1
|
Zhang W, Liu Z, Wang K, Zhang L, Liu S, He K, Wang H, Wang J, Wang Y, Yang Y, Zhang X, Wu H. Quantitative proteomic landscape of the pathophysiology of adhesive arachnoiditis and its clinical significance: Structure and mechanism of TNC and RANBP1 proteins. Int J Biol Macromol 2024; 287:138444. [PMID: 39662555 DOI: 10.1016/j.ijbiomac.2024.138444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
The pathophysiological mechanism of adhesive arachnoiditis (AA) is complex, involving the interaction of multiple proteins. In recent years, the development of quantitative proteomics technology has provided a new perspective to reveal its pathological mechanism. The main objective of this study was to reveal the changes of protein expression profiles in arachnoid tissue of patients with AA. Proteomic analysis of arachnoid tissue samples was performed by high-throughput mass spectrometry. Bioinformatics tools were used to process and analyze the data and screen out 712 differentially expressed proteins (DEPs). Specially, 2 hub DEPs (TNC and RANBP1) were found as potential diagnostic markers. The expression levels of TNC and RANBP1 proteins were significantly up-regulated in patients with AA. TNC protein played a key role in inflammation and extracellular matrix remodeling, while RANBP1 was involved in cytoplasmic transport and cell cycle regulation. The abnormal expression and modification of these two proteins were closely related to the pathophysiological process of the disease. Immunoinfiltration analysis found the pathological process of AA were related to the infiltration of memory B cells, activated NK cells and CD8(+) T cells. Additonally, weighted gene co-expression network analysis (WGCNA) showed the organization of the arachnoid proteome into a network of 28 biologically meaningful modules of co-expressed proteins. Of these, 2 modules were positively correlated to AA phenotypes. This study shows a distinct proteomic landscape of AA and non-adhesive arachnoiditis (nAA). These findings also provide valuable insights into the molecular changes associated with potential mechanisms underlying AA.
Collapse
Affiliation(s)
- Weikang Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Zhenlei Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Kai Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Lei Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Shaocheng Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Kun He
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - He Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Junyi Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Yaobin Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Yuhua Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Xiangyu Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Chang Chun street No.45, Xicheng, Beijing, China.
| |
Collapse
|
2
|
Gruber RC, Wirak GS, Blazier AS, Lee L, Dufault MR, Hagan N, Chretien N, LaMorte M, Hammond TR, Cheong A, Ryan SK, Macklin A, Zhang M, Pande N, Havari E, Turner TJ, Chomyk A, Christie E, Trapp BD, Ofengeim D. BTK regulates microglial function and neuroinflammation in human stem cell models and mouse models of multiple sclerosis. Nat Commun 2024; 15:10116. [PMID: 39578444 PMCID: PMC11584639 DOI: 10.1038/s41467-024-54430-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Neuroinflammation in the central nervous system (CNS), driven largely by resident phagocytes, has been proposed as a significant contributor to disability accumulation in multiple sclerosis (MS) but has not been addressed therapeutically. Bruton's tyrosine kinase (BTK) is expressed in both B-lymphocytes and innate immune cells, including microglia, where its role is poorly understood. BTK inhibition may provide therapeutic benefit within the CNS by targeting adaptive and innate immunity-mediated disease progression in MS. Using a CNS-penetrant BTK inhibitor (BTKi), we demonstrate robust in vivo effects in mouse models of MS. We further identify a BTK-dependent transcriptional signature in vitro, using the BTKi tolebrutinib, in mouse microglia, human induced pluripotent stem cell (hiPSC)-derived microglia, and a complex hiPSC-derived tri-culture system composed of neurons, astrocytes, and microglia, revealing modulation of neuroinflammatory pathways relevant to MS. Finally, we demonstrate that in MS tissue BTK is expressed in B-cells and microglia, with increased levels in lesions. Our data provide rationale for targeting BTK in the CNS to diminish neuroinflammation and disability accumulation.
Collapse
Affiliation(s)
- Ross C Gruber
- Sanofi, Cambridge, MA, USA
- Takeda, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - Nilesh Pande
- Sanofi, Cambridge, MA, USA
- Voyager Therapeutics Inc, Cambridge, MA, USA
| | | | | | - Anthony Chomyk
- Department of Neurosciences, Cleveland Clinic, Cambridge, OH, USA
| | - Emilie Christie
- Department of Neurosciences, Cleveland Clinic, Cambridge, OH, USA
| | - Bruce D Trapp
- Department of Neurosciences, Cleveland Clinic, Cambridge, OH, USA
| | | |
Collapse
|
3
|
Lim JH, Neuwirth A, Chung KJ, Grossklaus S, Soehnlein O, Hajishengallis G, Chavakis T. Formyl peptide receptor 2 regulates dendritic cell metabolism and Th17 cell differentiation during neuroinflammation. Front Immunol 2024; 15:1354074. [PMID: 39148732 PMCID: PMC11324504 DOI: 10.3389/fimmu.2024.1354074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 07/18/2024] [Indexed: 08/17/2024] Open
Abstract
Formyl peptide receptor 2 (FPR2) is a receptor for formylated peptides and specific pro-resolving mediators, and is involved in various inflammatory processes. Here, we aimed to elucidate the role of FPR2 in dendritic cell (DC) function and autoimmunity-related central nervous system (CNS) inflammation by using the experimental autoimmune encephalomyelitis (EAE) model. EAE induction was accompanied by increased Fpr2 mRNA expression in the spinal cord. FPR2-deficient (Fpr2 KO) mice displayed delayed onset of EAE compared to wild-type (WT) mice, associated with reduced frequencies of Th17 cells in the inflamed spinal cord at the early stage of the disease. However, FPR2 deficiency did not affect EAE severity after the disease reached its peak. FPR2 deficiency in mature DCs resulted in decreased expression of Th17 polarizing cytokines IL6, IL23p19, IL1β, and thereby diminished the DC-mediated activation of Th17 cell differentiation. LPS-activated FPR2-deficient DCs showed upregulated Nos2 expression and nitric oxide (NO) production, as well as reduced oxygen consumption rate and impaired mitochondrial function, including decreased mitochondrial superoxide levels, lower mitochondrial membrane potential and diminished expression of genes related to the tricarboxylic acid cycle and genes related to the electron transport chain, as compared to WT DCs. Treatment with a NO inhibitor reversed the reduced Th17 cell differentiation in the presence of FPR2-deficient DCs. Together, by regulating DC metabolism, FPR2 enhances the production of DC-derived Th17-polarizing cytokines and hence Th17 cell differentiation in the context of neuroinflammation.
Collapse
Affiliation(s)
- Jong-Hyung Lim
- Laboratory of Innate Immunity and Inflammation, Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ales Neuwirth
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Grossklaus
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - George Hajishengallis
- Laboratory of Innate Immunity and Inflammation, Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
4
|
Milne SM, Lahiri A, Sanchez CL, Marshall MJ, Jahan I, Meares GP. Myelin oligodendrocyte glycoprotein reactive Th17 cells drive Janus Kinase 1 dependent transcriptional reprogramming in astrocytes and alter cell surface cytokine receptor profiles during experimental autoimmune encephalomyelitis. Sci Rep 2024; 14:13146. [PMID: 38849434 PMCID: PMC11161502 DOI: 10.1038/s41598-024-63877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease affecting the central nervous system (CNS). T helper (Th) 17 cells are involved in the pathogenesis of MS and its animal model of experimental autoimmune encephalomyelitis (EAE) by infiltrating the CNS and producing effector molecules that engage resident glial cells. Among these glial cells, astrocytes have a central role in coordinating inflammatory processes by responding to cytokines and chemokines released by Th17 cells. In this study, we examined the impact of pathogenic Th17 cells on astrocytes in vitro and in vivo. We identified that Th17 cells reprogram astrocytes by driving transcriptomic changes partly through a Janus Kinase (JAK)1-dependent mechanism, which included increased chemokines, interferon-inducible genes, and cytokine receptors. In vivo, we observed a region-specific heterogeneity in the expression of cell surface cytokine receptors on astrocytes, including those for IFN-γ, IL-1, TNF-α, IL-17, TGFβ, and IL-10. Additionally, these receptors were dynamically regulated during EAE induced by adoptive transfer of myelin-reactive Th17 cells. This study overall provides evidence of Th17 cell reprogramming of astrocytes, which may drive changes in the astrocytic responsiveness to cytokines during autoimmune neuroinflammation.
Collapse
MESH Headings
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Animals
- Astrocytes/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Mice
- Myelin-Oligodendrocyte Glycoprotein
- Receptors, Cytokine/metabolism
- Receptors, Cytokine/genetics
- Janus Kinase 1/metabolism
- Mice, Inbred C57BL
- Cytokines/metabolism
- Cellular Reprogramming
- Female
- Cells, Cultured
Collapse
Affiliation(s)
- Sarah M Milne
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Anirudhya Lahiri
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Cristina L Sanchez
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA
| | - Micah J Marshall
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Ishrat Jahan
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA
| | - Gordon P Meares
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Neurology, The Ohio State University College of Medicine, IBMR 415D, 460 Medical Center Drive, Columbus, OH, 43210, USA.
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA.
- Rockefeller Neuroscience Institute, Morgantown, WV, 26506, USA.
| |
Collapse
|
5
|
Nigam M, Devi K, Coutinho HDM, Mishra AP. Exploration of gut microbiome and inflammation: A review on key signalling pathways. Cell Signal 2024; 118:111140. [PMID: 38492625 DOI: 10.1016/j.cellsig.2024.111140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
The gut microbiome, a crucial component of the human system, is a diverse collection of microbes that belong to the gut of human beings as well as other animals. These microbial communities continue to coexist harmoniously with their host organisms and perform various functions that affect the host's general health. Each person's gut microbiota has a unique makeup. The gut microbiota is well acknowledged to have a part in the local as well as systemic inflammation that underlies a number of inflammatory disorders (e.g., atherosclerosis, diabetes mellitus, obesity, and inflammatory bowel disease).The gut microbiota's metabolic products, such as short-chain fatty acids (butyrate, propionate, and acetate) inhibit inflammation by preventing immune system cells like macrophages and neutrophils from producing pro-inflammatory factors, which are triggered by the structural elements of bacteria (like lipopolysaccharide). The review's primary goal is to provide comprehensive and compiled data regarding the contribution of gut microbiota to inflammation and the associated signalling pathways.
