1
|
Interferon-γ-Treated Mesenchymal Stem Cells Modulate the T Cell-Related Chemokines and Chemokine Receptors in an Animal Model of Experimental Autoimmune Encephalomyelitis. Drug Res (Stuttg) 2023; 73:213-223. [PMID: 36754055 DOI: 10.1055/a-1995-6365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) modulate immune responses, and their immunomodulatory potential can be enhanced using inflammatory cytokines. Here, the modulatory effects of IFN-γ-licensed MSCs on expression of T cell-related chemokines and chemokine receptors were evaluated using an experimental autoimmune encephalomyelitis (EAE) model. MATERIAL AND METHODS EAE was induced in 3 groups of C57bl/6 mice and then treated with PBS, MSCs and IFN-γ-treated MSCs. The EAE manifestations were registered daily and finally, the brain and spinal cords were isolated for histopathological and gene expression studies. RESULTS The clinical scores were lowered in MSCs and IFN-γ-licensed MSCs groups, however, mice treated with IFN-γ-licensed MSCs exhibited lower clinical scores than MSCs-treated mice. Leukocyte infiltration into the brain was reduced after treatment with MSCs or IFN-γ-licensed MSCs compared to untreated group (P<0.05 and P<0.01, respectively). In comparison with untreated EAE mice, treatment with MSCs reduced CCL20 expression (P<0.001) and decreased CXCR3 and CCR6 expression (P<0.02 and P<0.04, respectively). In comparison with untreated EAE mice, treatment with IFN-γ-licensed MSCs reduced CXCL10, CCL17 and CCL20 expression (P<0.05, P<0.05, and P<0.001, respectively) as well as decreased CXCR3 and CCR6 expression (P<0.002 and P<0.02, respectively), whilst promoting expression of CCL22 and its receptor CCR4 (P<0.0001 and P<0.02, respectively). In comparison with MSC-treated group, mice treated with IFN-γ-licensed MSCs exhibited lower CXCL10 and CCR6 expression (P<0.002 and P<0.01, respectively), whereas greater expression of CCL22 and CCR4 (P<0.0001 and P<0.01, respectively). CONCLUSION Priming the MSC with IFN-γ can be an efficient approach to enhance the immunomodulatory potential of MSCs.
Collapse
|
2
|
Bernal‐Chico A, Tepavcevic V, Manterola A, Utrilla C, Matute C, Mato S. Endocannabinoid signaling in brain diseases: Emerging relevance of glial cells. Glia 2023; 71:103-126. [PMID: 35353392 PMCID: PMC9790551 DOI: 10.1002/glia.24172] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
The discovery of cannabinoid receptors as the primary molecular targets of psychotropic cannabinoid Δ9 -tetrahydrocannabinol (Δ9 -THC) in late 1980s paved the way for investigations on the effects of cannabis-based therapeutics in brain pathology. Ever since, a wealth of results obtained from studies on human tissue samples and animal models have highlighted a promising therapeutic potential of cannabinoids and endocannabinoids in a variety of neurological disorders. However, clinical success has been limited and major questions concerning endocannabinoid signaling need to be satisfactorily addressed, particularly with regard to their role as modulators of glial cells in neurodegenerative diseases. Indeed, recent studies have brought into the limelight diverse, often unexpected functions of astrocytes, oligodendrocytes, and microglia in brain injury and disease, thus providing scientific basis for targeting glial cells to treat brain disorders. This Review summarizes the current knowledge on the molecular and cellular hallmarks of endocannabinoid signaling in glial cells and its clinical relevance in neurodegenerative and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Ana Bernal‐Chico
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Neuroimmunology UnitBiocruces BizkaiaBarakaldoSpain
| | | | - Andrea Manterola
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Present address:
Parque Científico y Tecnológico de GuipuzkoaViralgenSan SebastianSpain
| | | | - Carlos Matute
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Susana Mato
- Department of NeurosciencesUniversity of the Basque Country UPV/EHULeioaSpain,Achucarro Basque Center for NeuroscienceLeioaSpain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Neuroimmunology UnitBiocruces BizkaiaBarakaldoSpain
| |
Collapse
|
3
|
Neurovascular Unit-Derived Extracellular Vesicles: From Their Physiopathological Roles to Their Clinical Applications in Acute Brain Injuries. Biomedicines 2022; 10:biomedicines10092147. [PMID: 36140248 PMCID: PMC9495841 DOI: 10.3390/biomedicines10092147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) form a heterogeneous group of membrane-enclosed structures secreted by all cell types. EVs export encapsulated materials composed of proteins, lipids, and nucleic acids, making them a key mediator in cell–cell communication. In the context of the neurovascular unit (NVU), a tightly interacting multicellular brain complex, EVs play a role in intercellular communication and in maintaining NVU functionality. In addition, NVU-derived EVs can also impact peripheral tissues by crossing the blood–brain barrier (BBB) to reach the blood stream. As such, EVs have been shown to be involved in the physiopathology of numerous neurological diseases. The presence of NVU-released EVs in the systemic circulation offers an opportunity to discover new diagnostic and prognostic markers for those diseases. This review outlines the most recent studies reporting the role of NVU-derived EVs in physiological and pathological mechanisms of the NVU, focusing on neuroinflammation and neurodegenerative diseases. Then, the clinical application of EVs-containing molecules as biomarkers in acute brain injuries, such as stroke and traumatic brain injuries (TBI), is discussed.
Collapse
|
4
|
Wu X, Wan T, Gao X, Fu M, Duan Y, Shen X, Guo W. Microglia Pyroptosis: A Candidate Target for Neurological Diseases Treatment. Front Neurosci 2022; 16:922331. [PMID: 35937897 PMCID: PMC9354884 DOI: 10.3389/fnins.2022.922331] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
In addition to its profound implications in the fight against cancer, pyroptosis have important role in the regulation of neuronal injury. Microglia are not only central members of the immune regulation of the central nervous system (CNS), but are also involved in the development and homeostatic maintenance of the nervous system. Under various pathological overstimulation, microglia pyroptosis contributes to the massive release of intracellular inflammatory mediators leading to neuroinflammation and ultimately to neuronal damages. In addition, microglia pyroptosis lead to further neurological damage by decreasing the ability to cleanse harmful substances. The pathogenic roles of microglia in a variety of CNS diseases such as neurodegenerative diseases, stroke, multiple sclerosis and depression, and many other neurological disorders have been gradually unveiled. In the context of different neurological disorders, inhibition of microglia pyroptosis by targeting NOD-like receptor family pyrin domain containing (NLRP) 3, caspase-1 and gasdermins (GSDMs) by various chemical agents as well as natural products significantly improve the symptoms or outcome in animal models. This study will provide new ideas for immunomodulatory treatment of CNS diseases.
Collapse
Affiliation(s)
- Xian Wu
- The First Affiliated Hospital of Hunan College of Traditional Chinese Medicine, Hunan Province Directly Affiliated TCM Hospital, Zhuzhou, China
| | - Teng Wan
- Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaoyu Gao
- Hengyang Medical College, University of South China, Hengyang, China
| | - Mingyuan Fu
- Hengyang Medical College, University of South China, Hengyang, China
| | - Yunfeng Duan
- The First Affiliated Hospital of Hunan College of Traditional Chinese Medicine, Hunan Province Directly Affiliated TCM Hospital, Zhuzhou, China
| | - Xiangru Shen
- Hengyang Medical College, University of South China, Hengyang, China
- *Correspondence: Xiangru Shen
| | - Weiming Guo
- Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Weiming Guo
| |
Collapse
|
5
|
Joshi P, Bisht A, Joshi S, Semwal D, Nema NK, Dwivedi J, Sharma S. Ameliorating potential of curcumin and its analogue in central nervous system disorders and related conditions: A review of molecular pathways. Phytother Res 2022; 36:3143-3180. [PMID: 35790042 DOI: 10.1002/ptr.7522] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/26/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Curcumin, isolated from turmeric (Curcuma longa L.) is one of the broadly studied phytomolecule owing to its strong antioxidant and anti-inflammatory potential and has been considered a promising therapeutic candidate in a wide range of disorders. Considering, its low bioavailability, different curcumin analogs have been developed to afford desired pharmacokinetic profile and therapeutic outcome in varied pathological states. Several preclinical and clinical studies have indicated that curcumin ameliorates mitochondrial dysfunction, inflammation, oxidative stress apoptosis-mediated neural cell degeneration and could effectively be utilized in the treatment of different neurodegenerative diseases. Hence, in this review, we have summarized key findings of experimental and clinical studies conducted on curcumin and its analogues with special emphasis on molecular pathways, viz. NF-kB, Nrf2-ARE, glial activation, apoptosis, angiogenesis, SOCS/JAK/STAT, PI3K/Akt, ERK1/2 /MyD88 /p38 MAPK, JNK, iNOS/NO, and MMP pathways involved in imparting ameliorative effects in the therapy of neurodegenerative disorders and associated conditions.
Collapse
Affiliation(s)
- Priyanka Joshi
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.,R & D, Patanjali Ayurved Ltd, Patanjali Food and Herbal Park, Haridwar, Uttarakhand, India
| | - Akansha Bisht
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India
| | - Sushil Joshi
- R & D, Patanjali Ayurved Ltd, Patanjali Food and Herbal Park, Haridwar, Uttarakhand, India
| | - Deepak Semwal
- Faculty of Biomedical Sciences, Uttarakhand Ayurved University, Dehradun, Uttarakhand, India
| | - Neelesh Kumar Nema
- Paramount Kumkum Private Limited, Prestige Meridian-1, Bangalore, Karnataka, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India
| |
Collapse
|
6
|
Tiane A, Schepers M, Riemens R, Rombaut B, Vandormael P, Somers V, Prickaerts J, Hellings N, van den Hove D, Vanmierlo T. DNA methylation regulates the expression of the negative transcriptional regulators ID2 and ID4 during OPC differentiation. Cell Mol Life Sci 2021; 78:6631-6644. [PMID: 34482420 PMCID: PMC8558293 DOI: 10.1007/s00018-021-03927-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022]
Abstract
The differentiation of oligodendrocyte precursor cells (OPCs) into myelinating oligodendrocytes is the prerequisite for remyelination in demyelinated disorders such as multiple sclerosis (MS). Epigenetic mechanisms, such as DNA methylation, have been suggested to control the intricate network of transcription factors involved in OPC differentiation. Yet, the exact mechanism remains undisclosed. Here, we are the first to identify the DNA-binding protein inhibitors, Id2 and Id4, as targets of DNA methylation during OPC differentiation. Using state-of-the-art epigenetic editing via CRISPR/dCas9-DNMT3a, we confirm that targeted methylation of Id2/Id4 drives OPC differentiation. Moreover, we show that in the pathological context of MS, methylation and gene expression levels of both ID2 and ID4 are altered compared to control human brain samples. We conclude that DNA methylation is crucial to suppress ID2 and ID4 during OPC differentiation, a process that appears to be dysregulated during MS. Our data do not only reveal new insights into oligodendrocyte biology, but could also lead to a better understanding of CNS myelin disorders.
