913976 DOI: 10.1007/s10735-016-9706-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/21/2016] [Indexed: 12/31/2022]
Abstract
Follistain-like protein 1 (FSTL1), has been recently demonstrated to be involved in the embryo development of nervous system and glioblastoma. However, the role of FSTL1 in neuroinflammation remains unexplored. In this study, the expression of FSTL1 in astrocytes was verified and its role was studied in neuroinflammation induced by in vivo intracerebroventricular (ICV) injection of lipopolysaccharide (LPS) or LPS treatment to astrocytes in vitro. FSTL1 was significantly induced after ICV LPS injection or LPS treatment. FSTL1 suppressed upregulation of pro-inflammatory cytokines in astrocytes after LPS treatment. Moreover, FSTL1 downregulated expression of pro-inflammatory cytokines through suppressing MAPK/p-ERK1/2 pathway in astrocytes. Our results suggest that FSTL1 may play an anti-inflammatory role in neuroinflammation mediated by astrocytes.
Collapse
Affiliation(s)
- Kai-Yuan Cheng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yi Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Ying-Guang Han
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Jing-Kun Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Jia-Lin Jia
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Bin Chen
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Zhi-Xiao Yao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Lin Nie
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Lei Cheng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
94
|
Ornelas IM, McLane LE, Saliu A, Evangelou AV, Khandker L, Wood TL. Heterogeneity in oligodendroglia: Is it relevant to mouse models and human disease? J Neurosci Res 2016; 94:1421-1433. [PMID: 27557736 PMCID: PMC5513674 DOI: 10.1002/jnr.23900] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 01/09/2023]
Abstract
There are many lines of evidence indicating that oligodendrocyte progenitor cells and oligodendrocyte populations in the central nervous system (CNS) are heterogeneous based on their developmental origins as well as from morphological and molecular criteria. Whether these distinctions reflect functional heterogeneity is less clear and has been the subject of considerable debate. Recent findings, particularly from knockout mouse models, have provided new evidence for regional variations in myelination phenotypes, particularly between brain and spinal cord. These data raise the possibility that oligodendrocytes in these regions have different functional capacities and/or ability to compensate for loss of a specific gene. The goal of this review is to briefly revisit the evidence for oligodendrocyte heterogeneity and then to present data from transgenic and demyelinating mouse models suggesting functional heterogeneity in myelination, demyelination, and remyelination in the CNS and, finally, to discuss the implications of these findings for human diseases. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Isis M Ornelas
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Lauren E McLane
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Aminat Saliu
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Angelina V Evangelou
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Luipa Khandker
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey.
| |
Collapse
|
95
|
Poutiainen P, Jaronen M, Quintana FJ, Brownell AL. Precision Medicine in Multiple Sclerosis: Future of PET Imaging of Inflammation and Reactive Astrocytes. Front Mol Neurosci 2016; 9:85. [PMID: 27695400 PMCID: PMC5023680 DOI: 10.3389/fnmol.2016.00085] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 08/30/2016] [Indexed: 12/29/2022] Open
Abstract
Non-invasive molecular imaging techniques can enhance diagnosis to achieve successful treatment, as well as reveal underlying pathogenic mechanisms in disorders such as multiple sclerosis (MS). The cooperation of advanced multimodal imaging techniques and increased knowledge of the MS disease mechanism allows both monitoring of neuronal network and therapeutic outcome as well as the tools to discover novel therapeutic targets. Diverse imaging modalities provide reliable diagnostic and prognostic platforms to better achieve precision medicine. Traditionally, magnetic resonance imaging (MRI) has been considered the golden standard in MS research and diagnosis. However, positron emission tomography (PET) imaging can provide functional information of molecular biology in detail even prior to anatomic changes, allowing close follow up of disease progression and treatment response. The recent findings support three major neuroinflammation components in MS: astrogliosis, cytokine elevation, and significant changes in specific proteins, which offer a great variety of specific targets for imaging purposes. Regardless of the fact that imaging of astrocyte function is still a young field and in need for development of suitable imaging ligands, recent studies have shown that inflammation and astrocyte activation are related to progression of MS. MS is a complex disease, which requires understanding of disease mechanisms for successful treatment. PET is a precise non-invasive imaging method for biochemical functions and has potential to enhance early and accurate diagnosis for precision therapy of MS. In this review we focus on modulation of different receptor systems and inflammatory aspect of MS, especially on activation of glial cells, and summarize the recent findings of PET imaging in MS and present the most potent targets for new biomarkers with the main focus on experimental MS research.