Collapse
Affiliation(s)
- Manisha Nigam
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India.
| | - Kanchan Devi
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India
| | | | - Abhay Prakash Mishra
- Department of Pharmacology, University of Free State, Bloemfontein 9300, South Africa.
| |
Collapse
|
6
|
Saedmocheshi S, Yousfi N, Chamari K. Breaking boundaries: the transformative role of exercise in managing multiple sclerosis. EXCLI JOURNAL 2024; 23:475-490. [PMID: 38741722 PMCID: PMC11089092 DOI: 10.17179/excli2024-6932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/13/2024] [Indexed: 05/16/2024]
Abstract
Multiple sclerosis (MS) is a prevalent cause of physical disability in adults, with inflammation-induced demyelination and neurodegeneration contributing to its etiology. This comprehensive review explores the multifaceted benefits of exercise in managing MS, including improvements in aerobic capacity, balance, muscle strength, immune and hormonal functions and mood. Various exercise modalities, such as aerobic, resistance, flexibility, and balance training, are discussed, along with tailored protocols for MS patients. Recommended exercise strategies are: aerobic exercise: 2-3x/week; 10-30 minutes (40 %-60 % of maximum heart rate (HRmax), HIIT: 1x/week, five 30-90-second intervals at 90 %-100 % HRmax, Resistance training: 2-3x/week, 5-10 exercises; 1-3 sets for each exercise, 8-15 repetitions/set. The review also examines the impact of exercise on neuroplasticity, cardiovascular responses, cytokine modulation, stress hormone regulation, brain structure, and function and fatigue perception. Emphasizing the importance of exercise in enhancing the quality of life for individuals with MS, the review proposes exercise prescriptions and highlights the promising link between physical activity, brain health, and improved hormonal and immune status in MS patients. This review aims to inform future research and guide clinical practices for effective MS management.
Collapse
Affiliation(s)
- Saber Saedmocheshi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj, Iran
| | - Narimen Yousfi
- Tunisian Research Laboratory "Sport Performance Optimisation", (LR09SEP01) National Center of Medicine and Science in Sport, Tunis, Tunisia
| | - Karim Chamari
- Higher Institute of Sport and Physical Education, ISSEP Ksar Said, Manouba University, Tunis, Tunisia
| |
Collapse
|
7
|
Kim M, Kim WS, Cha H, Kim B, Kwon YN, Kim SM. Early involvement of peripherally derived monocytes in inflammation in an NMO-like mouse model. Sci Rep 2024; 14:1177. [PMID: 38216632 PMCID: PMC10786844 DOI: 10.1038/s41598-024-51759-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune inflammatory disease that primarily affects the optic nerve and spinal cord within the central nervous system (CNS). Acute astrocyte injury caused by autoantibodies against aquaporin 4 (NMO-IgG) is a well-established key factor in the pathogenesis, ultimately leading to neuronal damage and patient disability. In addition to these humoral immune processes, numerous innate immune cells were found in the acute lesions of NMO patients. However, the origin and function of these innate immune cells remain unclear in NMO pathogenesis. Therefore, this study aims to analyze the origin and functions of these innate immune cells in an NMO-like mouse model and evaluate their role in the pathophysiology of NMO. The expression of Tmem119 on Iba1 + cells in brain tissue disappeared immediately after the injection of NMO-IgG + human complement mixture, while the expression of P2ry12 remained well-maintained at 1 day after injection. Based on these observations, it was demonstrated that monocytes infiltrate the brain during the early stages of the pathological process and are closely associated with the inflammatory response through the expression of the proinflammatory cytokine IL-1β. Understanding the variations in the expression patterns of P2ry12, Tmem119, and other markers could be helpful in distinguishing between these cell types and further analyzing their functions. Therefore, this research may contribute to a better understanding of the mechanisms and potential treatments for NMO.
Collapse
Affiliation(s)
- Moonhang Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea.
| | - Won Seok Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea
| | - Hyeuk Cha
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea
| | - Boram Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea
| | - Young Nam Kwon
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sung Min Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
8
|
Lafi And ZK, Mohammed BJ. Relationship between vitamin D receptor genotypes (FOK1rs2228570) and IL18 gene expression in sample of multiple sclerosis Iraqi patients. Hum Antibodies 2024; 32:1-8. [PMID: 38339924 DOI: 10.3233/hab-230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
BACKGROUND Multiple Sclerosis known as MS, this chronic inflammatory demyelinating condition affects the nervous system. It is a heterogenic and multifactorial disease. The goal of the current study was to investigate the relationship between MS patients' IL18 gene expression and the vitamin D receptor gene polymorphism (FOK1rs2228570). OBJECTIVE The aim of the study to investigate the association of vitamin D receptor (FOK1rs2228570) gene polymorphism and pro inflammatory cytokine (IL18) gene expression among multiple sclerosis Iraqi patients. Detection VDR polymorphism and determine whether this SNP is involved in susceptibility to multiple sclerosis and estimation IL18 gene expression and explore its relation with multiple sclerosis susceptibility. METHODS Blood samples were taken from 75 MS patients in Iraq (30 men and 45 women), as well as from 75 volunteers who seemed to be in a favorable state of health and fell within the age range of 20 to 50 years. Tetra-ARMS Polymerase Chain Reaction (Tetra-ARMS PCR) was used to find polymorphisms in the vitamin D receptor (VDR) gene, and Real-time Polymerase Chain Reaction (RT-PCR) was used to measure IL18 gene expression. RESULTS The findings from the analysis of VDR gene polymorphism in patients with MS indicated that the wild-type genotype T/T was present in 8 individuals, accounting for 10.6%, the heterogeneous genotype TC was 36 (48%), and the homogeneous genotype CC was 31 (41.3%), whilst T allele frequency was 52(34.6%) and C allele was 98(65.3%) with (P⩽ 0.01) significant difference and even as in control T/T genotype was 49(65.3%), TC genotype was 21(28%), CC genotype was 5(6.66%), T allele frequency was 119(79.3%) and C allele was 31(20.6%) with significant difference (P⩽ 0.001). While estimation of IL18 expression showed high elevation in patients' group (2.59 ± 0.51 fold) by significance difference (P⩽ 0.5) when compared to control group (1.35 ± 0.14 fold). The relationship between IL18 gene expression with VDR variant in MS patients demonstrated a significant rise (2.9 ± 0.51 fold) at CC genotype patients in IL18 folding gene expression, followed by (4.6 ± 0.17 fold) in TC genotype patients and finally (1.4 ± 0.08 fold) in TT genotype patients with highly significant (P⩽ 0.01). CONCLUSION The VDR(FOK1rs2228570) genotype was significantly correlated with IL18 expression in MS patients from Iraq.
Collapse
|
9
|
Denaroso GE, Smith Z, Angeliu CG, Cheli VT, Wang C, Paez PM. Deletion of voltage-gated calcium channels in astrocytes decreases neuroinflammation and demyelination in a murine model of multiple sclerosis. J Neuroinflammation 2023; 20:263. [PMID: 37964385 PMCID: PMC10644533 DOI: 10.1186/s12974-023-02948-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/05/2023] [Indexed: 11/16/2023] Open
Abstract
The experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis was used in combination with a Cav1.2 conditional knock-out mouse (Cav1.2KO) to study the role of astrocytic voltage-gated Ca++ channels in autoimmune CNS inflammation and demyelination. Cav1.2 channels were specifically ablated in Glast-1-positive astrocytes by means of the Cre-lox system before EAE induction. After immunization, motor activity was assessed daily, and a clinical score was given based on the severity of EAE symptoms. Cav1.2 deletion in astrocytes significantly reduced the severity of the disease. While no changes were found in the day of onset and peak disease severity, EAE mean clinical score was lower in Cav1.2KO animals during the chronic phase of the disease. This corresponded to better performance on the rotarod and increased motor activity in Cav1.2KO mice. Furthermore, decreased numbers of reactive astrocytes, activated microglia, and infiltrating lymphocytes were found in the lumbar section of the spinal cord of Cav1.2KO mice 40 days after immunization. The degree of myelin protein loss and size of demyelinated lesions were also attenuated in Cav1.2KO spinal cords. Similar results were found in EAE animals treated with nimodipine, a Cav1.2 Ca++ channel inhibitor with high affinity to the CNS. Mice injected with nimodipine during the acute and chronic phases of the disease exhibited lower numbers of reactive astrocytes, activated microglial, and infiltrating immune cells, as well as fewer demyelinated lesions in the spinal cord. These changes were correlated with improved clinical scores and motor performance. In summary, these data suggest that antagonizing Cav1.2 channels in astrocytes during EAE alleviates neuroinflammation and protects the spinal cord from autoimmune demyelination.
Collapse
Affiliation(s)
- G E Denaroso
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - Z Smith
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - C G Angeliu
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - V T Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - C Wang
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA
| | - P M Paez
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, NYS Center of Excellence, 701 Ellicott St., Buffalo, NY, 14203, USA.
| |
Collapse
|
10
|
Grimaldi-Bensouda L, Papeix C, Hamon Y, Benichou J, Abenhaim L. Association between vaccination and the risk of central demyelination: results from a case-referent study. J Neurol 2023; 270:4678-4686. [PMID: 37351662 PMCID: PMC10511379 DOI: 10.1007/s00415-023-11822-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Few studies documented the potential association between vaccination and the risk of central demyelination (CD). Specifically, anti-hepatitis B and anti-human papillomavirus (HPV) vaccines have been the subject of distrust with regard to their implication to trigger CD. METHODS From a systematic national registry, patients with first signs of CD (cases) were identified and documented for their exposure to vaccination up to 24 months before the first signs occurred. This exposure was compared to that of a representative sample of general practice patients without a history of CD, randomly selected from a national registry (referents). CD cases were 2:1 matched on age, sex, index date (ID), and region of residence. Vaccines against influenza, HPV, hepatitis B and diphtheria-tetanus-pertussis-poliomyelitis-haemophilus (DTPPHae) were considered. Associations between vaccination and CD were assessed using multivariate conditional logistic regressions, controlled for confounding factors. FINDINGS 564 CD cases were matched to 1,128 randomly selected referents (age range: 2-79 years old). Overall, 123 (22%) CD cases and 320 (28%) referents had received at least one vaccine within 24 months before ID. Adjusted odds ratios (ORs) for any vaccination were 0.69, 95% confidence interval (CI) [0.54-0.88] with respect to any CD first signs, 0.68 [0.51-0.90] for myelitis and 0.70 [0.42-1.17] for optic neuritis. Adjusted ORs for any CD first signs were 1.02 [0.71-1.47] for influenza vaccine (administered in 9.6% of cases and 10.4% of referents) and 0.72 [0.53-0.99] for DTPPHae vaccine (administered in 10.8% of cases and 14.5% of referents). Vaccines against hepatitis B and HPV were only administered in 1.1% and 1.2% of cases and in 2.9% and 3.2% of referents respectively, which statistically explained the point estimates < 1 (ORs of 0.39 [0.16-0.94] and of 0.32 [0.13-0.80]). INTERPRETATION No increased risk of CD incidence was observed amongst vaccinated patients. Lower rates of vaccination against hepatitis B and HPV observed in patients with CD compared to referents may be due to the reluctance of physicians to vaccinate patients considered at risk of CD.