Collapse
Affiliation(s)
- Assia Tiane
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Renzo Riemens
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Institute of Human Genetics, Julius Maximilians University, Wuerzburg, Germany
| | - Ben Rombaut
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Patrick Vandormael
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Veerle Somers
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Niels Hellings
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Daniel van den Hove
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium.
| |
Collapse
|
7
|
Mado H, Adamczyk-Sowa M, Bartman W, Wierzbicki K, Tadeusiak B, Sowa P. Plasma Interleukin-33 level in relapsing-remitting multiple sclerosis. Is it negatively correlated with central nervous system lesions in patients with mild disability? Clin Neurol Neurosurg 2021; 206:106700. [PMID: 34030079 DOI: 10.1016/j.clineuro.2021.106700] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/06/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cytokines and chemokines are undoubtedly involved in the pathogenesis of multiple sclerosis (MS). There are many reports that suggest a significant role for Interleukin-33 (IL-33) in the course of MS development, but it is not clear whether negative or positive. We therefore investigated plasma IL-33 levels in patients with relapsing-remitting MS (RRMS). METHODS The study consisted of RRMS patients (n = 73) and healthy subjects (n = 54). Blood samples were taken from all and plasma IL-33 levels were then determined using an enzyme-linked immunosorbent assay method. Patients also underwent laboratory and imaging tests and their disability status was assessed. RESULTS Plasma IL-33 levels were marginally significantly higher in patients with RRMS (p = 0.07). Higher IL-33 levels are significantly associated with higher age (p = 0.01). There was also a statistically significant negative correlation between plasma IL-33 levels and the number of high signal intensity lesions in T2-weighted MRI (p = 0.03). After dividing the number of lesions into groups < 9 and ≥ 9 T2-weighted lesions, the Student's t-test for unrelated variables showed a negative correlation, but not statistically significant (p = 0.22), while the Spearman's correlation showed a marginally significant correlation (p = 0.06) between IL-33 level and number of T2-weighted lesions. IL-33 was also shown to have a significant ability to differentiate RRMS patients from healthy subjects with a sensitivity of 99% and specificity of 70% (p = 0.00). CONCLUSIONS Patients with RRMS have elevated plasma IL-33 levels. In RRMS patients with mild disability, high plasma levels of IL-33 may have neuroprotective effects potentially by stimulating remyelination and/or suppressing autoimmune inflammation and damage. Further studies on this matter on a larger number of patients are needed.
Collapse
Affiliation(s)
- Hubert Mado
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland.
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Wojciech Bartman
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Krzysztof Wierzbicki
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Bartosz Tadeusiak
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Paweł Sowa
- Department of Otorhinolaryngology and Oncological Laryngology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
8
|
Haase S, Linker RA. Inflammation in multiple sclerosis. Ther Adv Neurol Disord 2021; 14:17562864211007687. [PMID: 33948118 PMCID: PMC8053832 DOI: 10.1177/17562864211007687] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that is characterised pathologically by demyelination, gliosis, neuro-axonal damage and inflammation. Despite intense research, the underlying pathomechanisms driving inflammatory demyelination in MS still remain incompletely understood. It is thought to be caused by an autoimmune response towards CNS self-antigens in genetically susceptible individuals, assuming autoreactive T cells as disease-initiating immune cells. Yet, B cells were recognized as crucial immune cells in disease pathology, including antibody-dependent and independent effects. Moreover, myeloid cells are important contributors to MS pathology, and it is becoming increasingly evident that different cell types act in concert during MS immunopathology. This is supported by the finding that the beneficial effects of actual existing disease-modifying therapies cannot be attributed to one single immune cell-type, but rather involve immunological cooperation. The current strategy of MS therapies thus aims to shift the immune cell repertoire from a pro-inflammatory towards an anti-inflammatory phenotype, involving regulatory T and B cells and anti-inflammatory macrophages. Although no existing therapy actually exists that directly induces an enhanced regulatory immune cell pool, numerous studies identified potential net effects on these cell types. This review gives a conceptual overview on T cells, B cells and myeloid cells in the immunopathology of relapsing-remitting MS and discusses potential contributions of actual disease-modifying therapies on these immune cell phenotypes.
Collapse
Affiliation(s)
- Stefanie Haase
- Neuroimmunologie, Klinik und Poliklinik für Neurologie, Universitätsklinik Regensburg, Franz-Josef-Strauss Allee, Regensburg, 93053, Germany
| | - Ralf A Linker
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
9
|
Pantazou V, Roux T, Oliveira Moreira V, Lubetzki C, Desmazières A. Interaction between Neurons and the Oligodendroglial Lineage in Multiple Sclerosis and Its Preclinical Models. Life (Basel) 2021; 11:231. [PMID: 33799653 PMCID: PMC7999210 DOI: 10.3390/life11030231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/06/2021] [Accepted: 03/07/2021] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is a complex central nervous system inflammatory disease leading to demyelination and associated functional deficits. Though endogenous remyelination exists, it is only partial and, with time, patients can enter a progressive phase of the disease, with neurodegeneration as a hallmark. Though major therapeutic advances have been made, with immunotherapies reducing relapse rate during the inflammatory phase of MS, there is presently no therapy available which significantly impacts disease progression. Remyelination has been shown to favor neuroprotection, and it is thus of major importance to better understand remyelination mechanisms in order to promote them and hence preserve neurons. A crucial point is how this process is regulated through the neuronal crosstalk with the oligodendroglial lineage. In this review, we present the current knowledge on neuron interaction with the oligodendroglial lineage, in physiological context as well as in MS and its experimental models. We further discuss the therapeutic possibilities resulting from this research field, which might allow to support remyelination and neuroprotection and thus limit MS progression.
Collapse
Affiliation(s)
- Vasiliki Pantazou
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
- Service de Neurologie, Centre Hospitalier Universitaire Vaudois, 46 Rue du Bugnon, 1011 Lausanne, Switzerland
| | - Thomas Roux
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
- Assistance Publique-Hôpitaux de Paris, Neurology Department, Pitié Salpêtrière University Hospital, 75013 Paris, France
| | - Vanessa Oliveira Moreira
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
| | - Catherine Lubetzki
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
- Assistance Publique-Hôpitaux de Paris, Neurology Department, Pitié Salpêtrière University Hospital, 75013 Paris, France
| | - Anne Desmazières
- Paris Brain Institute (ICM), Sorbonne Université, CNRS, Inserm, GH Pitié-Salpêtrière, 47 boulevard de l’Hôpital, 75013 Paris, France; (V.P.); (T.R.); (V.O.M.); (C.L.)
| |
Collapse
|
10
|
Lu L, Qi S, Chen Y, Luo H, Huang S, Yu X, Luo Q, Zhang Z. Targeted immunomodulation of inflammatory monocytes across the blood-brain barrier by curcumin-loaded nanoparticles delays the progression of experimental autoimmune encephalomyelitis. Biomaterials 2020; 245:119987. [DOI: 10.1016/j.biomaterials.2020.119987] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/08/2023]
|
11
|
Effects of EHP-101 on inflammation and remyelination in murine models of Multiple sclerosis. Neurobiol Dis 2020; 143:104994. [PMID: 32599064 DOI: 10.1016/j.nbd.2020.104994] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/05/2020] [Accepted: 06/20/2020] [Indexed: 01/26/2023] Open
Abstract
Multiple Sclerosis (MS) is characterized by a combination of inflammatory and neurodegenerative processes in the spinal cord and the brain. Natural and synthetic cannabinoids such as VCE-004.8 have been studied in preclinical models of MS and represent promising candidates for drug development. VCE-004.8 is a multitarget synthetic cannabidiol (CBD) derivative acting as a dual Peroxisome proliferator-activated receptor-gamma/Cannabinoid receptor type 2 (PPARγ/CB2) ligand agonist that also activates the Hypoxia-inducible factor (HIF) pathway. EHP-101 is an oral lipidic formulation of VCE-004.8 that has shown efficacy in several preclinical models of autoimmune, inflammatory, fibrotic, and neurodegenerative diseases. EHP-101 alleviated clinical symptomatology in EAE and transcriptomic analysis demonstrated that EHP-101 prevented the expression of many inflammatory genes closely associated with MS pathophysiology in the spinal cord. EHP-101 normalized the expression of several genes associated with oligodendrocyte function such as Teneurin 4 (Tenm4) and Gap junction gamma-3 (Gjc3) that were downregulated in EAE. EHP-101 treatment prevented microglia activation and demyelination in both the spinal cord and the brain. Moreover, EAE was associated with a loss in the expression of Oligodendrocyte transcription factor 2 (Olig2) in the corpus callosum, a marker for oligodendrocyte differentiation, which was restored by EHP-101 treatment. In addition, EHP-101 enhanced the expression of glutathione S-transferase pi (GSTpi), a marker for mature oligodendrocytes in the brain. We also found that a diet containing 0.2% cuprizone for six weeks induced a clear loss of myelin in the brain measured by Cryomyelin staining and Myelin basic protein (MBP) expression. Moreover, EHP-101 also prevented cuprizone-induced microglial activation, astrogliosis and reduced axonal damage. Our results provide evidence that EHP-101 showed potent anti-inflammatory activity, prevented demyelination, and enhanced remyelination. Therefore, EHP-101 represents a promising drug candidate for the potential treatment of different forms of MS.
Collapse
|
12
|
Malpas CB, Manouchehrinia A, Sharmin S, Roos I, Horakova D, Havrdova EK, Trojano M, Izquierdo G, Eichau S, Bergamaschi R, Sola P, Ferraro D, Lugaresi A, Prat A, Girard M, Duquette P, Grammond P, Grand’Maison F, Ozakbas S, Van Pesch V, Granella F, Hupperts R, Pucci E, Boz C, Sidhom Y, Gouider R, Spitaleri D, Soysal A, Petersen T, Verheul F, Karabudak R, Turkoglu R, Ramo-Tello C, Terzi M, Cristiano E, Slee M, McCombe P, Macdonell R, Fragoso Y, Olascoaga J, Altintas A, Olsson T, Butzkueven H, Hillert J, Kalincik T. Early clinical markers of aggressive multiple sclerosis. Brain 2020; 143:1400-1413. [DOI: 10.1093/brain/awaa081] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/21/2020] [Accepted: 02/05/2020] [Indexed: 11/13/2022] Open
Abstract
Abstract
Patients with the ‘aggressive’ form of multiple sclerosis accrue disability at an accelerated rate, typically reaching Expanded Disability Status Score (EDSS) ≥ 6 within 10 years of symptom onset. Several clinicodemographic factors have been associated with aggressive multiple sclerosis, but less research has focused on clinical markers that are present in the first year of disease. The development of early predictive models of aggressive multiple sclerosis is essential to optimize treatment in this multiple sclerosis subtype. We evaluated whether patients who will develop aggressive multiple sclerosis can be identified based on early clinical markers. We then replicated this analysis in an independent cohort. Patient data were obtained from the MSBase observational study. Inclusion criteria were (i) first recorded disability score (EDSS) within 12 months of symptom onset; (ii) at least two recorded EDSS scores; and (iii) at least 10 years of observation time, based on time of last recorded EDSS score. Patients were classified as having ‘aggressive multiple sclerosis’ if all of the following criteria were met: (i) EDSS ≥ 6 reached within 10 years of symptom onset; (ii) EDSS ≥ 6 confirmed and sustained over ≥6 months; and (iii) EDSS ≥ 6 sustained until the end of follow-up. Clinical predictors included patient variables (sex, age at onset, baseline EDSS, disease duration at first visit) and recorded relapses in the first 12 months since disease onset (count, pyramidal signs, bowel-bladder symptoms, cerebellar signs, incomplete relapse recovery, steroid administration, hospitalization). Predictors were evaluated using Bayesian model averaging. Independent validation was performed using data from the Swedish Multiple Sclerosis Registry. Of the 2403 patients identified, 145 were classified as having aggressive multiple sclerosis (6%). Bayesian model averaging identified three statistical predictors: age > 35 at symptom onset, EDSS ≥ 3 in the first year, and the presence of pyramidal signs in the first year. This model significantly predicted aggressive multiple sclerosis [area under the curve (AUC) = 0.80, 95% confidence intervals (CIs): 0.75, 0.84, positive predictive value = 0.15, negative predictive value = 0.98]. The presence of all three signs was strongly predictive, with 32% of such patients meeting aggressive disease criteria. The absence of all three signs was associated with a 1.4% risk. Of the 556 eligible patients in the Swedish Multiple Sclerosis Registry cohort, 34 (6%) met criteria for aggressive multiple sclerosis. The combination of all three signs was also predictive in this cohort (AUC = 0.75, 95% CIs: 0.66, 0.84, positive predictive value = 0.15, negative predictive value = 0.97). Taken together, these findings suggest that older age at symptom onset, greater disability during the first year, and pyramidal signs in the first year are early indicators of aggressive multiple sclerosis.