Collapse
Affiliation(s)
- Pekka Poutiainen
- Athinoula A Martinos Biomedical Imaging Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, MA, USA
| | - Merja Jaronen
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical SchoolBoston, MA, USA
| | - Francisco J. Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical SchoolBoston, MA, USA
| | - Anna-Liisa Brownell
- Athinoula A Martinos Biomedical Imaging Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, MA, USA
| |
Collapse
|
96
|
Ulivieri C, Savino MT, Luccarini I, Fanigliulo E, Aldinucci A, Bonechi E, Benagiano M, Ortensi B, Pelicci G, D'Elios MM, Ballerini C, Baldari CT. The Adaptor Protein Rai/ShcC Promotes Astrocyte-Dependent Inflammation during Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2016; 197:480-90. [PMID: 27288534 DOI: 10.4049/jimmunol.1502063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 05/13/2016] [Indexed: 01/13/2023]
Abstract
Th17 cells have been casually associated to the pathogenesis of autoimmune disease. We have previously demonstrated that Rai/ShcC, a member of the Shc family of adaptor proteins, negatively regulates Th17 cell differentiation and lupus autoimmunity. In this study, we have investigated the pathogenic outcome of the Th17 bias associated with Rai deficiency on multiple sclerosis development, using the experimental autoimmune encephalomyelitis (EAE) mouse model. We found that, unexpectedly, EAE was less severe in Rai(-/-) mice compared with their wild-type counterparts despite an enhanced generation of myelin-specific Th17 cells that infiltrated into the CNS. Nevertheless, when adoptively transferred into immunodeficient Rai(+/+) mice, these cells promoted a more severe disease compared with wild-type encephalitogenic Th17 cells. This paradoxical phenotype was caused by a dampened inflammatory response of astrocytes, which were found to express Rai, to IL-17. The results provide evidence that Rai plays opposite roles in Th17 cell differentiation and astrocyte activation, with the latter dominant over the former in EAE, highlighting this adaptor as a potential novel target for the therapy of multiple sclerosis.
Collapse
Affiliation(s)
- Cristina Ulivieri
- Department of Life Sciences, University of Siena, 2 53100, Siena, Italy;
| | | | | | | | | | - Elena Bonechi
- Department of Neurosciences, 6 50134 Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, 3 50134 Florence, Italy; and
| | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, 16 20139 Milan, Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, 16 20139 Milan, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, 3 50134 Florence, Italy; and
| | | | | |
Collapse
|
97
|
Mayo L, Cunha APD, Madi A, Beynon V, Yang Z, Alvarez JI, Prat A, Sobel RA, Kobzik L, Lassmann H, Quintana FJ, Weiner HL. IL-10-dependent Tr1 cells attenuate astrocyte activation and ameliorate chronic central nervous system inflammation. Brain 2016; 139:1939-57. [PMID: 27246324 PMCID: PMC4939696 DOI: 10.1093/brain/aww113] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 03/21/2016] [Indexed: 01/09/2023] Open
Abstract
See Winger and Zamvil (doi:
10.1093/brain/aww121
) for a scientific commentary on this article.
The innate immune system plays a central role in the chronic central nervous system inflammation that drives neurological disability in progressive forms of multiple sclerosis, for which there are no effective treatments. The mucosal immune system is a unique tolerogenic organ that provides a physiological approach for the induction of regulatory T cells. Here we report that nasal administration of CD3-specific antibody ameliorates disease in a progressive animal model of multiple sclerosis. This effect is IL-10-dependent and is mediated by the induction of regulatory T cells that share a similar transcriptional profile to Tr1 regulatory cells and that suppress the astrocyte inflammatory transcriptional program. Treatment results in an attenuated inflammatory milieu in the central nervous system, decreased microglia activation, reduced recruitment of peripheral monocytes, stabilization of the blood–brain barrier and less neurodegeneration. These findings suggest a new therapeutic approach for the treatment of progressive forms of multiple sclerosis and potentially other types of chronic central nervous system inflammation.