Collapse
Affiliation(s)
- Lamiae Grimaldi-Bensouda
- The PGRx Study Group, Paris, France
- Pharmacology Department, Hospital Group Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Garches, France
- University of Versailles-Paris Saclay, Montigny Le Bretonneux, France
- Inserm U 1018 CESP, Villejuif, France
| | - Caroline Papeix
- Neurology Department of Hospital foundation A de Rothschild, Paris, France
- Paris-Cité University, Paris, France
| | - Yann Hamon
- The PGRx Study Group, Paris, France
- RESAL, LA-SER Group, Paris, France
| | - Jacques Benichou
- Inserm U 1018 CESP, Villejuif, France
- Department of Biostatistics and Clinical Research, CHU Rouen, 76000 Rouen, France
- Université de Rouen-Normandie, Rouen, France
| | - Lucien Abenhaim
- The PGRx Study Group, Paris, France
- RESAL, LA-SER Group, Paris, France
- Department of Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- Centre for Risk Research Inc., Montreal, Canada
| |
Collapse
|
11
|
de Almeida SM, Beltrame MP, Tang B, Rotta I, Justus JLP, Schluga Y, da Rocha MT, Martins E, Liao A, Abramson I, Vaida F, Schrier R, Ellis RJ. CD3 +CD56 + and CD3 -CD56 + lymphocytes in the cerebrospinal fluid of persons with HIV-1 subtypes B and C. J Neuroimmunol 2023; 377:578067. [PMID: 36965365 PMCID: PMC10817703 DOI: 10.1016/j.jneuroim.2023.578067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/19/2023]
Abstract
The transactivator of transcription (Tat) is a HIV regulatory protein which promotes viral replication and chemotaxis. HIV-1 shows extensive genetic diversity, HIV-1 subtype C being the most dominant subtype in the world. Our hypothesis is the frequency of CSF CD3+CD56+ and CD3-CD56dim is reduced in HIV-1C compared to HIV-1B due to the Tat C30S31 substitution in HIV-1C. 34 CSF and paired blood samples (PWH, n = 20; PWoH, n = 14) were studied. In PWH, the percentage of CD3+CD56+ was higher in CSF than in blood (p < 0.001), comparable in both compartments in PWoH (p = 0.20). The proportion of CD3-CD56dim in CSF in PWH was higher than PWoH (p = 0.008). There was no subtype differences. These results showed CNS compartmentalization of NKT cell response in PWH.
Collapse
Affiliation(s)
- Sergio M de Almeida
- Virology Laboratory, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, PR, Brazil.
| | | | - Bin Tang
- HIV Neurobehavioral Research Center (HNRC), UCSD, San Diego, CA, USA
| | - Indianara Rotta
- Virology Laboratory, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Julie Lilian P Justus
- Immunophenotyping Laboratory, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Yara Schluga
- Immunophenotyping Laboratory, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Maria Tadeu da Rocha
- Immunophenotyping Laboratory, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Edna Martins
- Immunophenotyping Laboratory, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Antony Liao
- HIV Neurobehavioral Research Center (HNRC), UCSD, San Diego, CA, USA
| | - Ian Abramson
- HIV Neurobehavioral Research Center (HNRC), UCSD, San Diego, CA, USA
| | - Florin Vaida
- HIV Neurobehavioral Research Center (HNRC), UCSD, San Diego, CA, USA
| | - Rachel Schrier
- HIV Neurobehavioral Research Center (HNRC), UCSD, San Diego, CA, USA
| | - Ronald J Ellis
- HIV Neurobehavioral Research Center (HNRC), UCSD, San Diego, CA, USA
| |
Collapse
|
12
|
Tian J, Jiang L, Chen Z, Yuan Q, Liu C, He L, Jiang F, Rui K. Tissue-resident immune cells in the pathogenesis of multiple sclerosis. Inflamm Res 2023; 72:363-372. [PMID: 36547688 DOI: 10.1007/s00011-022-01677-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system (CNS) in which genetic and environmental factors contribute to disease progression. Both innate and adaptive immune cells, including T cells, B cells, activated macrophages and microglia, have been identified to be involved in the pathogenesis of MS, leading to the CNS inflammation, neurodegeneration and demyelination. In recent years, there has been considerable progress in understanding the contribution of tissue-resident immune cells in the pathogenesis of MS. METHODS We performed a keyword-based search in PubMed database. We combined "multiple sclerosis" with keywords, such as tissue-resident memory T cells, microglia to search for relevant literatures in PubMed. RESULTS AND CONCLUSION In this review, we comprehensively describe the characteristics of tissue-resident memory T cells and microglia, summarize their role in the pathogenesis of MS, and discuss their interaction with other immune cells in the CNS.
Collapse
Affiliation(s)
- Jie Tian
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
- Department of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Lingli Jiang
- Department of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zixiang Chen
- Department of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qingfang Yuan
- Department of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chang Liu
- Department of Laboratory Medicine, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Longfeng He
- Department of Obstetrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Feng Jiang
- Department of Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China.
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
13
|
Xiao X, Chu SJ, Tang JH, Zhang LY, Zhang BB. Leukoencephalopathy in children with acute lymphoblastic leukemia after chemotherapy: a retrospective monocenter study. Transl Cancer Res 2023; 12:340-350. [PMID: 36915585 PMCID: PMC10007887 DOI: 10.21037/tcr-22-2180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/06/2022] [Indexed: 01/10/2023]
Abstract
Background To investigate the clinical and neuroimaging characteristics of leukoencephalopathy among children with acute lymphoblastic leukemia (ALL), especially after chemotherapy. Methods Clinical data for 17 pediatric patients with leukoencephalopathy and 17 matched controls were retrospectively analyzed. All participants were children with ALL admitted to the Children's Hospital of Soochow University from May 2011 to April 2021. The data mainly consisted of general information, laboratory studies, and imaging diagnostic results. Results Overall, 94.12% of the patients experienced neurological symptoms. The most common symptoms were seizure (7/17, 41.18%), nausea (5/17, 29.41%), vomiting (5/17, 29.41%), paralysis (5/17, 29.41%), and numbness (4/17, 23.53%). On neuroimaging, multiple and irregular lesions were observed, distributed mainly in the periventricular area (9/17, 52.94%), parietal lobe (6/17, 35.29%), and basal ganglia (5/17, 29.41%). Moreover, there were significant differences in serum sodium (P=0.0001), C-reactive protein (P=0.0124) and blood pressure (P=0.0271) between patients with and without leukoencephalopathy. After aggressive treatment, the clinical symptoms (12/17, 70.59%) and imaging lesions (11/13, 84.62%) gradually improved in most patients. Conclusions Chemotherapy is an important risk factor related to leukoencephalopathy. Although the clinical symptoms of leukoencephalopathy vary widely, there is a high degree of consistency in its radiological features. Abnormal laboratory results may also help the identification of leukoencephalopathy. Early detection and treatment can improve brain development in the long term.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Si-Jia Chu
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Ji-Hong Tang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| | - Li-Ya Zhang
- Department of Internal Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Bing-Bing Zhang
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Erdoğan T, Koçer B, Şen S, Balcı BP, Terzi M. Newly Diagnosed Tumefactive Demyelinating Lesion and Multiple Sclerosis After COVID-19 Infection. Noro Psikiyatr Ars 2023; 60:223-230. [PMID: 37645083 PMCID: PMC10461761 DOI: 10.29399/npa.28142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 08/18/2022] [Indexed: 08/31/2023] Open
Abstract
Introduction To describe the parainfectious or postinfectious effects of COVID-19 infection on the first demyelinating presentation of Multiple Sclerosis and tumefactive demyelinating lesion (TDL) developing with Longitudinally Extensive Transverse Myelitis (LETM). Methods We present six patients who presented with a first CNS demyelination event or whose demyelinating lesions had aggravated after COVID-19 infection between May and December 2020. Nasopharyngeal swab SARS-CoV-2 PCR positivity was detected in five cases and cerebrospinal fluid (CSF) PCR was positive in one. The symptoms, neurological signs, radiological and CSF findings of the cases were examined. Results A 24-year-old woman presented with LETM aggravated by COVID-19, accompanied by a newly developed open-ring enhanced TDL. Four patients were diagnosed with the first presentation of MS, and one presented with clinically isolated syndrome according to the McDonald 2017 criteria. The interval between SARS-CoV-2 infection and the onset of clinical symptoms ranged from 4-93 days. All of the cases present with pyramidal or brain stem findings and have high brain and/or spinal MRI load. This suggests the moderate activity of CNS demyelinating disease after COVID-19 infection. Conclusions Based on this case series, all these first demyelinating events suggested that COVID-19 infection might trigger or exacerbate CNS demyelinating disease. SARS-CoV-2 plays a role in the clinical onset of Multiple Sclerosis. Active delayed demyelination developed within the first three months. This can be explained by COVID-triggered neuroimmune response that had been latent, and the initiation of the active disease process began with triggering or aggravation of the lesions in MRI. Multiple Sclerosis should be maintained during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Tuğba Erdoğan
- Gazi University School of Medicine, Department of Neurology, Ankara, Turkey
| | - Belgin Koçer
- Gazi University School of Medicine, Department of Neurology, Ankara, Turkey
| | - Sedat Şen
- Ondokuz Mayıs University School of Medicine, Department of Neurology, Samsun, Turkey
| | - Belgin Petek Balcı
- İstanbul Hamidiye Faculty of Medicine, University of Health Sciences, Department of Neurology, İstanbul, Turkey
| | - Murat Terzi
- Ondokuz Mayıs University School of Medicine, Department of Neurology, Samsun, Turkey
| |
Collapse
|
15
|
Conklin B, Conley BM, Hou Y, Chen M, Lee KB. Advanced theragnostics for the central nervous system (CNS) and neurological disorders using functional inorganic nanomaterials. Adv Drug Deliv Rev 2023; 192:114636. [PMID: 36481291 DOI: 10.1016/j.addr.2022.114636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/13/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Various types of inorganic nanomaterials are capable of diagnostic biomarker detection and the therapeutic delivery of a disease or inflammatory modulating agent. Those multi-functional nanomaterials have been utilized to treat neurodegenerative diseases and central nervous system (CNS) injuries in an effective and personalized manner. Even though many nanomaterials can deliver a payload and detect a biomarker of interest, only a few studies have yet to fully utilize this combined strategy to its full potential. Combining a nanomaterial's ability to facilitate targeted delivery, promote cellular proliferation and differentiation, and carry a large amount of material with various sensing approaches makes it possible to diagnose a patient selectively and sensitively while offering preventative measures or early disease-modifying strategies. By tuning the properties of an inorganic nanomaterial, the dimensionality, hydrophilicity, size, charge, shape, surface chemistry, and many other chemical and physical parameters, different types of cells in the central nervous system can be monitored, modulated, or further studies to elucidate underlying disease mechanisms. Scientists and clinicians have better understood the underlying processes of pathologies for many neurologically related diseases and injuries by implementing multi-dimensional 0D, 1D, and 2D theragnostic nanomaterials. The incorporation of nanomaterials has allowed scientists to better understand how to detect and treat these conditions at an early stage. To this end, having the multi-modal ability to both sense and treat ailments of the central nervous system can lead to favorable outcomes for patients suffering from such injuries and diseases.