Collapse
Affiliation(s)
- Charles B Malpas
- CORe Unit, Department of Medicine, University of Melbourne, Melbourne, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Australia
| | - Ali Manouchehrinia
- Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sifat Sharmin
- CORe Unit, Department of Medicine, University of Melbourne, Melbourne, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Australia
| | - Izanne Roos
- CORe Unit, Department of Medicine, University of Melbourne, Melbourne, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Australia
| | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Eva Kubala Havrdova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Maria Trojano
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | | | - Sara Eichau
- Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | - Patrizia Sola
- Department of Neuroscience, Azienda Ospedaliera Universitaria, Modena, Italy
| | - Diana Ferraro
- Department of Neuroscience, Azienda Ospedaliera Universitaria, Modena, Italy
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Lugaresi
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | | - Marc Girard
- CHUM and Universite de Montreal, Montreal, Canada
| | | | | | | | | | - Vincent Van Pesch
- Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Université Catholique de Louvain, Brussels, Belgium
| | - Franco Granella
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Eugenio Pucci
- UOC Neurologia, Azienda Sanitaria Unica Regionale Marche - AV3, Macerata, Italy
| | - Cavit Boz
- KTU Medical Faculty Farabi Hospital, Trabzon, Turkey
| | - Youssef Sidhom
- Department of Neurology, Razi Hospital, Manouba, Tunisia
| | - Riadh Gouider
- Department of Neurology, Razi Hospital, LR 18SP03, Clinical Investigation Center Neurosciences and Mental Health, Faculty of Medicine University Tunis El Manar, Tunis, Tunisia
| | - Daniele Spitaleri
- Azienda Ospedaliera di Rilievo Nazionale San Giuseppe Moscati Avellino, Avellino, Italy
| | - Aysun Soysal
- Bakirkoy Education and Research Hospital for Psychiatric and Neurological Diseases, Istanbul, Turkey
| | | | | | | | - Recai Turkoglu
- Haydarpasa Numune Training and Research Hospital, Istanbul, Turkey
| | | | - Murat Terzi
- Medical Faculty, 19 Mayis University, Samsun, Turkey
| | | | - Mark Slee
- Flinders University, Adelaide, Australia
| | - Pamela McCombe
- University of Queensland, Brisbane, Australia
- Royal Brisbane and Women’s Hospital, Brisbane, Australia
| | | | - Yara Fragoso
- Universidade Metropolitana de Santos, Santos, Brazil
| | - Javier Olascoaga
- Instituto de Investigación Sanitaria Biodonostia, Hospital Universitario Donostia, San Sebastián, Spain
| | - Ayse Altintas
- Koc University, School of Medicine, Department of Neurology, Istanbul, Turkey
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Sweden
| | - Helmut Butzkueven
- Central Clinical School, Monash University, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, Australia
- Department of Neurology, Box Hill Hospital, Monash University, Melbourne, Australia
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Sweden
| | - Tomas Kalincik
- CORe Unit, Department of Medicine, University of Melbourne, Melbourne, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
13
|
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease that affects the central nervous system (CNS), particularly, in young adults. Current MS treatments aim to reduce demyelination; however, these have limited efficacy, display side effects and lack of regenerative activities. Oligodendrocyte progenitor cells (OPCs) represents the major source for new myelin. Upon demyelination, OPCs get activated, proliferate, migrate towards the lesion, and differentiate into remyelinating oligodendrocytes. Although myelin repair (remyelination) represents a robust response to myelin damage, during MS, this regenerative phenomenon decays in efficiency or even fails. CNS-resident pericytes (CNS-PCs) are essential for vascular homeostasis regulating blood-brain barrier (BBB) permeability and stability as well as endothelial cells (ECs) function during angiogenesis and neovascularization. Recent studies indicate that CNS-PCs also play a crucial role regulating OPC function during remyelination, and very importantly, these cells are substantially affected in MS. This chapter summarizes important aspects of MS and CNS remyelination as well as it provides new insights supporting the contribution of CNS-PCs to myelin regeneration and to MS pathology. Currently, there is evidence arguing in favor of CNS-PCs as novel therapeutic targets for the development of future treatments for MS.
Collapse
|
14
|
Safaeinejad F, Bahrami S, Redl H, Niknejad H. Inhibition of Inflammation, Suppression of Matrix Metalloproteinases, Induction of Neurogenesis, and Antioxidant Property Make Bryostatin-1 a Therapeutic Choice for Multiple Sclerosis. Front Pharmacol 2018; 9:625. [PMID: 29971003 PMCID: PMC6018466 DOI: 10.3389/fphar.2018.00625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/24/2018] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease characterized by inflammation and myelin damage. Pro-inflammatory cytokines, oxidative stress, high level of matrix metalloproteinases (MMPs) activity and blood-brain barrier (BBB) damage, immune-mediated destruction of myelin and neuron loss are involved in the pathogenesis of MS. The currently approved treatments for MS include injectable drugs (interferon-β and glatiramer acetate), oral drugs (fingolimod), and monoclonal antibodies (natalizumab). The mentioned therapeutic choices are mostly focused on the inhibition of inflammation. Therefore, the search for a multi-target therapeutic choice remains unchallenged. It seems that a drug with anti-inflammatory, oxidative stress inhibitory, reduction of MMPs activity, and neurogenesis stimulatory properties may be effective for treatment of MS. In this regard, Bryostatin-1 as a macrolide and marine natural product has been selected as a therapeutic choice. Studies indicate that Bryostatin-1 has anti-inflammatory and antioxidant properties and decreases MMPs level and BBB damage. Furthermore, Bryostatin-1 has a neuroprotective effect and promotes neurogenesis and differentiation of oligodendrocyte progenitor stem cells as a critical step for remyelination/myelogenesis. Based on these properties, we hypothesized here that Bryostatin-1 is an effective treatment in MS.
Collapse
Affiliation(s)
- Fahimeh Safaeinejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Caspase-1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis. Proc Natl Acad Sci U S A 2018; 115:E6065-E6074. [PMID: 29895691 DOI: 10.1073/pnas.1722041115] [Citation(s) in RCA: 352] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease of the CNS of unknown cause that remains incurable. Inflammasome-associated caspases mediate the maturation and release of the proinflammatory cytokines IL-1β and IL-18 and activate the pore-forming protein gasdermin D (GSDMD). Inflammatory programmed cell death, pyroptosis, was recently shown to be mediated by GSDMD. Here, we report molecular evidence for GSDMD-mediated inflammasome activation and pyroptosis in both myeloid cells (macrophages/microglia) and, unexpectedly, in myelin-forming oligodendrocytes (ODCs) in the CNS of patients with MS and in the MS animal model, experimental autoimmune encephalomyelitis (EAE). We observed inflammasome activation and pyroptosis in human microglia and ODCs in vitro after exposure to inflammatory stimuli and demonstrate caspase-1 inhibition by the small-molecule inhibitor VX-765 in both cell types. GSDMD inhibition by siRNA transduction suppressed pyroptosis in human microglia. VX-765 treatment of EAE animals reduced the expression of inflammasome- and pyroptosis-associated proteins in the CNS, prevented axonal injury, and improved neurobehavioral performance. Thus, GSDMD-mediated pyroptosis in select glia cells is a previously unrecognized mechanism of inflammatory demyelination and represents a unique therapeutic opportunity for mitigating the disease process in MS and other CNS inflammatory diseases.
Collapse
|
16
|
Chrobok NL, Bol JGJM, Jongenelen CA, Brevé JJP, El Alaoui S, Wilhelmus MMM, Drukarch B, van Dam AM. Characterization of Transglutaminase 2 activity inhibitors in monocytes in vitro and their effect in a mouse model for multiple sclerosis. PLoS One 2018; 13:e0196433. [PMID: 29689097 PMCID: PMC5918173 DOI: 10.1371/journal.pone.0196433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/12/2018] [Indexed: 02/07/2023] Open
Abstract
The neurodegenerative disease multiple sclerosis (MS) is pathologically characterized by the massive influx of immune cells into the central nervous system. This contributes to demyelination and axonal damage which causes symptoms such as motor and cognitive dysfunctions. The migration of leukocytes from the blood vessel is orchestrated by a multitude of factors whose determination is essential in reducing cellular influx in MS patients and the experimental autoimmune encephalomyelitis (EAE) animal model. The here studied enzyme tissue Transglutaminase (TG2) is present intracellularly, on the cell surface and extracellularly. There it contributes to cellular adhesion and migration via its transamidation activity and possibly by facilitating cellular interaction with the extracellular matrix. Previous data from our group showed reduced motor symptoms and cellular infiltration after using a pharmacological TG2 transamidation activity inhibitor in a rat EAE model. However, it remained elusive if the cross-linking activity of the enzyme resulted in the observed effects. To follow-up, we now characterized two new small molecule TG2 activity inhibitors, BJJF078 and ERW1041E. Both compounds are potent inhibitor of recombinant human and mouse Transglutaminase enzyme activity, mainly TG2 and the close related enzyme TG1. In addition they did not affect the binding of TG2 to the extracellular matrix substrate fibronectin, a process via which TG2 promotes cellular adhesion and migration. We found, that ERW1041E but not BJJF078 resulted in reduced EAE disease motor-symptoms while neither caused apparent changes in pathology (cellular influx), Transglutaminase activity or expression of inflammation related markers in the spinal cord, compared to vehicle treated controls. Although we cannot exclude issues on bioavailability and in vivo efficacy of the used compounds, we hypothesize that extracellular TG1/TG2 activity is of greater importance than (intra-)cellular activity in mouse EAE pathology.
Collapse
Affiliation(s)
- Navina L. Chrobok
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - John G. J. M. Bol
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Cornelis A. Jongenelen
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - John J. P. Brevé
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Micha M. M. Wilhelmus
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Benjamin Drukarch
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Critical Roles of IL-33/ST2 Pathway in Neurological Disorders. Mediators Inflamm 2018; 2018:5346413. [PMID: 29507527 PMCID: PMC5817350 DOI: 10.1155/2018/5346413] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/06/2017] [Indexed: 01/21/2023] Open
Abstract
Interleukin-33 (IL-33) is an IL-1 family member, which exhibits both pro- and anti-inflammatory properties solely based on the type of the disease itself. Generally, IL-33 is expressed by both endothelial and epithelial cells and mediates its function based on the interaction with various receptors, mainly with ST2 variants. IL-33 is a potent inducer for the Th2 immune response which includes defence mechanism in brain diseases. Thus, in this paper, we review the biological features of IL-33 and the critical roles of IL-33/ST2 pathway in selected neurological disorders including Alzheimer's disease, multiple sclerosis, and malaria infection to discuss the involvement of IL-33/ST2 pathway during these brain diseases and its potential as future immunotherapeutic agents or for intervention purposes.