Collapse
Affiliation(s)
- Lior Mayo
- 1 Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA 2 Cell Research and Immunology Department, Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 699788, Israel
| | - Andre Pires Da Cunha
- 1 Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Asaf Madi
- 3 Evergrande Center for Immunologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vanessa Beynon
- 1 Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhiping Yang
- 4 Environmental Health Department, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jorge I Alvarez
- 5 Neuroimmunology Research Lab, CRCHUM, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada 6 Pathobiology Department, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandre Prat
- 5 Neuroimmunology Research Lab, CRCHUM, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | | | - Lester Kobzik
- 4 Environmental Health Department, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Hans Lassmann
- 8 Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Wien, Austria
| | - Francisco J Quintana
- 1 Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard L Weiner
- 1 Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
98
|
McMurran CE, Jones CA, Fitzgerald DC, Franklin RJM. CNS Remyelination and the Innate Immune System. Front Cell Dev Biol 2016; 4:38. [PMID: 27200350 PMCID: PMC4853384 DOI: 10.3389/fcell.2016.00038] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022] Open
Abstract
A misguided inflammatory response is frequently implicated in myelin damage. Particularly prominent among myelin diseases, multiple sclerosis (MS) is an autoimmune condition, with immune–mediated damage central to its etiology. Nevertheless, a robust inflammatory response is also essential for the efficient regeneration of myelin sheaths after such injury. Here, we discuss the functions of inflammation that promote remyelination, and how these have been experimentally disentangled from the pathological facets of the immune response. We focus on the contributions that resident microglia and monocyte-derived macrophages make to remyelination and compare the roles of these two populations of innate immune cells. Finally, the current literature is framed in the context of developing therapies that manipulate the innate immune response to promote remyelination in clinical myelin disease.
Collapse
Affiliation(s)
- Christopher E McMurran
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge Cambridge, UK
| | | | - Denise C Fitzgerald
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Science, Queens University Belfast Belfast, UK
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge Cambridge, UK
| |
Collapse
|
99
|
Burman J, Svenningsson A. Cerebrospinal fluid concentration of Galectin-9 is increased in secondary progressive multiple sclerosis. J Neuroimmunol 2016; 292:40-4. [DOI: 10.1016/j.jneuroim.2016.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/11/2016] [Accepted: 01/14/2016] [Indexed: 12/14/2022]
|
100
|
Zhang Y, Huang R, Zhang Y, Yi H, Bai Y, Chao J, Yao H. IL-17 induces MIP-1α expression in primary mouse astrocytes via TRPC channel. Inflammopharmacology 2016; 24:33-42. [PMID: 26782821 DOI: 10.1007/s10787-015-0256-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/14/2015] [Indexed: 10/22/2022]
Abstract
Our previous study demonstrated IL-17-mediated induction of MIP-1α through its binding to the cognate IL-17RA and MIP-1α was involved in astrocyte activation. Transient receptor potential canonical (TRPC) channel was involved in astrocyte activation, however, whether TRPC channel regulates MIP-1α expression in the context of multiple sclerosis (MS) remains largely unknown. In this study we identify the essential role of TRPC channel in IL-17-mediated MIP-1α expression and astrocyte activation. Moreover, treatment of astrocytes with IL-17 activated MAPKs and PI3K/Akt signaling pathways with downstream NF-κB pathways. Interestingly, the TRPC blocker-SKF96365 (10 μM) and Norgestimate (10 μM) significantly inhibited the increased expression of MIP-1α via suppression of IL-17-mediated ERK, p38 and JNK MAPKs and PI3K/Akt pathway activation, thereby underscoring the role of TRPC channel in this process. Together these data underpin the role of TRPC channel as a novel target that regulates MIP-1α expression and cell activation-mediated by IL-17 with implications for therapeutic intervention for reversal of neuroinflammation inflicted by IL-17. Understanding the regulation of MIP-1α expression may provide insights into the development of potential therapeutic targets for neuroinflammation associated with MS.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Rongrong Huang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yanhong Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Hongwei Yi
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.,State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Jie Chao
- Department of Physiology, Medical School of Southeast University, Nanjing, China
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China. .,Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|