Collapse
Affiliation(s)
- Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Brian M Conley
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Meizi Chen
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123, Bevier Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
16
|
Montalban X, Wallace D, Genovese MC, Tomic D, Parsons-Rich D, Le Bolay C, Kao AH, Guehring H. Characterisation of the safety profile of evobrutinib in over 1000 patients from phase II clinical trials in multiple sclerosis, rheumatoid arthritis and systemic lupus erythematosus: an integrated safety analysis. J Neurol Neurosurg Psychiatry 2023; 94:1-9. [PMID: 36418156 PMCID: PMC9763187 DOI: 10.1136/jnnp-2022-328799] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Analyse the integrated safety profile of evobrutinib, a Bruton's tyrosine kinase inhibitor (BTKi), using pooled data from multiple sclerosis (MS), rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) trials. METHODS Phase II, randomised, double-blind, placebo-controlled trial data were analysed (N=1083; MS: n=213, 48 weeks (W); RA: n=390, 12W; SLE: n=480, 52W). The analysis included all patients who received ≥1 dose of evobrutinib (25 mg or 75 mg once daily, or 50 mg or 75 mgtwice daily) or placebo. Descriptive statistics and exposure-adjusted incidence rates (EAIR) were used to report treatment-emergent adverse events (TEAEs). RESULTS Data from 1083 patients were pooled: evobrutinib, n=861; placebo, n=271 (sum >1083 due to MS trial design: n=49 received both placebo (W0-24) and evobrutinib 25 mg (W25-48)); median follow-up time (pt-years): evobrutinib, 0.501; placebo, 0.463. Across indications, the proportion of patients with TEAEs and the EAIR were similar for evobrutinib and placebo (66.2% (247.6 events/100 pt-years) vs 62.4% (261.4 events/100 pt-years)). By indication, the EAIR (events/100 pt-years) of TEAEs for evobrutinib versus placebo were: MS: 119.7 vs 148.3; RA: 331.8 vs 306.8; SLE: 343.0 vs 302.1. Two fatal events occurred (in SLE). The serious infections EAIR was 2.7 and 2.1 events/100 pt-years for evobrutinib and placebo. For previously reported BTKi-class effects, the EAIR of transient elevated alanine aminotransferase/aspartate aminotransferase TEAEs (events/100 pt-years) with evobrutinib versus placebo was 4.8 vs 2.8/3.5 vs 0.7, respectively. IgG levels were similar in evobrutinib/placebo-treated patients. CONCLUSIONS This is the first BTKi-integrated safety analysis that includes patients with MS. Overall, evobrutinib treatment (all doses) was generally well tolerated across indications. TRIAL REGISTRATION NUMBERS NCT02975349, NCT03233230, NCT02975336.
Collapse
Affiliation(s)
- Xavier Montalban
- Department of Neurology-Neuroimmunology, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Daniel Wallace
- Cedars-Sinai Medical Center, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Mark C Genovese
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, California, USA
| | - Davorka Tomic
- Global Clinical Development, Ares Trading SA, Eysins, Switzerland, an affiliate of Merck KGaA
| | - Dana Parsons-Rich
- Global Clinical Development, EMD Serono Research & Development Institute, Inc, Billerica, Massachusetts, USA, an affiliate of Merck KGaA (affiliation at the time the research was conducted)
- ECD-Early Clinical Development, Pfizer, Cambridge, Massachusetts, USA
| | | | - Amy H Kao
- Translational Innovation Platform in Immunology & Neuroscience, EMD Serono Research & Development Institute, Inc, Billerica, Massachusetts, USA, an affiliate of Merck KGaA
| | - Hans Guehring
- Global Patient Safety, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
17
|
Immunotherapy as a New Therapeutic Approach for Brain and Spinal Cord Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1394:73-84. [PMID: 36587382 DOI: 10.1007/978-3-031-14732-6_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Historically, the central nervous system (CNS) was considered an immune-privileged organ. However, recent studies have shown that the immune system plays a significant role in the CNS. Thus, there is renewed interest in applying cancer immunotherapy to CNS malignancies with the hope of generating a robust anti-tumor immune response and creating long-lasting immunity in patients. There has been some work with non-specific immunotherapy such as IL-2 for brain metastasis. Unfortunately, the results from non-specific immunotherapy studies were lackluster, so the focus has shifted to more specific CNS immunotherapies including cancer vaccines, immune checkpoint inhibitors, oncolytic virus therapy, and chimeric antigen receptor (CAR) T cell therapy. With respect to cancer vaccines, rindopepimut has been well-studied in glioblastoma (GBM) patients with the EGFRvIII mutation, with early results from phase II trials showing possible efficacy in carefully selected GBM patients. Other antigen-specific CNS tumor vaccines are still in the early stages. Immune checkpoint inhibitors are amongst the most promising and widely studied CNS immunotherapy strategies. Anti-PD-1 showed promising results in many non-CNS solid tumors, however, results from early clinical trials show poor efficacy for anti-PD-1 in GBM patients. Anti-PD-1 is also under investigation for CNS metastasis and showed some efficacy in non-small cell lung cancer and renal cell carcinoma patients. Anti-PD-1 is under early stage investigation for other CNS tumors such as chordoma. Oncolytic virus therapy is the strategy of infecting tumor cells with a virus that in turn triggers an innate immune response leading to tumor cell lysis. Oncolytic viruses currently under investigation include several adenovirus-based therapies and a herpes simplex virus-based therapy. Phase I studies have demonstrated the safety of oncolytic virus therapies in GBM patients. Current studies are evaluating the efficacy of these therapies both alone and in combination with other immunotherapy approaches such as checkpoint inhibition in patients with CNS tumors. CAR T cell therapy is a newer immunotherapy approach. CAR T cell therapies, directed against EGFRvIII mutation and HER-2 mutation, demonstrate an acceptable safety profile, although there is no conclusive evidence of the survival benefit of these therapies in early trials. Studies are currently underway to determine optimal tumor-specific antigen selection and modality of administration for CAR T cell therapy. Overall, the prognosis is generally poor for patients with CNS malignancies. The promising results of cancer immunotherapy for non-CNS tumors have created significant interest in applying these therapies for CNS malignancies. Preliminary results have not demonstrated robust efficacy for CNS immunotherapy. However, it is important to keep in mind that the field is still in its infancy and many clinical trials are still early-phase. Several, clinical trials are currently underway to further explore the role of immunotherapy for CNS malignancies.
Collapse
|
18
|
He J, Shen X, Fu D, Yang Y, Xiong K, Zhao L, Xie H, Pelekos G, Li Y. Human periodontitis-associated salivary microbiome affects the immune response of diabetic mice. J Oral Microbiol 2022; 14:2107814. [PMID: 35958276 PMCID: PMC9359160 DOI: 10.1080/20002297.2022.2107814] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background The bidirectional association between periodontitis and diabetes mellitus has been well accepted; however, pathways connecting them remain unclear. Some oral bacteria are able to induce immunologic changes favoring insulin resistance individually. However, it is unclear if and how the systemic immune system responds to a disturbed oral microbial community in diabetic sufferers. Aim This study aimed to investigate the impact of the human periodontitis-associated salivary microbiome on the splenic immune responses of diabetic mice. Methods An in vivo diabetic animal model was established by feeding high fat food. After microbial depletion with quadruple antibiotic treatment, human saliva from healthy and periodontitis volunteers was transplanted into the mouth of these diabetic mice (N = 3), respectively. Results Osteoclasts and expression levels of TNF-α and IL-1β were significantly increased in periodontal tissues of mice receiving periodontitis patients donated microbiome compared to these transplanted with healthy subjects donated microbiome. The proportion of monocyte (an innate immunocyte) decreased in mice receiving periodontitis patients donated microbiome. However, the abundance of an adaptive immunocyte Th17 was up-regulated. The IL17 production of ILC3 cells in human periodontitis-associated salivary microbiome recipient mice was significantly impaired. Conclusions A disturbed oral microbiome imposes a stress on the splenic immune responses of diabetic mice.