Collapse
|
18
|
Quantitative analysis of lipid debris accumulation caused by cuprizone induced myelin degradation in different CNS areas. Brain Res Bull 2018; 137:277-284. [PMID: 29325992 DOI: 10.1016/j.brainresbull.2018.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/22/2017] [Accepted: 01/04/2018] [Indexed: 01/09/2023]
Abstract
Degradation of myelin sheath is thought to be the cause of neurodegenerative diseases, such as multiple sclerosis (MS), but definitive agreement on the mechanism of how myelin is lost is currently lacking. Autoimmune initiation of MS has been recently questioned by proposing that the immune response is a consequence of oligodendrocyte degeneration. To study the process of myelin breakdown, we induced demyelination with cuprizone and applied coherent anti-Stokes Raman scattering (CARS) microscopy, a non-destructive label-free method to image lipid structures in living tissue. We confirmed earlier results showing a brain region dependent myelin destructive effect of cuprizone. In addition, high resolution in situ CARS imaging revealed myelin debris forming lipid droplets alongwith myelinated axon fibers. Quantification of lipid debris with custom-made software for segmentation and three dimensional reconstruction revealed brain region dependent accumulation of lipid drops inversely correlated with the thickness of myelin sheaths. Finally, we confirmed that in situ CARS imaging is applicable to living human brain tissue in brain slices derived from a patient. Thus, CARS microscopy is potent tool for quantitative monitoring of myelin degradation in unprecedented spatiotemporal resolution during oligodendrocyte damage. We think that the accumulation of lipid drops around degrading myelin might be instrumental in triggering subsequent inflammatory processes.
Collapse
|
19
|
Zoghi A, Petramfar P, Nikseresht A, Sakhaee E. Investigation of ischemic and demyelinating lesions by cerebral vasoreactivity based on transcranial Doppler sonography: a comparative study. Neuropsychiatr Dis Treat 2018; 14:2323-2328. [PMID: 30254443 PMCID: PMC6141114 DOI: 10.2147/ndt.s150062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Variations of cerebral blood flow in response to hypoxia and hyperoxia in different disease conditions can provide new insights into disease etiopathogenesis. This study aimed to determine the characteristics of cerebral vasoreactivity for ischemia and demyelination. MATERIALS AND METHODS This case-control study included: 28 patients with lacunar infarctions verified by history, physical examination, and MRI; 28 age- and sex-matched healthy controls; 28 patients with relapsing-remitting multiple sclerosis (MS), based on McDonald criteria; and 28 age- and sex-matched healthy controls for the MS group. Transcranial Doppler sonography was undertaken in all subjects to calculate the mean flow velocity (MFV) of the right middle cerebral artery (MCA) and, after a breath-holding (BH) maneuver, the breath-holding index (BHI) was determined. RESULTS There was no significant difference of BHI and changes of MFV of the MCA in MS patients compared to controls (1.02 ± 0.4 vs 1.02 ± 0.3, p = 0.993; and 16.8 ± 8.1 vs 11.3 ± 10.8, p = 0.057). BHI in patients with lacunar infarctions was significantly lower (0.8 ± 0.4 vs 1.2 ± 0.3, p < 0.001) compared to controls. The BHI (p = 0.040) and variations of MFV of MCA (p = 0.007) in MS patients were significantly higher than in patients with lacunar infarctions. The vasoreactivity of demyelinating lesions was higher than that of ischemic ones. CONCLUSION Therefore, cerebral vasoreactivity determined by transcranial Doppler could be utilized for differentiating demyelinating from ischemic lesions.
Collapse
Affiliation(s)
- Anahita Zoghi
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Peyman Petramfar
- Clinical Neurology Research Center, Department of Neurology, Shiraz University of Medical Sciences, Shiraz, Iran,
| | - Alireza Nikseresht
- Clinical Neurology Research Center, Department of Neurology, Shiraz University of Medical Sciences, Shiraz, Iran,
| | - Ehsan Sakhaee
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Alsahebfosoul F, Rahimmanesh I, Shajarian M, Etemadifar M, Sedaghat N, Hejazi Z, Naderi S. Interleukin-33 plasma levels in patients with relapsing-remitting multiple sclerosis. Biomol Concepts 2017; 8:55-60. [PMID: 28107165 DOI: 10.1515/bmc-2016-0026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/25/2016] [Indexed: 02/08/2023] Open
Abstract
Cytokines are implicated in the immunopathogenesis of multiple sclerosis (MS). Interleukin (IL)-33, one of the recently discovered members of the IL-1 superfamily, is a dual functional cytokine involved in various autoimmune disorders. In a case-control study, venous blood was collected from healthy subjects categorized as control group (n=44) and MS patients (n=44). All recruited patients were clinically diagnosed with relapsing-remitting MS (RRMS), including patients without treatment (new identified cases, n=16) and those treated with interferon beta (IFN-β) (n=28). The plasma levels of IL-33 in subjects were measured with ELISA. Significantly elevated IL-33 plasma levels were observed in RRMS patients (p=0.005). Furthermore, IFN-β-treated MS patients had lower levels of IL-33 compared to the untreated patients (p<0.001). Increased IL-33 plasma levels in the patient group might be associated with development of MS. These results could contribute to our better understanding about the role of IL-33 in the immunopathogenesis of MS.
Collapse
Affiliation(s)
- Fereshteh Alsahebfosoul
- Isfahan Research Center of Multiple Sclerosis, Isfahan, Iran.,Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Isfahan Research Center of Multiple Sclerosis, Isfahan, Iran.,Departments of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Shajarian
- Isfahan Research Center of Multiple Sclerosis, Isfahan, Iran.,Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoud Etemadifar
- Isfahan Research Center of Multiple Sclerosis, Isfahan, Iran.,Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Sedaghat
- Isfahan Research Center of Multiple Sclerosis, Isfahan, Iran.,Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Hejazi
- Isfahan Research Center of Multiple Sclerosis, Isfahan, Iran.,Departments of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shamsi Naderi
- Isfahan Research Center of Multiple Sclerosis, Isfahan, Iran.,Departments of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
21
|
Menezes KM, Algarve TD, Flôres FS, Cruz IBM, Copetti F, Silveira AF. DNA damage and postural balance in multiple sclerosis patients. FISIOTERAPIA EM MOVIMENTO 2017. [DOI: 10.1590/1980-5918.030.s01.ao08] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Abstract Introduction: Multiple Sclerosis (MS) is a chronic inflammatory disease characterized by infiltration of inflammatory cells on the Central Nervous System (CNS). There is evidence that cumulative DNA damage can contribute to various mechanisms underlying MS lesions. Changes in postural balance are frequent observations in subjects with MS. Objective: Evaluated the DNA damage index (DDI)) and postural balance in patients with MS. Methods: A case-control study was conducted with 28 subjects matched for sex, age, and body mass index, divided into MS group and control. The DDI was assessed by comet assay and postural balance through recording the body oscillations of the center of pressure (COP), in the anterior-posterior and lateral middle directions. Results: Showed higher DDI in MS patients (21.3 ± 4.8) than controls (7.9 ± 6.1). Significant differences between groups were also noted in postural control parameters. The wider ranges of postural sway were observed in the MS group. The associations between DDI and postural control parameters showed weak, but significant correlations. No associations were found between DDI and time of diagnosis of MS. Conclusion: People with MS had higher DDI and larger body oscillations than healthy individuals.
Collapse
|
22
|
Akkermann R, Beyer F, Küry P. Heterogeneous populations of neural stem cells contribute to myelin repair. Neural Regen Res 2017; 12:509-517. [PMID: 28553319 PMCID: PMC5436337 DOI: 10.4103/1673-5374.204999] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
As ingenious as nature's invention of myelin sheaths within the mammalian nervous system is, as fatal can be damage to this specialized lipid structure. Long-term loss of electrical insulation and of further supportive functions myelin provides to axons, as seen in demyelinating diseases such as multiple sclerosis (MS), leads to neurodegeneration and results in progressive disabilities. Multiple lines of evidence have demonstrated the increasing inability of oligodendrocyte precursor cells (OPCs) to replace lost oligodendrocytes (OLs) in order to restore lost myelin. Much research has been dedicated to reveal potential reasons for this regeneration deficit but despite promising approaches no remyelination-promoting drugs have successfully been developed yet. In addition to OPCs neural stem cells of the adult central nervous system also hold a high potential to generate myelinating OLs. There are at least two neural stem cell niches in the brain, the subventricular zone lining the lateral ventricles and the subgranular zone of the dentate gyrus, and an additional source of neural stem cells has been located in the central canal of the spinal cord. While a substantial body of literature has described their neurogenic capacity, still little is known about the oligodendrogenic potential of these cells, even if some animal studies have provided proof of their contribution to remyelination. In this review, we summarize and discuss these studies, taking into account the different niches, the heterogeneity within and between stem cell niches and present current strategies of how to promote stem cell-mediated myelin repair.
Collapse
Affiliation(s)
- Rainer Akkermann
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Felix Beyer
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patrick Küry
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
23
|
Akkermann R, Jadasz JJ, Azim K, Küry P. Taking Advantage of Nature's Gift: Can Endogenous Neural Stem Cells Improve Myelin Regeneration? Int J Mol Sci 2016; 17:ijms17111895. [PMID: 27854261 PMCID: PMC5133894 DOI: 10.3390/ijms17111895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/28/2016] [Accepted: 11/09/2016] [Indexed: 01/18/2023] Open
Abstract
Irreversible functional deficits in multiple sclerosis (MS) are directly correlated to axonal damage and loss. Neurodegeneration results from immune-mediated destruction of myelin sheaths and subsequent axonal demyelination. Importantly, oligodendrocytes, the myelinating glial cells of the central nervous system, can be replaced to some extent to generate new myelin sheaths. This endogenous regeneration capacity has so far mainly been attributed to the activation and recruitment of resident oligodendroglial precursor cells. As this self-repair process is limited and increasingly fails while MS progresses, much interest has evolved regarding the development of remyelination-promoting strategies and the presence of alternative cell types, which can also contribute to the restoration of myelin sheaths. The adult brain comprises at least two neurogenic niches harboring life-long adult neural stem cells (NSCs). An increasing number of investigations are beginning to shed light on these cells under pathological conditions and revealed a significant potential of NSCs to contribute to myelin repair activities. In this review, these emerging investigations are discussed with respect to the importance of stimulating endogenous repair mechanisms from germinal sources. Moreover, we present key findings of NSC-derived oligodendroglial progeny, including a comprehensive overview of factors and mechanisms involved in this process.
Collapse
Affiliation(s)
- Rainer Akkermann
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| | - Janusz Joachim Jadasz
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| | - Kasum Azim
- Focus Translational Neuroscience, Institute of Physiological Chemistry, University of Mainz, 55122 Mainz, Germany.