Collapse
Affiliation(s)
- Jinzhi He
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xin Shen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
| | - Di Fu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
| | - Yutao Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
| | - Kaixin Xiong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
| | - Huixu Xie
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
| | - Georege Pelekos
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Yan Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan, China
| |
Collapse
|
19
|
McPherson SW, Heuss ND, Abedin M, Roehrich H, Pierson MJ, Gregerson DS. Parabiosis reveals the correlation between the recruitment of circulating antigen presenting cells to the retina and the induction of spontaneous autoimmune uveoretinitis. J Neuroinflammation 2022; 19:295. [PMID: 36494807 PMCID: PMC9733026 DOI: 10.1186/s12974-022-02660-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Characterizing immune cells and conditions that govern their recruitment and function in autoimmune diseases of the nervous system or in neurodegenerative processes is an area of active investigation. We sought to analyze the origin of antigen presenting cells associated with the induction of retinal autoimmunity using a system that relies on spontaneous autoimmunity, thus avoiding uncertainties associated with immunization with adjuvants at remotes sites or adoptive transfer of in vitro activated T cells. METHODS R161H mice (B10.RIII background), which spontaneously and rapidly develop severe spontaneous autoimmune uveoretinitis (SAU), were crossed to CD11cDTR/GFP mice (B6/J) allowing us to track the recruitment to and/or expansion within the retina of activated, antigen presenting cells (GFPhi cells) in R161H+/- × CD11cDTR/GFP F1 mice relative to the course of SAU. Parabiosis between R161H+/- × CD11cDTR/GFP F1 mice and B10.RIII × B6/J F1 (wild-type recipient) mice was done to explore the origin and phenotype of antigen presenting cells crucial for the induction of autoimmunity. Analysis was done by retinal imaging, flow cytometry, and histology. RESULTS Onset of SAU in R161H+/- × CD11cDTR/GFP F1 mice was delayed relative to B10.RIII-R161H+/- mice revealing a disease prophase prior to frank autoimmunity that was characterized by expansion of GFPhi cells within the retina prior to any clinical or histological evidence of autoimmunity. Parabiosis between mice carrying the R161H and CD11cDTR/GFP transgenes and transgene negative recipients showed that recruitment of circulating GFPhi cells into retinas was highly correlative with the occurrence of SAU. CONCLUSIONS Our results here contrast with our previous findings showing that retinal antigen presenting cells expanding in response to either sterile mechanical injury or neurodegeneration were derived from myeloid cells within the retina or optic nerve, thus highlighting a unique facet of retinal autoimmunity.
Collapse
Affiliation(s)
- Scott W. McPherson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Neal D. Heuss
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Md. Abedin
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Heidi Roehrich
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Mark J. Pierson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| | - Dale S. Gregerson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, Lions Research Building, Room 482A, Minneapolis, MN 55455 USA
| |
Collapse
|
20
|
Cabezas Perez RJ, Ávila Rodríguez MF, Rosero Salazar DH. Exogenous Antioxidants in Remyelination and Skeletal Muscle Recovery. Biomedicines 2022; 10:biomedicines10102557. [PMID: 36289819 PMCID: PMC9599955 DOI: 10.3390/biomedicines10102557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory, oxidative, and autoimmune responses cause severe damage to the nervous system inducing loss of myelin layers or demyelination. Even though demyelination is not considered a direct cause of skeletal muscle disease there is extensive damage in skeletal muscles following demyelination and impaired innervation. In vitro and in vivo evidence using exogenous antioxidants in models of demyelination is showing improvements in myelin formation alongside skeletal muscle recovery. For instance, exogenous antioxidants such as EGCG stimulate nerve structure maintenance, activation of glial cells, and reduction of oxidative stress. Consequently, this evidence is also showing structural and functional recovery of impaired skeletal muscles due to demyelination. Exogenous antioxidants mostly target inflammatory pathways and stimulate remyelinating mechanisms that seem to induce skeletal muscle regeneration. Therefore, the aim of this review is to describe recent evidence related to the molecular mechanisms in nerve and skeletal muscle regeneration induced by exogenous antioxidants. This will be relevant to identifying further targets to improve treatments of neuromuscular demyelinating diseases.
Collapse
|
21
|
Zhang S, Mao C, Li X, Miao W, Teng J. Advances in Potential Cerebrospinal Fluid Biomarkers for Autoimmune Encephalitis: A Review. Front Neurol 2022; 13:746653. [PMID: 35937071 PMCID: PMC9355282 DOI: 10.3389/fneur.2022.746653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Autoimmune encephalitis (AE) is a severe inflammatory disease of the brain. Patients with AE demonstrate amnesia, seizures, and psychosis. Recent studies have identified numerous associated autoantibodies (e.g., against NMDA receptors (NMDARs), LGI1, etc.) involved in the pathogenesis of AE, and the levels of diagnosis and treatment are thus improved dramatically. However, there are drawbacks of clinical diagnosis and treatment based solely on antibody levels, and thus the application of additional biomarkers is urgently needed. Considering the important role of immune mechanisms in AE development, we summarize the relevant research progress in identifying cerebrospinal fluid (CSF) biomarkers with a focus on cytokines/chemokines, demyelination, and nerve damage.
Collapse
|
22
|
Hadjiagapiou MS, Krashias G, Deeba E, Christodoulou C, Pantzaris M, Lambrianides A. Antibodies to blood coagulation components are implicated in patients with multiple sclerosis. Mult Scler Relat Disord 2022; 62:103775. [DOI: 10.1016/j.msard.2022.103775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
|
23
|
Barati S, Kashani IR, Tahmasebi F. The effects of mesenchymal stem cells transplantation on A1 neurotoxic reactive astrocyte and demyelination in the cuprizone model. J Mol Histol 2022; 53:333-346. [PMID: 35031895 DOI: 10.1007/s10735-021-10046-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS), which is an autoimmune disease, is characterized by symptoms such as demyelination, axonal damage, and astrogliosis. As the most abundant type of glial cells, astrocytes play an important role in MS pathogenesis. Mesenchymal stem cells (MSCs) are a subset of stromal cells that have the potential for migration, immune-modulation, differentiation, remyelination, and neuroregeneration. Therefore, the present study evaluates the effects of MSC transplantation on A1 reactive astrocytes and the remyelination process in the cuprizone mouse model. The study used 30 male C57BL/6 mice, which were randomly distributed into three subgroups (n = 10), i.e., control, cuprizone, and transplanted MSCs groups. In order to generate a chronic demyelination model, the mice in the cuprizone group received food mixed with 0.2% cuprizone powder for 12 weeks. Then, 2 μl of DMEM containing approximately 3 × 105 DiI labeled cells was injected with a 4-min interval into the right lateral ventricle using a 10-μl Hamilton syringe. After 2 weeks of cell transplantation, we used the rotarod test to evaluate the behavioral deficits, while the remyelination process was assessed by transmission electron microscopy (TEM) and Luxol Fast Blue (LFB) staining. We assessed the population of A1 astrocytes and oligodendrocytes using specific markers, such as C3, GFAP, and Olig2, using the immunefleurocent method. The pro-inflammatory and trophic factors were assessed by a real-time polymerase chain reaction. According to our data, the specific marker of A1 astrocytes (C3) decreased in the MSCs group, while the number of oligodendrocytes significantly increased in this group compared to the cuprizone mice. Additionally, MSC was able to enhance the remyelination process after cuprizone usage, as shown by LFB and TEM images. The molecular results showed that MSCs could reduce pro-inflammatory factors, such as IL-1 and TNF-α, through the secretion of BDNF and TGF-β as trophic factors. The obtained results indicated that MSC could reduce demyelination and inflammation by decreasing A1 neurotoxic reactive astrocytes and neurotrophic and immunomodulatory factors secretion in the chronic cuprizone demyelination model.
Collapse
Affiliation(s)
- Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
24
|
The Effects of Modified Curcumin Preparations on Glial Morphology in Aging and Neuroinflammation. Neurochem Res 2022; 47:813-824. [PMID: 34988899 DOI: 10.1007/s11064-021-03499-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is characterized by reactive microglia and astrocytes (collectively called gliosis) in the central nervous system and is considered as one of the main pathological hallmarks in different neurodegenerative diseases such as Alzheimer's disease, age-related dementia, and multiple sclerosis. Upon activation, glia undergoes structural and morphological changes such as the microglial cells swell in size and astrocytes become bushy, which play both beneficial and detrimental roles. Hence, they are unable to perform the normal physiological role in brain immunity. Curcumin, a cytokine suppressive anti-inflammatory drug, has a high proven pre-clinical potency and efficacy to reverse chronic neuroinflammation by attenuating the activation and morphological changes that occur in the microglia and astrocytes. This review will highlight the recent findings on the tree structure changes of microglia and astrocytes in neuroinflammation and the effects of curcumin against the activation and morphology of glial cells.
Collapse
|
25
|
Huang S, Wu T, Lau AY, Au C, Huang H, Wang X, Kim JY. Attention to time-of-day variability improves the reproducibility of gene expression patterns in multiple sclerosis. iScience 2021; 24:103247. [PMID: 34746708 PMCID: PMC8551071 DOI: 10.1016/j.isci.2021.103247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 08/30/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022] Open
Abstract
Low reproducibility in gene expression profiles has been observed in transcriptome studies, and this often limits applying findings to clinical practice. Here, we show time-of-day effects on gene expression and analytical schemes to increase the reproducibility in expression patterns. We recruited patients with relapsing-remitting multiple sclerosis (RRMS) and healthy subjects and collected blood from individuals twice a day, day (2 pm) and night (9 pm). RNA sequencing analyses found that gene expression in RRMS in relapse (Relapse) is significantly changed at night compared with either Relapse at day or RRMS in remission (Remission). Gene set overrepresentation analysis demonstrated that gene sets significantly changed in Relapse at night are enriched to immune responses related to MS pathology. In those gene sets, 68 genes are significantly changed expression in Relapse at night compared with Relapse at day and Remission. This supports that times of sample collections should be standardized to obtain reproducible gene expression patterns. Times of day affect gene expression patterns in patients with RRMS in relapse Transcriptome profiles in Relapse are changed from day to night In Relapse, immune response-related genes change the expression at night
Collapse
Affiliation(s)
- Suihong Huang
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Tan Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Alexander Y Lau
- Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Cheryl Au
- Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hao Huang
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Jin Young Kim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
26
|
Satheesh NJ, Salloum-Asfar S, Abdulla SA. The Potential Role of COVID-19 in the Pathogenesis of Multiple Sclerosis-A Preliminary Report. Viruses 2021; 13:2091. [PMID: 34696521 PMCID: PMC8540806 DOI: 10.3390/v13102091] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/17/2022] Open
Abstract
Coronavirus 2019 (COVID-19) is an infectious respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that mainly affects the lungs. COVID-19 symptoms include the presence of fevers, dry coughs, fatigue, sore throat, headaches, diarrhea, and a loss of taste or smell. However, it is understood that SARS-CoV-2 is neurotoxic and neuro-invasive and could enter the central nervous system (CNS) via the hematogenous route or via the peripheral nerve route and causes encephalitis, encephalopathy, and acute disseminated encephalomyelitis (ADEM) in COVID-19 patients. This review discusses the possibility of SARS-CoV-2-mediated Multiple Sclerosis (MS) development in the future, comparable to the surge in Parkinson's disease cases following the Spanish Flu in 1918. Moreover, the SARS-CoV-2 infection is associated with a cytokine storm. This review highlights the impact of these modulated cytokines on glial cell interactions within the CNS and their role in potentially prompting MS development as a secondary disease by SARS-CoV-2. SARS-CoV-2 is neurotropic and could interfere with various functions of neurons leading to MS development. The influence of neuroinflammation, microglia phagocytotic capabilities, as well as hypoxia-mediated mitochondrial dysfunction and neurodegeneration, are mechanisms that may ultimately trigger MS development.
Collapse
Affiliation(s)
| | - Salam Salloum-Asfar
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar;
| | - Sara A. Abdulla
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar;
| |
Collapse
|
27
|
Zhang J, Xu X, Liu H, Jin L, Shen X, Xie C, Xiang W, Yang D, Feng W, Wang J, Wang M, Dong T, Qiu H, Wu L, Wang Y, Zhang X, Huang Z. Astrocytic YAP prevents the demyelination through promoting expression of cholesterol synthesis genes in experimental autoimmune encephalomyelitis. Cell Death Dis 2021; 12:907. [PMID: 34611127 PMCID: PMC8492624 DOI: 10.1038/s41419-021-04203-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/13/2021] [Accepted: 09/22/2021] [Indexed: 01/20/2023]
Abstract
Cholesterols are the main components of myelin, and are mainly synthesized in astrocytes and transported to oligodendrocytes and neurons in the adult brain. It has been reported that Hippo/yes-associated protein (YAP) pathways are involved in cholesterol synthesis in the liver, however, it remains unknown whether YAP signaling can prevent the demyelination through promoting cholesterol synthesis in experimental autoimmune encephalomyelitis (EAE), a commonly used animal model of multiple sclerosis characterized by neuroinflammation and demyelination. Here, we found that YAP was upregulated and activated in astrocytes of spinal cords of EAE mice through suppression of the Hippo pathway. YAP deletion in astrocytes aggravated EAE with earlier onset, severer inflammatory infiltration, demyelination, and more loss of neurons. Furthermore, we found that the neuroinflammation was aggravated and the proliferation of astrocytes was decreased in YAPGFAP-CKO EAE mice. Mechanically, RNA-seq revealed that the expression of cholesterol-synthesis pathway genes such as HMGCS1 were decreased in YAP-/- astrocytes. qPCR, western blot, and immunostaining further confirmed the more significant reduction of HMGCS1 in spinal cord astrocytes of YAPGFAP-CKO EAE mice. Interestingly, upregulation of cholesterol-synthesis pathways by diarylpropionitrile (DPN) (an ERβ-ligand, to upregulate the expression of HMGCS1) treatment partially rescued the demyelination deficits in YAPGFAP-CKO EAE mice. Finally, activation of YAP by XMU-MP-1 treatment promoted the expression of HMGCS1 in astrocytes and partially rescued the demyelination and inflammatory infiltration deficits in EAE mice. These findings identify unrecognized functions of astrocytic YAP in the prevention of demyelination through promoting cholesterol synthesis in EAE, and reveal a novel pathway of YAP/HMGCS1 for cholesterol synthesis in EAE pathology.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Body Weight
- Cell Proliferation
- Cholesterol/biosynthesis
- Demyelinating Diseases/genetics
- Down-Regulation/genetics
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Gene Expression Regulation
- Hippo Signaling Pathway
- Inflammation/pathology
- Mice, Knockout
- Models, Biological
- Neurons/metabolism
- Neurons/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recovery of Function
- Spinal Cord/pathology
- Spinal Cord/ultrastructure
- Up-Regulation/genetics
- YAP-Signaling Proteins/deficiency
- YAP-Signaling Proteins/metabolism
- Mice
Collapse
Affiliation(s)
- Jingjing Zhang
- School of Pharmacy, and Department of Neurosurgery of the Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, China
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xingxing Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huitao Liu
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingting Jin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiya Shen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Changnan Xie
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiwei Xiang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Danlu Yang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjin Feng
- Zhejiang Sinogen Medical Equipment Co., Ltd., Wenzhou, Zhejiang, China
| | - Jiaojiao Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mianxian Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tianyingying Dong
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haoyu Qiu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lihao Wu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Wang
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Xu Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Zhihui Huang
- School of Pharmacy, and Department of Neurosurgery of the Affiliated Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, China.
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
28
|
Goldfarb S, Fainstein N, Ganz T, Vershkov D, Lachish M, Ben-Hur T. Electric neurostimulation regulates microglial activation via retinoic acid receptor α signaling. Brain Behav Immun 2021; 96:40-53. [PMID: 33989746 DOI: 10.1016/j.bbi.2021.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/20/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Brain stimulation by electroconvulsive therapy is effective in neuropsychiatric disorders by unknown mechanisms. Microglial toxicity plays key role in neuropsychiatric, neuroinflammatory and degenerative diseases. We examined the mechanism by which electroconvulsive seizures (ECS) regulates microglial phenotype and response to stimuli. Microglial responses were examined by morphological analysis, Iba1 and cytokine expression. ECS did not affect resting microglial phenotype or morphology but regulated their activation by Lipopolysaccharide stimulation. Microglia were isolated after ECS or sham sessions in naïve mice for transcriptome analysis. RNA sequencing identified 141 differentially expressed genes. ECS modulated multiple immune-associated gene families and attenuated neurotoxicity-associated gene expression. Blood brain barrier was examined by injecting Biocytin-TMR tracer. There was no breakdown of the BBB, nor increase in gene-signature of peripheral monocytes, suggesting that ECS effect is mainly on resident microglia. Unbiased analysis of regulatory sequences identified the induction of microglial retinoic acid receptor α (RARα) gene expression and a putative common RARα-binding motif in multiple ECS-upregulated genes. The effects of AM580, a selective RARα agonist on microglial response to LPS was examined in vitro. AM580 prevented LPS-induced cytokine expression and reactive oxygen species production. Chronic murine experimental autoimmune encephalomyelitis (EAE) was utilized to confirm the role RARα signaling as mediator of ECS-induced transcriptional pathway in regulating microglial toxicity. Continuous intracerebroventricular delivery of AM580 attenuated effectively EAE severity. In conclusion, ECS regulates CNS innate immune system responses by activating microglial retinoic acid receptor α pathway, signifying a novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Dan Vershkov
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel; The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
29
|
Dieu RS, Wais V, Sørensen MZ, Marczynska J, Dubik M, Kavan S, Thomassen M, Burton M, Kruse T, Khorooshi R, Owens T. Central Nervous System-Endogenous TLR7 and TLR9 Induce Different Immune Responses and Effects on Experimental Autoimmune Encephalomyelitis. Front Neurosci 2021; 15:685645. [PMID: 34211367 PMCID: PMC8241214 DOI: 10.3389/fnins.2021.685645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/19/2021] [Indexed: 11/13/2022] Open
Abstract
Innate receptors, including Toll like receptors (TLRs), are implicated in pathogenesis of CNS inflammatory diseases such as multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). TLR response to pathogens or endogenous signals includes production of immunoregulatory mediators. One of these, interferon (IFN)β, a Type I IFN, plays a protective role in MS and EAE. We have previously shown that intrathecal administration of selected TLR ligands induced IFNβ and infiltration of blood-derived myeloid cells into the central nervous system (CNS), and suppressed EAE in mice. We have now extended these studies to evaluate a potential therapeutic role for CNS-endogenous TLR7 and TLR9. Intrathecal application of Imiquimod (TLR7 ligand) or CpG oligonucleotide (TLR9 ligand) into CNS of otherwise unmanipulated mice induced IFNβ expression, with greater magnitude in response to CpG. CD45+ cells in the meninges were identified as source of IFNβ. Intrathecal CpG induced infiltration of monocytes, neutrophils, CD4+ T cells and NK cells whereas Imiquimod did not recruit blood-derived CD45+ cells. CpG, but not Imiquimod, had a beneficial effect on EAE, when given at time of disease onset. This therapeutic effect of CpG on EAE was not seen in mice lacking the Type I IFN receptor. In mice with EAE treated with CpG, the proportion of monocytes was significantly increased in the CNS. Infiltrating cells were predominantly localized to spinal cord meninges and demyelination was significantly reduced compared to non-treated mice with EAE. Our findings show that TLR7 and TLR9 signaling induce distinct inflammatory responses in the CNS with different outcome in EAE and point to recruitment of blood-derived cells and IFNβ induction as possible mechanistic links between TLR9 stimulation and amelioration of EAE. The protective role of TLR9 signaling in the CNS may have application in treatment of diseases such as MS.
Collapse
Affiliation(s)
- Ruthe Storgaard Dieu
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Vian Wais
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Michael Zaucha Sørensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Joanna Marczynska
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Magdalena Dubik
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Stephanie Kavan
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mark Burton
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Torben Kruse
- Department of Clinical Genetics, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
30
|
Huff TC, Sant DW, Camarena V, Van Booven D, Andrade NS, Mustafi S, Monje PV, Wang G. Vitamin C regulates Schwann cell myelination by promoting DNA demethylation of pro-myelinating genes. J Neurochem 2021; 157:1759-1773. [PMID: 32219848 PMCID: PMC7530063 DOI: 10.1111/jnc.15015] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022]
Abstract
Ascorbic acid (vitamin C) is critical for Schwann cells to myelinate peripheral nerve axons during development and remyelination after injury. However, its exact mechanism remains elusive. Vitamin C is a dietary nutrient that was recently discovered to promote active DNA demethylation. Schwann cell myelination is characterized by global DNA demethylation in vivo and may therefore be regulated by vitamin C. We found that vitamin C induces a massive transcriptomic shift (n = 3,848 genes) in primary cultured Schwann cells while simultaneously producing a global increase in genomic 5-hydroxymethylcytosine (5hmC), a DNA demethylation intermediate which regulates transcription. Vitamin C up-regulates 10 pro-myelinating genes which exhibit elevated 5hmC content in both the promoter and gene body regions of these loci following treatment. Using a mouse model of human vitamin C metabolism, we found that maternal dietary vitamin C deficiency causes peripheral nerve hypomyelination throughout early development in resulting offspring. Additionally, dietary vitamin C intake regulates the expression of myelin-related proteins such as periaxin (PRX) and myelin basic protein (MBP) during development and remyelination after injury in mice. Taken together, these results suggest that vitamin C cooperatively promotes myelination through 1) increased DNA demethylation and transcription of pro-myelinating genes, and 2) its known role in stabilizing collagen helices to form the basal lamina that is necessary for myelination.