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| |
Collapse
|
24
|
Monocyte behaviour and tissue transglutaminase expression during experimental autoimmune encephalomyelitis in transgenic CX3CR1 gfp/gfp mice. Amino Acids 2016; 49:643-658. [PMID: 27826792 PMCID: PMC5332504 DOI: 10.1007/s00726-016-2359-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/01/2016] [Indexed: 02/06/2023]
Abstract
Leukocyte infiltration into the central nervous system (CNS) is a key pathological feature in multiple sclerosis (MS) and the MS animal model experimental autoimmune encephalomyelitis (EAE). Recently, preventing leukocyte influx into the CNS of MS patients is the main target of MS therapies and insight into cell behaviour in the circulation is needed for further elucidation of such therapies. In this study, we aimed at in vivo visualization of monocytes in a time-dependent manner during EAE. Using intravital two-photon microscopy (IVM), we imaged CX3CR1gfp/gfp mice during EAE, visualizing CX3CR1-GFP+ monocytes and their dynamics in the spinal cord vasculature. Our observations showed that intraluminal crawling of CX3CR1-GFP+ monocytes increased even before the clinical onset of EAE due to immunization of the animals. Furthermore, intraluminal crawling remained elevated during ongoing clinical disease. Besides, the displacement of these cells was larger during the peak of EAE compared to the control animals. In addition, we showed that the enzyme tissue transglutaminase (TG2), which is present in CNS-infiltrated cells in MS patients, is likewise found in CX3CR1-GFP+ monocytes in the spinal cord lesions and at the luminal side of the vasculature during EAE. It might thereby contribute to adhesion and crawling of monocytes, facilitating extravasation into the CNS. Thus, we put forward that interference with monocyte adhesion, by e.g. inhibition of TG2, should be applied at a very early stage of EAE and possibly MS, to effectively combat subsequent pathology.
Collapse
|
25
|
Borghi M, Carletto S, Ostacoli L, Scavelli F, Pia L, Pagani M, Bertolotto A, Malucchi S, Signori A, Cavallo M. Decline of Neuropsychological Abilities in a Large Sample of Patients with Multiple Sclerosis: A Two-Year Longitudinal Study. Front Hum Neurosci 2016; 10:282. [PMID: 27375468 PMCID: PMC4896920 DOI: 10.3389/fnhum.2016.00282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/25/2016] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE In this longitudinal study, we monitored two large groups of multiple sclerosis (MS) patients and healthy controls (HC) for 2 years, with the aim of comparing their neuropsychological profile over time. METHOD Three hundred and twenty-two patients with MS and 303 HC were administered the Brief Repeatable Battery of Neuropsychological tests (BRB-N); neuropsychiatric measures were also administered. Two follow-ups were scheduled at 1 and 2 years, respectively. RESULTS A linear mixed model (LMM) with random intercept was run by considering participants' performance on each test of the BRB-N at the three assessment points (baseline and follow-ups) as the within-subjects variable, and group (patients and controls) as the between-subjects factor. The interaction term was statistically significant for the tests: Symbol Digit Modalities test (SDMT) (p = 0.044), Paced Auditory Serial Addition test (PASAT) (p = 0.011) and Word List Generation (WLG) (p < 0.001), whereas for the PASAT-3 approached statistical significance (p = 0.05). In addition, a LMM with random intercept was also run by identifying three groups (controls, relapsing-remitting course of MS (i.e. RR-MS), and prog-MS). The interaction term was statistically significant for: PASAT-3 (p = 0.017), PASAT-2 (p = 0.0026), and WLG (p = 0.0022). CONCLUSIONS Our results corroborate on a very large scale evidence that the abilities tapped by the tasks SDMT, PASAT and WLG are particularly sensitive to MS, and further extend this issue by showing that these abilities are likely to be more sensitive than others to the progression of the disease, as compared to HC.
Collapse
Affiliation(s)
- Martina Borghi
- Clinical Psychology and Psychosomatics Service, University Hospital San Luigi Gonzaga, University of Turin Orbassano, Italy
| | - Sara Carletto
- Clinical Psychology and Psychosomatics Service, University Hospital San Luigi Gonzaga, University of Turin Orbassano, Italy
| | - Luca Ostacoli
- Clinical Psychology and Psychosomatics Service, University Hospital San Luigi Gonzaga, University of Turin Orbassano, Italy
| | - Francesco Scavelli
- Clinical Psychology and Psychosomatics Service, University Hospital San Luigi Gonzaga, University of Turin Orbassano, Italy
| | - Lorenzo Pia
- SAMBA (SpAtial, Motor and Bodily Awareness) Research Group, Department of Psychology, University of TurinTurin, Italy; Neuroscience Institute of Turin (NIT), University of TurinTurin, Italy
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, Consiglio Nazionale delle Ricerche (CNR), Rome Italy
| | - Antonio Bertolotto
- Neurologia 2 - CRESM (Regional Reference Centre for Multiple Sclerosis), "San Luigi Gonzaga" Hospital Medical School Orbassano, Italy
| | - Simona Malucchi
- Neurologia 2 - CRESM (Regional Reference Centre for Multiple Sclerosis), "San Luigi Gonzaga" Hospital Medical School Orbassano, Italy
| | - Alessio Signori
- Department of Health Sciences (DISSAL), Section of Biostatistics, University of Genoa Genoa, Italy
| | - Marco Cavallo
- Faculty of Psychology, eCampus UniversityNovedrate, Italy; Department of Mental Health, Azienda Sanitaria Locale Torino 3Collegno, Italy
| |
Collapse
|
26
|
Tekgöl Uzuner G, Uzuner N. Neurovascular coupling in patients with relapsing-remitting multiple sclerosis. Clin Neurol Neurosurg 2016; 146:24-8. [PMID: 27136094 DOI: 10.1016/j.clineuro.2016.04.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 03/16/2016] [Accepted: 04/25/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Also to the inflammatory demyelinating lesions and degenerative process, altered cerebrovascular reactivity or neurovascular coupling (NVC) might be considered as playing another role in the pathogenesis of multiple sclerosis. The objective of this study is to assess the NVC of patients with relapsing-remitting multiple sclerosis (RRMS) during the acute exacerbation period. PATIENTS AND METHODS Four hundred fifty-eight patients with RRMS and 160 healthy subjects were screened for this study during the last 14 years. We performed transtemporal transcranial Doppler recordings from the P2-segments of both posterior cerebral arteries simultaneously during simple or complex visual stimulation. The NVC was defined as a relative increase of the blood flow velocities as a percentage change of the baseline values during visual stimulation. RESULTS The NVC to simple visual stimulation was significantly higher in the patients on both sides (37.2±13.5% and 36.0±14.8%; right and left side, respectively) from those of the controls (30.9±9.9% and 30.0±8.8%; right and left side, respectively) (p<0.01). Similarly, the NVC to complex visual stimulation was significantly higher in the patients (43.3±14.1% and 41.7±13.5%; right and left side, respectively) from those of the controls (38.6±14.2% and 37.6±14.1%; right and left side, respectively) (p<0.05). CONCLUSION Our results suggest that patients with RRMS during exacerbation period have more reactive neurovascular units in the occipital cortex.
Collapse
Affiliation(s)
- Gulnur Tekgöl Uzuner
- Eskisehir Osmangazi University, Faculty of Medicine, Department of Neurology, Eskisehir, Turkey.
| | - Nevzat Uzuner
- Eskisehir Osmangazi University, Faculty of Medicine, Department of Neurology, Eskisehir, Turkey.
| |
Collapse
|
27
|
Malan-Müller S, Fairbairn L, Daniels WMU, Dashti MJS, Oakeley EJ, Altorfer M, Kidd M, Seedat S, Gamieldien J, Hemmings SMJ. Molecular mechanisms of D-cycloserine in facilitating fear extinction: insights from RNAseq. Metab Brain Dis 2016; 31:135-56. [PMID: 26400817 DOI: 10.1007/s11011-015-9727-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/02/2015] [Indexed: 01/24/2023]
Abstract
D-cycloserine (DCS) has been shown to be effective in facilitating fear extinction in animal and human studies, however the precise mechanisms whereby the co-administration of DCS and behavioural fear extinction reduce fear are still unclear. This study investigated the molecular mechanisms of intrahippocampally administered D-cycloserine in facilitating fear extinction in a contextual fear conditioning animal model. Male Sprague Dawley rats (n = 120) were grouped into four experimental groups (n = 30) based on fear conditioning and intrahippocampal administration of either DCS or saline. The light/dark avoidance test was used to differentiate maladapted (MA) (anxious) from well-adapted (WA) (not anxious) subgroups. RNA extracted from the left dorsal hippocampus was used for RNA sequencing and gene expression data was compared between six fear-conditioned + saline MA (FEAR + SALINE MA) and six fear-conditioned + DCS WA (FEAR + DCS WA) animals. Of the 424 significantly downregulated and 25 significantly upregulated genes identified in the FEAR + DCS WA group compared to the FEAR + SALINE MA group, 121 downregulated and nine upregulated genes were predicted to be relevant to fear conditioning and anxiety and stress-related disorders. The majority of downregulated genes transcribed immune, proinflammatory and oxidative stress systems molecules. These molecules mediate neuroinflammation and cause neuronal damage. DCS also regulated genes involved in learning and memory processes, and genes associated with anxiety, stress-related disorders and co-occurring diseases (e.g., cardiovascular diseases, digestive system diseases and nervous system diseases). Identifying the molecular underpinnings of DCS-mediated fear extinction brings us closer to understanding the process of fear extinction.
Collapse
Affiliation(s)
- Stefanie Malan-Müller
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| | - Lorren Fairbairn
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Willie M U Daniels
- Department of Human Physiology, University of KwaZulu-Natal, Durban, South Africa
| | | | - Edward J Oakeley
- Novartis Institutes for BioMedical Research, Biomarker Development - Human Genetics and Genomics, Genome Technologies, Basel, Switzerland
| | - Marc Altorfer
- Novartis Institutes for BioMedical Research, Biomarker Development - Human Genetics and Genomics, Genome Technologies, Basel, Switzerland
| | - Martin Kidd
- Centre for Statistical Consultation, Stellenbosch University, Stellenbosch, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Junaid Gamieldien
- University of the Western Cape, South African National Bioinformatics Institute, Cape Town, South Africa
| | - Sîan Megan Joanna Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
28
|
Abstract
Multiple sclerosis (MS) is a CNS disorder characterized by inflammation, demyelination and neurodegeneration, and is the most common cause of acquired nontraumatic neurological disability in young adults. The course of the disease varies between individuals: some patients accumulate minimal disability over their lives, whereas others experience a rapidly disabling disease course. This latter subset of patients, whose MS is marked by the rampant progression of disability over a short time period, is often referred to as having 'aggressive' MS. Treatment of patients with aggressive MS is challenging, and optimal strategies have yet to be defined. It is important to identify patients who are at risk of aggressive MS as early as possible and implement an effective treatment strategy. Early intervention might protect patients from irreversible damage and disability, and prevent the development of a secondary progressive course, which thus far lacks effective therapy.