Collapse
Affiliation(s)
- Tyler C. Huff
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David W. Sant
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vladimir Camarena
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nadja S. Andrade
- Department of Psychiatry & Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sushmita Mustafi
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Paula V. Monje
- Department of Neurological Surgery, Indiana University, Indianapolis, IN, USA
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
31
|
Mrad MF, Saba ES, Nakib L, Khoury SJ. Exosomes From Subjects With Multiple Sclerosis Express EBV-Derived Proteins and Activate Monocyte-Derived Macrophages. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/4/e1004. [PMID: 34006621 PMCID: PMC8130999 DOI: 10.1212/nxi.0000000000001004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/26/2021] [Indexed: 12/19/2022]
Abstract
Objective To investigate in a cross-sectional study the effect of serum-derived exosomes on primary human blood monocyte-derived macrophages (MDMs) comparing exosomes from healthy donors vs patients with relapsing-remitting multiple sclerosis in remission and in relapse and to assess whether the response correlates with exosomal Epstein-Barr virus (EBV) protein expression. Methods A total of 45 serum-derived exosome preparations were isolated from patients and healthy controls and verified for the expression of exosomal and EBV markers. MDMs were differentiated from monocytes for 7 days and incubated for 24 hours with exosomes, and then, cell supernatants were collected for cytokine measurement by cytometric bead array. Cells were immunophenotyped before and after differentiation. Results Serum-derived exosomes of patients with multiple sclerosis (MS) expressed higher levels of EBV proteins than healthy controls. Of interest, expression of EBV nuclear antigen EBNA1 and latent membrane proteins LMP1 and 2A was higher on exosomes derived from patients with active RRMS compared with healthy controls and stable patients. After data normalization, we observed that incubation with EBV(+) exosomes induced CXCL10 and CCL2 secretion by MDMs. MDMs differentiated from patients with active disease were better secretors of CXCL10 and other interferon-γ–inducible chemokines, including CCL2 and CXCL9, than MDMs from healthy and stable MS groups. MDMs from active patients had a higher frequency of a CD14(++) subset that correlated with the secreted CXCL10. Conclusion Exosomes expressing EBV proteins correlate with disease activity and induce an inflammatory response in MDMs that is compounded by the origin of the responder cells.
Collapse
Affiliation(s)
- May F Mrad
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Esber S Saba
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Layane Nakib
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon
| | - Samia J Khoury
- From the Nehme and Therese Tohme Multiple Sclerosis Center (M.F.M.), Faculty of Medicine, American University of Beirut Medical Center; Department of Experimental Pathology (E.S.S., L.N.), Immunology and Microbiology, Faculty of Medicine, American University of Beirut; and Nehme and Therese Tohme Multiple Sclerosis Center (S.J.K.), and Abu Haidar Neuroscience Institute, Faculty of Medicine, American University of Beirut Medical Center, Lebanon.
| |
Collapse
|
32
|
Ghareghani M, Ghanbari A, Eid A, Shaito A, Mohamed W, Mondello S, Zibara K. Hormones in experimental autoimmune encephalomyelitis (EAE) animal models. Transl Neurosci 2021; 12:164-189. [PMID: 34046214 PMCID: PMC8134801 DOI: 10.1515/tnsci-2020-0169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/05/2021] [Accepted: 04/14/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) in which activated immune cells attack the CNS and cause inflammation and demyelination. While the etiology of MS is still largely unknown, the interaction between hormones and the immune system plays a role in disease progression, but the mechanisms by which this occurs are incompletely understood. Several in vitro and in vivo experimental, but also clinical studies, have addressed the possible role of the endocrine system in susceptibility and severity of autoimmune diseases. Although there are several demyelinating models, experimental autoimmune encephalomyelitis (EAE) is the oldest and most commonly used model for MS in laboratory animals which enables researchers to translate their findings from EAE into human. Evidences imply that there is great heterogeneity in the susceptibility to the induction, the method of induction, and the response to various immunological or pharmacological interventions, which led to conflicting results on the role of specific hormones in the EAE model. In this review, we address the role of endocrine system in EAE model to provide a comprehensive view and a better understanding of the interactions between the endocrine and the immune systems in various models of EAE, to open up a ground for further detailed studies in this field by considering and comparing the results and models used in previous studies.
Collapse
Affiliation(s)
- Majid Ghareghani
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Amir Ghanbari
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ali Eid
- Biomedical and Pharmaceutical Research Unit and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdullah Shaito
- Department of Biological and Chemical Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - Wael Mohamed
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Shibin Al Kawm, Egypt
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Pahang, Malaysia
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Kazem Zibara
- PRASE, Lebanese University, Beirut, Lebanon
- Biology Department, Faculty of Sciences – I, Lebanese University, Beirut, Lebanon
| |
Collapse
|
33
|
Ge F, Huo Z, Li C, Wang R, Wang R, Liu Y, Chen J, Lu Y, Wen Y, Jiang Y, Peng H, Wu X, Liang H, He J, Liang W. Lung cancer risk in patients with multiple sclerosis: a Mendelian randomization analysis. Mult Scler Relat Disord 2021; 51:102927. [PMID: 33812221 DOI: 10.1016/j.msard.2021.102927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The relationship of multiple sclerosis (MS) with lung cancer is under debate. Conventional observational studies have reported conflicting findings, but such studies are susceptible to confounding and reverse causation. With a Mendelian Randomization approach, we were able to evaluate the causality between MS and lung cancer. METHODS According to published genome-wide association studies (GWASs), we obtained 35 MS-related single-nucleotide polymorphisms, which were used as instrumental variables in our study. Summary data of individual-level genetic information were obtained from the International Lung Cancer Consortium (ILCCO), with a total of 15,861 controls and 11,348 cases; the latter is composed of patients with lung adenocarcinoma and squamous cell lung cancer. The inverse variance-weighted method was applied to estimate the causation between MS and lung cancer. To further evaluate the pleiotropy, the MR-Egger and Weighted median methods were implemented. RESULTS The results of MR analysis suggested a causal effect of MS on lung cancer incidence, with evidence of an increased risk for overall lung cancer [odds ratio (OR): 1.0648; 95% confidence interval (CI): 1.0163-1.1156; p = 0.0082]. However, subgroup analyses showed no significant causal relationships between MS and lung adenocarcinoma (OR = 1.0716; 95% CI 0.9840-1.1671, p = 0.1119) and squamous cell lung cancer (OR = 1.0284; 95% CI 0.9575-1.1045, p = 0.4424). In addition, no pleiotropy was found in our study. CONCLUSION Our study indicated that MS is a causal risk factor in the development of lung cancer. Further work is needed to elucidate the potential mechanisms.
Collapse
Affiliation(s)
- Fan Ge
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, First Clinical School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhenyu Huo
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Caichen Li
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Runchen Wang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rui Wang
- Department of Clinical Medicine, First Clinical School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yeling Liu
- Department of Clinical Medicine, Third Clinical School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiana Chen
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi Lu
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yaokai Wen
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu Jiang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Haoxin Peng
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiangrong Wu
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hengrui Liang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Department of Oncology, the First People's Hospital of Zhaoqing, Zhaoqing, 526000, China.
| |
Collapse
|
34
|
Sun Y, Ji J, Zha Z, Zhao H, Xue B, Jin L, Wang L. Effect and Mechanism of Catalpol on Remyelination via Regulation of the NOTCH1 Signaling Pathway. Front Pharmacol 2021; 12:628209. [PMID: 33708131 PMCID: PMC7940842 DOI: 10.3389/fphar.2021.628209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/21/2021] [Indexed: 11/13/2022] Open
Abstract
Promoting the differentiation of oligodendrocyte precursor cells (OPCs) is important for fostering remyelination in multiple sclerosis. Catalpol has the potential to promote remyelination and exert neuroprotective effects, but its specific mechanism is still unclear. Recent studies have shown that the NOTCH1 signaling pathway is involved in mediating OPC proliferation and differentiation. In this study, we elucidated that catalpol promoted OPC differentiation in vivo and vitro and explored the regulatory role of catalpol in specific biomolecular processes. Following catalpol administration, better and faster recovery of body weight and motor balance was observed in mice with cuprizone (CPZ)-induced demyelination. Luxol fast blue staining (LFB) and transmission electron microscopy (TEM) showed that catalpol increased the myelinated area and improved myelin ultrastructure in the corpus callosum in demyelinated mice. In addition, catalpol enhanced the expression of CNPase and MBP, indicating that it increased OPC differentiation. Additionally, catalpol downregulated the expression of NOTCH1 signaling pathway-related molecules, such as JAGGED1, NOTCH1, NICD1, RBPJ, HES5, and HES1. We further demonstrated that in vitro, catalpol enhanced the differentiation of OPCs into OLs and inhibited NOTCH1 signaling pathway activity. Our data suggested that catalpol may promote OPC differentiation and remyelination through modulation of the NOTCH1 pathway. This study provides new insight into the mechanism of action of catalpol in the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Yaqin Sun
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Jing Ji
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Zheng Zha
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Bing Xue
- Core Facility Center, Capital Medical University, Beijing, China
| | - Liangyun Jin
- Core Facility Center, Capital Medical University, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| |
Collapse
|
35
|
Liu K, Cai GL, Zhuang Z, Pei SY, Xu SN, Wang YN, Wang H, Wang X, Cui C, Sun MC, Guo SH, Jia KP, Wang XZ, Cai GF. Interleukin-1β-Treated Mesenchymal Stem Cells Inhibit Inflammation in Hippocampal Astrocytes Through Exosome-Activated Nrf-2 Signaling. Int J Nanomedicine 2021; 16:1423-1434. [PMID: 33654394 PMCID: PMC7910114 DOI: 10.2147/ijn.s289914] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interleukin-1β (IL-1)-treated mesenchymal stem cells (MSCs) and IL-1-MSCs-conditioned medium (CM) exert anti-inflammatory roles. Astrocytes are essential for the modulation of synaptic activity and neuronal homeostasis in the brain. Exosomes are the critical mediators in intercellular communication. However, the mechanism underlying the anti-inflammatory effect of IL-1-treated MSCs remains unknown. METHODS In this study, exosomes (IL-1-Exo) were isolated from IL-1-treated MSCs. In addition, lipopolysaccharide (LPS)-treated hippocampal astrocytes and status epilepticus (SE) mice were treated with IL-1-Exo. Inflammatory activity, astrogliosis, and cognitive performance were measured to determine the effect of IL-1-Exo on inflammation. RESULTS The results revealed that IL-1-Exo significantly inhibited LPS-induced astrogliosis and inflammatory responses of astrocytes. Also, IL-1-Exo reversed the LPS-induced effect on calcium signaling. The Nrf2 signaling pathway was associated with the effect of IL-1-Exo in LPS-treated astrocytes. Furthermore, IL-1-Exo reduced the inflammatory response and improved the cognitive performance of SE mice. CONCLUSION The results suggest that IL-1-Exo inhibited LPS-induced inflammatory responses in astrocytes and SE mice and that the effect of IL-1-Exo was primarily mediated through the Nrf-2 signaling pathway. This study provides a new understanding of the molecular mechanism of inflammation-associated brain diseases and an avenue to develop nanotherapeutic agents for the treatment of inflammatory conditions in the brain.