Collapse
|
29
|
Börnsen L, Romme Christensen J, Ratzer R, Hedegaard C, Søndergaard HB, Krakauer M, Hesse D, Nielsen CH, Sorensen PS, Sellebjerg F. Endogenous interferon-β-inducible gene expression and interferon-β-treatment are associated with reduced T cell responses to myelin basic protein in multiple sclerosis. PLoS One 2015; 10:e0118830. [PMID: 25738751 PMCID: PMC4349448 DOI: 10.1371/journal.pone.0118830] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 01/08/2015] [Indexed: 11/18/2022] Open
Abstract
Autoreactive CD4+ T-cells are considered to play a major role in the pathogenesis of multiple sclerosis. In experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, exogenous and endogenous type I interferons restrict disease severity. Recombinant interferon-β is used for treatment of multiple sclerosis, and some untreated multiple sclerosis patients have increased expression levels of type I interferon-inducible genes in immune cells. The role of endogenous type I interferons in multiple sclerosis is controversial: some studies found an association of high expression levels of interferon-β-inducible genes with an increased expression of interleukin-10 and a milder disease course in untreated multiple sclerosis patients, whereas other studies reported an association with a poor response to treatment with interferon-β. In the present study, we found that untreated multiple sclerosis patients with an increased expression of interferon-β-inducible genes in peripheral blood mononuclear cells and interferon-β-treated multiple sclerosis patients had decreased CD4+ T-cell reactivity to the autoantigen myelin basic protein ex vivo. Interferon-β-treated multiple sclerosis patients had increased IL10 and IL27 gene expression levels in monocytes in vivo. In vitro, neutralization of interleukin-10 and monocyte depletion increased CD4+ T-cell reactivity to myelin basic protein while interleukin-10, in the presence or absence of monocytes, inhibited CD4+ T-cell reactivity to myelin basic protein. Our findings suggest that spontaneous expression of interferon-β-inducible genes in peripheral blood mononuclear cells from untreated multiple sclerosis patients and treatment with interferon-β are associated with reduced myelin basic protein-induced T-cell responses. Reduced myelin basic protein-induced CD4+ T-cell autoreactivity in interferon-β-treated multiple sclerosis patients may be mediated by monocyte-derived interleukin-10.
Collapse
Affiliation(s)
- Lars Börnsen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Jeppe Romme Christensen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Ratzer
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Chris Hedegaard
- Institute for Inflammation Research, Department of Rheumatology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Helle B. Søndergaard
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Martin Krakauer
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Dan Hesse
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Claus H. Nielsen
- Institute for Inflammation Research, Department of Rheumatology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Per S. Sorensen
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Gharibi T, Ahmadi M, Seyfizadeh N, Jadidi-Niaragh F, Yousefi M. Immunomodulatory characteristics of mesenchymal stem cells and their role in the treatment of multiple sclerosis. Cell Immunol 2015; 293:113-21. [PMID: 25596473 DOI: 10.1016/j.cellimm.2015.01.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 12/31/2014] [Accepted: 01/02/2015] [Indexed: 12/12/2022]
Abstract
Multiple Sclerosis (MS) is a chronic inflammatory neurodegenerative disease of central nervous system (CNS). Although the main cause of MS is not clear, studies suggest that MS is an autoimmune disease which attacks myelin sheath of neurons. There are different therapeutic regimens for MS patients including interferon (IFN)-β, glatiramer acetate (GA), and natalizumab. However, such therapies are not quite effective and are associated with some side effects. So which, there is no complete therapeutic method for MS patients. Regarding the potent immunomodulatory effects of mesenchymal stem cells (MSCs) and their ameliorative effects in experimental autoimmune encephalopathy (EAE), it seems that MSCs may be a new therapeutic method in MS therapy. MSC transplantation is an approach to regulate the immune system in the region of CNS lesions. In this review, we have tried to discuss about the immunomodulatory properties of MSCs and their therapeutic mechanisms in MS patients.
Collapse
Affiliation(s)
- Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Seyfizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Moore S, Khalaj AJ, Patel R, Yoon J, Ichwan D, Hayardeny L, Tiwari-Woodruff SK. Restoration of axon conduction and motor deficits by therapeutic treatment with glatiramer acetate. J Neurosci Res 2014; 92:1621-36. [PMID: 24989965 PMCID: PMC4305217 DOI: 10.1002/jnr.23440] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/25/2014] [Accepted: 05/27/2014] [Indexed: 12/20/2022]
Abstract
Glatiramer acetate (GA; Copaxone) is an approved drug for the treatment of multiple sclerosis (MS). The underlying multifactorial anti-inflammatory, neuroprotective effect of GA is in the induction of reactive T cells that release immunomodulatory cytokines and neurotrophic factors at the injury site. These GA-induced cytokines and growth factors may have a direct effect on axon function. Building on previous findings that suggest a neuroprotective effect of GA, we assessed the therapeutic effects of GA on brain and spinal cord pathology and functional correlates using the chronic experimental autoimmune encephalomyelitis (EAE) mouse model of MS. Therapeutic regimens were utilized based on promising prophylactic efficacy. More specifically, C57BL/6 mice were treated with 2 mg/mouse/day GA for 8 days beginning at various time points after EAE post-induction day 15, yielding a thorough, clinically relevant assessment of GA efficacy within the context of severe progressive disease. Therapeutic treatment with GA significantly decreased clinical scores and improved rotorod motor performance in EAE mice. These functional improvements were supported by an increase in myelinated axons and fewer amyloid precursor protein-positive axons in the spinal cords of GA-treated EAE mice. Furthermore, therapeutic GA decreased microglia/macrophage and T cell infiltrates and increased oligodendrocyte numbers in both the spinal cord and corpus callosum of EAE mice. Finally, GA improved callosal axon conduction and nodal protein organization in EAE. Our results demonstrate that therapeutic GA treatment has significant beneficial effects in a chronic mouse model of MS, in which its positive effects on both myelinated and non-myelinated axons results in improved axon function.
Collapse
Affiliation(s)
- Spencer Moore
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Anna J Khalaj
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Rhusheet Patel
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - JaeHee Yoon
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Daniel Ichwan
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Liat Hayardeny
- Pharmacology Unit, Global Innovative Research and Development, Teva Pharmaceutical IndustriesNetanya, Israel
| | - Seema K Tiwari-Woodruff
- Department of Neurology, UCLA School of MedicineLos Angeles, California
- Brain Research Institute, UCLA School of MedicineLos Angeles, California
| |
Collapse
|
32
|
Ocwieja M, Meiser K, David OJ, Valencia J, Wagner F, Schreiber SJ, Pleyer U, Ziemer S, Schmouder R. Effect of fingolimod (FTY720) on cerebral blood flow, platelet function and macular thickness in healthy volunteers. Br J Clin Pharmacol 2014; 78:1354-65. [PMID: 24976291 PMCID: PMC4256624 DOI: 10.1111/bcp.12454] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 06/23/2014] [Indexed: 12/16/2022] Open
Abstract
AIM Fingolimod, a sphingosine 1-phosphate receptor modulator, is the first oral disease modifying therapy approved for the treatment of relapsing multiple sclerosis. The aim of this double-blind, placebo-controlled study was to evaluate the effect of fingolimod on cerebral blood flow, platelet function and macular thickness in healthy volunteers. METHODS The study included 88 healthy volunteers who received fingolimod 0.5 mg or 1.25 mg or matched placebo over a period of 4 weeks. Transcranial colour coded sonography was performed to measure mean blood flow velocities, the platelet function was measured by the PFA-100® assay using a collagen/epinephrine cartridge and macular thickness was measured using optical coherence tomography. An assessment of non-inferiority of fingolimod vs. placebo was performed against a reference value (20% of the overall baseline value). RESULTS All 88 randomized participants completed the study. At day 28 compared with baseline value, for 0.5 mg, 1.25 mg and placebo treatments, the mean middle cerebral artery blood flow velocity decreased by 4, 1 and 3.7 cm s(-1), respectively. The platelet function analyzer closure time increase was not significant (7.8, 7.5 and 10.4 s, respectively). The mean percentage change in the central foveal thickness from baseline for both eyes was below 3% for all groups. The safety profile of fingolimod in this study was found consistent with the previous reports. CONCLUSIONS In healthy volunteers, the changes seen with both fingolimod doses were found to be within normal variability, non-inferior and comparable with those observed with placebo for all the pharmacodynamic parameters assessed.
Collapse
Affiliation(s)
| | | | | | | | - Frank Wagner
- Charité Research Organisation GmbHCharitéplatz 1, Berlin, Germany
| | - Stephan J Schreiber
- Department of Neurology, Universitätsmedizin BerlinCharitéplatz 1, Berlin, Germany
- Department of Neurology, Charité UniversitätsmedizinCharitéplatz 1, Berlin, Germany
| | - Uwe Pleyer
- Department of Ophthalmology, Universitätsmedizin BerlinAugustenburger Platz 1, Berlin, Germany
- Department of Ophthalmology, Charité UniversitätsmedizinCharitéplatz 1, Berlin, Germany
| | - Sabine Ziemer
- Institute for Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Universitätsmedizin BerlinCharitéplatz 1, Berlin, Germany
- Coagulation Laboratory, Charité UniversitätsmedizinCharitéplatz 1, Berlin, Germany
| | - Robert Schmouder
- Novartis Institutes for Biomedical Research, Inc.Massachusetts Avenue, Cambridge, MA, USA
| |
Collapse
|
33
|
Tepavčević V, Kerninon C, Aigrot MS, Meppiel E, Mozafari S, Arnould-Laurent R, Ravassard P, Kennedy TE, Nait-Oumesmar B, Lubetzki C. Early netrin-1 expression impairs central nervous system remyelination. Ann Neurol 2014; 76:252-68. [DOI: 10.1002/ana.24201] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/16/2014] [Accepted: 06/16/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Vanja Tepavčević
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Christophe Kerninon
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Marie Stéphane Aigrot
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Elodie Meppiel
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Sabah Mozafari
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Raphaelle Arnould-Laurent
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Philippe Ravassard
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Timothy E. Kennedy
- Department of Neurology and Neurosurgery; Montreal Neurological Institute, McGill University; Montreal Quebec Canada
| | - Brahim Nait-Oumesmar
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
| | - Catherine Lubetzki
- Pierre and Marie Curie University, University of Paris 06, UM-75, Sorbonne Universities, ICM-GH Pitié-Salpêtrière; Paris France
- National Institute of Health and Medical Research (INSERM); U1127 Paris France
- National Center for Scientific Research (CNRS), Mixed Unit of Research 7225; Paris France
- Pitié-Salpêtrière Hospital; Public Hospital Network of Paris (AP-HP); Paris France
| |
Collapse
|
34
|
Role of IL-33 and its receptor in T cell-mediated autoimmune diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:587376. [PMID: 25032216 PMCID: PMC4084552 DOI: 10.1155/2014/587376] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/26/2014] [Accepted: 05/27/2014] [Indexed: 12/14/2022]
Abstract
Interleukin-33 (IL-33) is a new cytokine of interleukin-1 family, whose specific receptor is ST2. IL-33 exerts its functions via its target cells and plays different roles in diseases. ST2 deletion and exclusion of IL-33/ST2 axis are accompanied by enhanced susceptibility to dominantly T cell-mediated organ-specific autoimmune diseases. It has been reported that IL-33/ST2 pathway plays a key role in host defense and immune regulation in inflammatory and infectious diseases. This review focuses on new findings in the roles of IL-33 and ST2 in several kinds of T cell-mediated autoimmune diseases.
Collapse
|
35
|
Soluble CD163 as a marker of macrophage activity in newly diagnosed patients with multiple sclerosis. PLoS One 2014; 9:e98588. [PMID: 24886843 PMCID: PMC4041861 DOI: 10.1371/journal.pone.0098588] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 05/05/2014] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Soluble CD163 (sCD163) is a macrophage specific protein known to be up-regulated in serum from patients with multiple sclerosis (MS). OBJECTIVE To investigate sCD163 in serum and CSF (cerebrospinal fluid) from patients undergoing MS diagnostic work-up and analyse its potential as a diagnostic biomarker. METHODS After a full MS diagnostic work-up, including collection of paired samples of CSF and serum, 183 patients were evaluated for inclusion in this study. Patients were divided into groups based on their diagnosis. Patients with normal clinical and paraclinical findings were grouped as symptomatic controls. Serum and CSF levels of sCD163 were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS sCD163 could be measured in all serum and CSF samples. A high sCD163 CSF/serum ratio in relation to molecular weight was found, strongly indicating local production in the CNS. Median levels of sCD163 were significantly decreased in serum and significantly elevated in CSF in patients with relapsing-remitting, and primary-progressive MS. There were, however, some overlaps of the measures between groups. In a receiver operating characteristic (ROC) analysis sCD163 CSF/serum ratio had an area under the curve of 0.72. CONCLUSION The sCD163 CSF/serum ratio was significantly increased in patients with MS and may reflect macrophage activation in MS lesions. These results suggest that primary progressive MS also is driven by inflammation in which the innate immune system plays a pivotal role.
Collapse
|
36
|
Rosenberger TA. Targeting calpain-mediated proteolysis and peptide signaling as a strategy to reduce injury in multiple sclerosis. J Neurochem 2014; 130:161-4. [PMID: 24844646 DOI: 10.1111/jnc.12732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 04/01/2014] [Accepted: 04/02/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Thad A Rosenberger
- University of North Dakota School of Medicine and Health Sciences, Department of Basic Sciences, Grand Forks, North Dakota
| |
Collapse
|
37
|
Contrast sensitivity in relapsing-remitting multiple sclerosis assessed by sine-wave gratings and angular frequency stimuli. Vis Neurosci 2014; 31:381-6. [PMID: 24834838 DOI: 10.1017/s0952523814000182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Previous studies have shown that multiple sclerosis (MS) affects the visual system, mainly by reducing contrast sensitivity (CS), a function that can be assessed by measuring contrast sensitivity function (CSF). To this end, we measured both the CSF for sine-wave gratings and angular frequency stimuli with 20 participants aged between 21 and 44 years, of both genders, with normal or corrected to normal visual acuity. Of these 20 participants, there were 10 volunteers with clinically defined MS of the relapsing-remitting clinical form, with no history of optic neuritis (ON), as well as 10 healthy volunteers who served as the control group (CG). We used a forced-choice detection paradigm. The results showed reduced CS to both classes of stimuli. Differences were found for sine-wave gratings at spatial frequencies of 0.5, 1.25, and 2.5 cycles per degree (cpd) (P < 0.002) and for angular frequency stimuli of 4, 24, and 48 cycles/360° (P < 0.05). On the one hand, comparing the maxima of the respective CSFs, the CS to angular frequency stimuli (24 cycles/360°) was 1.61-fold higher than that of the CS to vertical sine-wave gratings (4.0 cpd) in the CG; for the MS group, these values were 1.55-fold higher. On the other hand, CS in the MS group attained only 75% for 24 cycles/360° and 78% for 4.0 cpd of the 100% CS estimates found for the CG at the peak frequencies. These findings suggest that MS affects the visual system, mostly at its maximum contrast sensitivities. Also, since angular frequencies and sine-wave gratings operate at distinct levels of contrast in the visual system, MS seems to affect CS at both high and low levels of contrast.
Collapse
|
38
|
Yousefi F, Ebtekar M, Soleimani M, Soudi S, Hashemi SM. Comparison of in vivo immunomodulatory effects of intravenous and intraperitoneal administration of adipose-tissue mesenchymal stem cells in experimental autoimmune encephalomyelitis (EAE). Int Immunopharmacol 2013; 17:608-16. [PMID: 23973288 DOI: 10.1016/j.intimp.2013.07.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 07/14/2013] [Accepted: 07/29/2013] [Indexed: 02/06/2023]
Abstract
Due to their immunomodulatory and anti-inflammatory competence, mesenchymal stem cells (MSCs) have been considered as a suitable candidate for treatment of autoimmune diseases. Earlier studies have shown that treatment with bone marrow-derived MSCs may modulate immune responses and reduce disease severity in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Here we compare the immune regulatory properties of adipose tissue MSCs (AT-MSCs) in two independent routes of injection; namely intraperitoneal (i.p.) and intravenous (i.v.). We investigated the splenic CD4+CD25+FOXP3+ T cell population known as regulatory T cells, by flow cytometry and their brain cell infiltration by hematoxylin-eosin staining in both i.p. and i.v. routes of AT-MSC administration. We also evaluated the inflammatory cytokine profile including IFN-γ and IL-17 and anti-inflammatory cytokines such as IL-4 by ELISA technique in both routes of cell administration. We show that the i.p. route has a more pronounced effect in maintaining the splenic CD4+CD25+FOXP3+ T cell population and increase of IL-4 secretion. We also showed that i.p. injection of cells resulted in lower IFN-γ secretion and reduced cell infiltration in brain more effectively as compared to the i.v. route. The effects of AT-MSCs on down-regulation of splenocyte proliferation, IL-17 secretion and alleviating the severity of clinical scores were similar in i.p. and i.v. routes. Our data show that, due to their immunomodulative and neuroprotective effects, AT-MSCs may be a proper candidate for stem cell based MS therapy.
Collapse
Affiliation(s)
- Forouzan Yousefi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | |
Collapse
|
39
|
Global Protein Differential Expression Profiling of Cerebrospinal Fluid Samples Pooled from Chinese Sporadic CJD and non-CJD Patients. Mol Neurobiol 2013; 49:290-302. [DOI: 10.1007/s12035-013-8519-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 07/17/2013] [Indexed: 12/14/2022]
|
40
|
Borghi M, Cavallo M, Carletto S, Ostacoli L, Zuffranieri M, Picci RL, Scavelli F, Johnston H, Furlan PM, Bertolotto A, Malucchi S. Presence and significant determinants of cognitive impairment in a large sample of patients with multiple sclerosis. PLoS One 2013; 8:e69820. [PMID: 23922813 PMCID: PMC3726772 DOI: 10.1371/journal.pone.0069820] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 06/12/2013] [Indexed: 11/19/2022] Open
Abstract
Objectives To investigate the presence and the nature of cognitive impairment in a large sample of patients with Multiple Sclerosis (MS), and to identify clinical and demographic determinants of cognitive impairment in MS. Methods 303 patients with MS and 279 healthy controls were administered the Brief Repeatable Battery of Neuropsychological tests (BRB-N); measures of pre-morbid verbal competence and neuropsychiatric measures were also administered. Results Patients and healthy controls were matched for age, gender, education and pre-morbid verbal Intelligence Quotient. Patients presenting with cognitive impairment were 108/303 (35.6%). In the overall group of participants, the significant predictors of the most sensitive BRB-N scores were: presence of MS, age, education, and Vocabulary. The significant predictors when considering MS patients only were: course of MS, age, education, vocabulary, and depression. Using logistic regression analyses, significant determinants of the presence of cognitive impairment in relapsing-remitting MS patients were: duration of illness (OR = 1.053, 95% CI = 1.010–1.097, p = 0.015), Expanded Disability Status Scale score (OR = 1.247, 95% CI = 1.024–1.517, p = 0.028), and vocabulary (OR = 0.960, 95% CI = 0.936–0.984, p = 0.001), while in the smaller group of progressive MS patients these predictors did not play a significant role in determining the cognitive outcome. Conclusions Our results corroborate the evidence about the presence and the nature of cognitive impairment in a large sample of patients with MS. Furthermore, our findings identify significant clinical and demographic determinants of cognitive impairment in a large sample of MS patients for the first time. Implications for further research and clinical practice were discussed.
Collapse
Affiliation(s)
- Martina Borghi
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
| | - Marco Cavallo
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
- Department of Translational Medicine, “Amedeo Avogadro” University of Eastern Piedmont, Novara, Italy
- * E-mail:
| | - Sara Carletto
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
| | - Luca Ostacoli
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
| | - Marco Zuffranieri
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
| | - Rocco Luigi Picci
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
| | - Francesco Scavelli
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
| | - Harriet Johnston
- School of Psychology, University of St Andrews, St Andrews, Scotland, United Kingdom
| | - Pier Maria Furlan
- Department of Mental Health, “San Luigi Gonzaga” Hospital Medical School, University of Turin, ASL TO3, Orbassano, Italy
| | - Antonio Bertolotto
- Neurologia 2 – CRESM (Regional Reference Centre for Multiple Sclerosis), “San Luigi Gonzaga” Hospital Medical School, Orbassano, Italy
| | - Simona Malucchi
- Neurologia 2 – CRESM (Regional Reference Centre for Multiple Sclerosis), “San Luigi Gonzaga” Hospital Medical School, Orbassano, Italy
| |
Collapse
|
41
|
Menezes KM, Copetti F, Wiest MJ, Trevisan CM, Silveira AF. Efeito da equoterapia na estabilidade postural de portadores de esclerose múltipla: estudo preliminar. FISIOTERAPIA E PESQUISA 2013. [DOI: 10.1590/s1809-29502013000100008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJETIVO: Este estudo se propôs a verificar se a estimulação por meio da equoterapia é capaz de desencadear alterações no controle postural de portadores de esclerose múltipla (EM). MÉTODO: Fizeram parte deste estudo 11 portadores de EM divididos em Grupo Intervenção (GI) e Grupo Controle (GC). O GI foi inserido num programa de hipoterapia durante 4 meses, sendo conduzidas 2 sessões semanais com duração de 50 minutos cada. A estabilidade postural foi avaliada utilizando uma plataforma de força (para calcular o deslocamento do centro de pressão (COP), durante 30 segundos, em postura ereta quasi-estática, com olhos abertos e fechados, antes e após o treinamento com equoterapia. O tratamento estatístico foi feito através do ANOVA e Post hoc de Tukey com p<0,05. RESULTADOS: A amplitude de oscilação anteroposterior reduziu significativamente no GI após a estimulação, enquanto o GC manteve um comportamento inalterado entre as avaliações. Em ambos os grupos, a estabilidade postural foi reduzida com a supressão da informação visual. CONCLUSÕES: A adaptação funcional proporcionada pela equoterapia foi capaz de melhorar a estabilidade postural dos portadores de EM. Os resultados corroboram a literatura que suporta o uso da equoterapia como uma intervenção em potencial nas desordens de controle postural em portadores de EM.
Collapse
|
42
|
Brain pericyte plasticity as a potential drug target in CNS repair. Drug Discov Today 2012; 18:456-63. [PMID: 23266366 DOI: 10.1016/j.drudis.2012.12.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/23/2012] [Accepted: 12/11/2012] [Indexed: 01/09/2023]
Abstract
Brain pericytes (BrPCs) are essential cellular components of the central nervous system neurovascular unit involved in the regulation of blood flow, blood-brain barrier function, as well as in the stabilization of the vessel architecture. More recently, it became evident that BrPCs, besides their regulatory activities in brain vessel function and homeostasis, have pleiotropic functions in the adult CNS ranging from stromal and regeneration promoting activities to stem cell properties. This special characteristic confers BrPC cell plasticity, being able to display features of other cells within the organism. BrPCs might also be causally involved in certain brain diseases. Due to these properties BrPCs might be potential drug targets for future therapies of neurological disorders. This review summarizes BrPC properties, disorders in which this cell type might be involved, and provides suggestions for future therapeutic developments targeting BrPCs.
Collapse
|
43
|
Liu J, Yin L, Dong H, Xu E, Zhang L, Qiao Y, Liu Y, Li L, Jia J. Decreased serum levels of nucleolin protein fragment, as analyzed by bead-based proteomic technology, in multiple sclerosis patients compared to controls. J Neuroimmunol 2012; 250:71-6. [PMID: 22633274 DOI: 10.1016/j.jneuroim.2012.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 05/01/2012] [Accepted: 05/02/2012] [Indexed: 11/24/2022]
Abstract
In order to investigate the biomarkers associated with relapsing-remitting multiple sclerosis (RRMS), we analyzed 72 patients with RRMS and 65 healthy controls using proteome technology. Peptides in sera were purified using magnetic beads, and analyzed by matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry and ClinProTool software. Thirteen peptides were significantly different between patients with RRMS and healthy controls. Furthermore, a pattern of peaks was selected for genetic algorithm (GA), supervised neural network (SNN) and quick classifier (QC) model building. Among these three models, GA method was best with 93.49% of recognition capability and 82.66% of cross-validation and discriminated the proteomic spectra in patients with RRMS from healthy controls, with a sensitivity of 80% and a specificity of 91.3%. Meanwhile, the first peptide with m/z 2023.3 was identified as fragment of nucleolin protein. There is a possible relationship between the fragment peptide of nucleolin and the trigger of relapse in MS. Sera nucleolin may serve as a possible biomarker of RRMS.
Collapse
Affiliation(s)
- Jianghong Liu
- Department of Neurology, Xuan Wu Hospital of Capital Medical University, Beijing, 100053, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Petersen T, Møller-Larsen A, Ellermann-Eriksen S, Thiel S, Christensen T. Effects of interferon-beta therapy on elements in the antiviral immune response towards the human herpesviruses EBV, HSV, and VZV, and to the human endogenous retroviruses HERV-H and HERV-W in multiple sclerosis. J Neuroimmunol 2012; 249:105-8. [PMID: 22608883 DOI: 10.1016/j.jneuroim.2012.04.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 04/23/2012] [Accepted: 04/24/2012] [Indexed: 12/22/2022]
Abstract
Effects of treatment of multiple sclerosis patients with IFN-β on elements of the antiviral immune response to herpesviruses were analysed in a longitudinal study. We found significantly increased seroreactivity to EBV EBNA-1, and to VZV, in patients who did not respond to IFN-β therapy. We found no significant changes in seroreactivity to EBV EA, or to HSV. For the same patient cohort, we have previously demonstrated significant decreases in seroreactivities to envelope antigens for the two human endogenous retroviruses HERV-H and HERV-W, closely linked to efficacy of therapy. We further searched for correlations between seroreactivities to EBV, HSV, and VZV, and levels of mannan-binding lectin (MBL), and MBL-associated serine protease 3. We found no such correlations. Our results are in accord with recent reports of increased seroreactivity to EBV EBNA-1, and to VZV in active MS, and they support that the herpesviruses EBV and VZV together with HERV-H/HERV-W and the antiviral immune response may play a role in MS development.
Collapse
Affiliation(s)
- Thor Petersen
- Department of Neurology, Aarhus University Hospital, Nørrebrogade 44, Aarhus C, Denmark
| | | | | | | | | |
Collapse
|
45
|
Kumagai C, Kalman B, Middleton FA, Vyshkina T, Massa PT. Increased promoter methylation of the immune regulatory gene SHP-1 in leukocytes of multiple sclerosis subjects. J Neuroimmunol 2012; 246:51-7. [PMID: 22458980 DOI: 10.1016/j.jneuroim.2012.03.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 12/21/2022]
Abstract
The protein tyrosine phosphatase, SHP-1, is a negative regulator of proinflammatory signaling and autoimmune disease. We have previously reported reduced SHP-1 expression in peripheral blood leukocytes of subjects with multiple sclerosis (MS). Recent evidence indicates that virus-induced DNA methylation of the SHP-1 promoter is responsible for aberrant silencing of SHP-1 expression and function in hematopoietic cells that might relate to inflammatory diseases. In the present study, bisulfite sequencing of the SHP-1 promoter demonstrated that over a third of MS subjects had abnormally high promoter methylation. As SHP-1 is deficient in MS leukocytes and SHP-1-regulated proinflammatory genes are correspondingly upregulated, we propose that increased SHP-1 promoter methylation may relate in part to decreased SHP-1 expression and increased leukocyte-mediated inflammation in MS.
Collapse
Affiliation(s)
- Chiharu Kumagai
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | | | | | | | |
Collapse
|
46
|
Christophi GP, Gruber RC, Panos M, Christophi RL, Jubelt B, Massa PT. Interleukin-33 upregulation in peripheral leukocytes and CNS of multiple sclerosis patients. Clin Immunol 2012; 142:308-19. [PMID: 22189043 PMCID: PMC3288946 DOI: 10.1016/j.clim.2011.11.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 11/11/2011] [Accepted: 11/21/2011] [Indexed: 01/16/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system (CNS). Here we document for the first time that the cytokine IL-33 is upregulated in both the periphery and the CNS of MS patients. Plasma IL-33 was elevated in MS patients compared to normal subjects and a three-month treatment of MS patients with interferon β-1a resulted in a significant decrease of IL-33 levels. Similarly, stimulated cultured lymphocytes and macrophages from MS patients had elevated IL-33 levels compared to normal subjects. In parallel, the transcription factor NF-κB that mediates IL-33 transcription was also elevated in leukocytes of MS patients. IL-33 was elevated in normal-appearing white matter and plaque areas from MS brains and astrocytes were identified as an important source of IL-33 expression in the CNS. In summary, IL-33 levels are elevated in the periphery and CNS of MS patients, implicating IL-33 in the pathogenesis of MS.
Collapse
Affiliation(s)
- George P. Christophi
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ross C. Gruber
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Panos
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rebecca L. Christophi
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Burk Jubelt
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paul T. Massa
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
47
|
Jadasz JJ, Aigner L, Rivera FJ, Küry P. The remyelination Philosopher's Stone: stem and progenitor cell therapies for multiple sclerosis. Cell Tissue Res 2012; 349:331-47. [PMID: 22322424 DOI: 10.1007/s00441-012-1331-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 01/16/2012] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease that leads to oligodendrocyte loss and subsequent demyelination of the adult central nervous system (CNS). The pathology is characterized by transient phases of recovery during which remyelination can occur as a result of resident oligodendroglial precursor and stem/progenitor cell activation. However, myelin repair efficiency remains low urging the development of new therapeutical approaches that promote remyelination activities. Current MS treatments target primarily the immune system in order to reduce the relapse rate and the formation of inflammatory lesions, whereas no therapies exist in order to regenerate damaged myelin sheaths. During the last few years, several transplantation studies have been conducted with adult neural stem/progenitor cells and glial precursor cells to evaluate their potential to generate mature oligodendrocytes that can remyelinate axons. In parallel, modulation of the endogenous progenitor niche by neural and mesenchymal stem cell transplantation with the aim of promoting CNS progenitor differentiation and myelination has been studied. Here, we summarize these findings and discuss the properties and consequences of the various molecular and cell-mediated remyelination approaches. Moreover, we address age-associated intrinsic cellular changes that might influence the regenerative outcome. We also evaluate the extent to which these experimental treatments might increase the regeneration capacity of the demyelinated human CNS and hence be turned into future therapies.
Collapse
Affiliation(s)
- Janusz J Jadasz
- Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | | | | | | |
Collapse
|
48
|
Martino G, Pluchino S, Bonfanti L, Schwartz M. Brain regeneration in physiology and pathology: the immune signature driving therapeutic plasticity of neural stem cells. Physiol Rev 2011; 91:1281-304. [PMID: 22013212 PMCID: PMC3552310 DOI: 10.1152/physrev.00032.2010] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Regenerative processes occurring under physiological (maintenance) and pathological (reparative) conditions are a fundamental part of life and vary greatly among different species, individuals, and tissues. Physiological regeneration occurs naturally as a consequence of normal cell erosion, or as an inevitable outcome of any biological process aiming at the restoration of homeostasis. Reparative regeneration occurs as a consequence of tissue damage. Although the central nervous system (CNS) has been considered for years as a "perennial" tissue, it has recently become clear that both physiological and reparative regeneration occur also within the CNS to sustain tissue homeostasis and repair. Proliferation and differentiation of neural stem/progenitor cells (NPCs) residing within the healthy CNS, or surviving injury, are considered crucial in sustaining these processes. Thus a large number of experimental stem cell-based transplantation systems for CNS repair have recently been established. The results suggest that transplanted NPCs promote tissue repair not only via cell replacement but also through their local contribution to changes in the diseased tissue milieu. This review focuses on the remarkable plasticity of endogenous and exogenous (transplanted) NPCs in promoting repair. Special attention will be given to the cross-talk existing between NPCs and CNS-resident microglia as well as CNS-infiltrating immune cells from the circulation, as a crucial event sustaining NPC-mediated neuroprotection. Finally, we will propose the concept of the context-dependent potency of transplanted NPCs (therapeutic plasticity) to exert multiple therapeutic actions, such as cell replacement, neurotrophic support, and immunomodulation, in CNS repair.
Collapse
Affiliation(s)
- Gianvito Martino
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.
| | | | | | | |
Collapse
|
49
|
Efficient CRM197-mediated drug targeting to monocytes. J Control Release 2011; 158:139-47. [PMID: 21982901 DOI: 10.1016/j.jconrel.2011.09.091] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 09/21/2011] [Accepted: 09/23/2011] [Indexed: 01/04/2023]
Abstract
Efficient delivery of drugs to specific cellular reservoirs is of particular importance for therapeutics that are not able to pass cellular barriers and that may have unwanted side effects in off-target tissues. Heparin-binding epidermal growth factor (HB-EGF) is expressed on leukocytes and may be targeted for specific drug delivery using cross-reacting material (CRM)197, a non-toxic variant of diphtheria toxin and exogenous substrate for HB-EGF. We used fluorescently labeled CRM197 and CRM197-coated liposomes to investigate their potential use for drug delivery to leukocytes. We demonstrate that CRM197-guided systems are efficiently taken up by human leukocytes in vitro. CRM197 was also found to specifically target leukocytes in vivo in mice with components of the human immune system (HIS mice) and hamsters. Monocytes represent the most prominent subset of leukocytes that showed highly specific CRM197-mediated uptake. We therefore propose the application of CRM197 as a novel targeting approach in diseases that require the selective treatment of monocytes.
Collapse
|
50
|
Yang Y, Tian SJ, Wu L, Huang DH, Wu WP. Fibrinogen depleting agent batroxobin has a beneficial effect on experimental autoimmune encephalomyelitis. Cell Mol Neurobiol 2011; 31:437-48. [PMID: 21165693 DOI: 10.1007/s10571-010-9637-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 12/02/2010] [Indexed: 10/18/2022]
Abstract
Multiple sclerosis (MS) was characterized with widespread demyelination and axonal loss of central nervous system (CNS). Fibrinogen (fibrin) deposition was considered as one of the pathogenesis of MS. Therefore, we explored the effects of fibrinogen depleting agent batroxobin in experimental autoimmune encephalomyelitis (EAE) mice model. Our study showed that prevention and suppression with batroxobin significantly ameliorated clinical severity of EAE, reduced inflammatory cells infiltration, and demyelination, and suppressed the activation of astrocytes and macrophages comprising the CD11b(+) population. Batroxobin treatment leads to reduced expression of p-Akt and increased expression of MBP as compared to control. In addition, batroxobin treatment partly reversed the dendric-like formation of macrophages irritated by fibrinogen in vitro. The reduced severity of EAE mice treated with batroxobin suggests that strategy targeting fibrin as a potential therapy for EAE may be beneficial for the treatment of MS patients.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurology, The General Hospital of Chinese PLA, Beijing, China.
| | | | | | | | | |
Collapse
|