Collapse
Affiliation(s)
- Kai Liu
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Guo-Liang Cai
- Postdoctoral Research Workstation of Harbin Sport University, Harbin, 150001, People’s Republic of China
- Department of Sport Science and Health, Harbin Sport University, Harbin, 150008, People’s Republic of China
| | - Zhe Zhuang
- Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Si-Ying Pei
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Sheng-Nan Xu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Ya-Nan Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Hong Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Xin Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Cheng Cui
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Man-Chao Sun
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Si-Hui Guo
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Kun-Ping Jia
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Xiu-Zhen Wang
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| | - Guo-Feng Cai
- Hanan Branch of Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150001, People’s Republic of China
| |
Collapse
|
36
|
Association between familial Mediterranean fever and multiple sclerosis: A case series from the JIR cohort and systematic literature review. Mult Scler Relat Disord 2021; 50:102834. [PMID: 33609923 DOI: 10.1016/j.msard.2021.102834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/19/2021] [Accepted: 02/05/2021] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Familial Mediterranean fever (FMF) is the most frequent monogenic autoinflammatory disorder; and leads to the uncontrolled production of interleukin (IL)-1β. Multiple sclerosis (MS) is an inflammatory disease of the central nervous system; and its development seems to be partly correlated with IL-1β levels. It is hypothesized that FMF could be associated with MS. We aim to describe the features of patients displaying both diseases and to investigate the MEFV mutation rate in MS patients. METHODS Patients with definite MS were retrieved from the cohort of FMF patients in the Reference Center for Rare Auto-inflammatory Diseases and Amyloidosis (CEREMAIA). We also performed a systematic literature review of articles from PubMed that were published from 1990 to 2020. RESULTS Twenty-four patients were included in the case series: five patients (1.3%) from our cohort of 364 and 19 patients from the literature. The sex ratio was 2:1. The mean age at diagnosis of FMF was 19 years old; and that for MS was 29 years old. Seven studies investigating the MEFV mutation rate in MS patients were included. Three studies found a higher mutation rate in MS patients than in the control group. CONCLUSION FMF and MS features were comparable to those of patients with unrelated diseases; and MEFV mutation carriage was not positively correlated with MS. However; MS prevalence in FMF patients was higher than was expected in a healthy population. To a lesser extent; FMF prevalence in MS patients was higher than expected in a healthy population and the difference might not be significant. These data suggest that FMF could be associated with MS; and further studies are needed to investigate a potential causal association.
Collapse
|
37
|
Role of Peripheral Immune Cells in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis. SCI 2021. [DOI: 10.3390/sci3010012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease that affects the myelination of the neurons present in the central nervous system (CNS). The exact etiology of MS development is unclear, but various environmental and genetic factors might play a role in initiating the disease. Experimental autoimmune encephalomyelitis (EAE) is a mouse model that is used to study the pathophysiology of MS disease as well as the effects of possible therapeutic agents. In addition, autoreactive immune cells trigger an inflammatory process upon the recognition of CNS antigens, which leads to destruction of the neurons. These include innate immune cells such as macrophages, dendritic cells, and natural killer cells. Additionally, the activation and extravasation of adaptive immune cells such as CD4+ T cells into the CNS may lead to further exacerbation of the disease. However, many studies revealed that immune cells could have either a protective or pathological role in MS. In this review, we highlight the roles of innate and adaptive immune cellular and soluble players that contribute to the pathogenesis of MS and EAE, which may be used as potential targets for therapy.
Collapse
|
38
|
Borgonetti V, Sanna MD, Lucarini L, Galeotti N. Targeting the RNA-Binding Protein HuR Alleviates Neuroinflammation in Experimental Autoimmune Encephalomyelitis: Potential Therapy for Multiple Sclerosis. Neurotherapeutics 2021; 18:412-429. [PMID: 33200288 PMCID: PMC8116432 DOI: 10.1007/s13311-020-00958-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory and neurodegenerative disease of the central nervous system characterized by demyelination, axonal loss, and motor dysfunction. Activated microglia are associated with the destruction of myelin in the CNS. Activated microglia produce cytokines and proinflammatory factors, favoring neuroinflammation, myelin damage, and neuronal loss, and it is thought to be involved in the disease pathogenesis. The present study investigated the role of post-transcriptional regulation of gene expression on the neuroinflammation related to experimental autoimmune encephalomyelitis (EAE) in mice, by focusing on HuR, an RNA-binding protein involved in inflammatory and immune phenomena. Spinal cord sections of EAE mice showed an increased HuR immunostaining that was abundantly detected in the cytoplasm of activated microglia, a pattern associated with its increased activity. Intrathecal administration of an anti-HuR antisense oligonucleotide (ASO) decreased the proinflammatory activated microglia, inflammatory infiltrates, and the expression of the proinflammatory cytokines IL-1β, TNF-α, and IL-17, and inhibited the activation of the NF-κB pathway. The beneficial effect of anti-HuR ASO in EAE mice corresponded also to a decreased permeability of the blood-brain barrier. EAE mice showed a reduced spinal CD206 immunostaining that was restored by anti-HuR ASO, indicating that HuR silencing promotes a shift to the anti-inflammatory and regenerative microglia phenotype. Mice that received anti-HuR ASO exhibited improved EAE-related motor dysfunction, pain hypersensitivity, and body weight loss. Targeting HuR might represent an innovative and promising perspective to control neurological disturbances in MS patients.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Maria Domenica Sanna
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Laura Lucarini
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Nicoletta Galeotti
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| |
Collapse
|
39
|
Marenna S, Huang SC, Castoldi V, d’Isa R, Costa GD, Comi G, Leocani L. Functional evolution of visual involvement in experimental autoimmune encephalomyelitis. Mult Scler J Exp Transl Clin 2020; 6:2055217320963474. [PMID: 35145730 PMCID: PMC8822451 DOI: 10.1177/2055217320963474] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
Background Experimental autoimmune encephalomyelitis (EAE) is a common animal model of multiple sclerosis (MS). C57BL/6 mice immunized with myelin oligodendrocyte glycoprotein exhibit chronic disease course, together with optic neuritis, consisting of demyelination/axonal loss of the optic nerve. Objectives To characterize functional and structural visual damages in two different phases of EAE: pre- and post-motor onset. Methods Visual alterations were detected with Visual Evoked Potential (VEP), Electroretinogram (ERG) and Optical Coherence Tomography (OCT). Optic nerve histology was performed at 7 (pre-motor onset) or 37 (post-motor onset) days post-immunization (dpi). Results At 7 dpi, optic nerve inflammation was similar in EAE eyes with and without VEP latency delay. Demyelination was detected in EAE eyes with latency delay (p < 0.0001), while axonal loss (p < 0.0001) and ERG b-wave amplitude (p = 0.004) were decreased in EAE eyes without latency delay compared to Healthy controls. At 37 dpi, functional and structural optic nerve damage were comparable between EAE groups, while a decrease of ERG amplitude and NGCC thickness were found in EAE eyes with VEP latency delay detected post-motor onset. Conclusions Thanks to non-invasive methods, we studied the visual system in a MS model, which could be useful for developing specific therapeutic strategies to target different disease phases.
Collapse
Affiliation(s)
- Silvia Marenna
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
| | - Su-Chun Huang
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
| | - Valerio Castoldi
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
| | - Raffaele d’Isa
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
| | - Gloria Dalla Costa
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giancarlo Comi
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
| | - Letizia Leocani
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
40
|
Santos G, Barateiro A, Brites D, Fernandes A. S100B Impairs Oligodendrogenesis and Myelin Repair Following Demyelination Through RAGE Engagement. Front Cell Neurosci 2020; 14:279. [PMID: 33100970 PMCID: PMC7500156 DOI: 10.3389/fncel.2020.00279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/06/2020] [Indexed: 01/30/2023] Open
Abstract
Increased expression of S100B and its specific receptor for advanced glycation end products (RAGE) has been described in patients with multiple sclerosis (MS), being associated with an active demyelinating process. We previously showed that a direct neutralization of S100B reduces lysophosphatidylcholine (LPC)-induced demyelination and inflammation using an ex vivo demyelinating model. However, whether S100B actions occur through RAGE and how oligodendrogenesis and remyelination are affected are not clarified. To evaluate the role of the S100B–RAGE axis in the course of a demyelinating insult, organotypic cerebellar slice cultures (OCSC) were demyelinated with LPC in the presence or absence of RAGE antagonist FPS-ZM1. Then, we explored the effects of the S100B–RAGE axis inhibition on glia reactivity and inflammation, myelination and neuronal integrity, and on oligodendrogenesis and remyelination. In the present study, we confirmed that LPC-induced demyelination increased S100B and RAGE expression, while RAGE antagonist FPS-ZM1 markedly reduced their content and altered RAGE cellular localization. Furthermore, FPS-ZM1 prevented LPC-induced microgliosis and astrogliosis, as well as NF-κB activation and pro-inflammatory cytokine gene expression. In addition, RAGE antagonist reduced LPC-induced demyelination having a beneficial effect on axonal and synaptic protein preservation. We have also observed that RAGE engagement is needed for LPC-induced oligodendrocyte (OL) maturation arrest and loss of mature myelinating OL, with these phenomena being prevented by FPS-ZM1. Our data suggest that increased levels of mature OL in the presence of FPS-ZM1 are related to increased expression of microRNAs (miRs) associated with OL differentiation and remyelination, such as miR-23a, miR-219a, and miR-338, which are defective upon LPC incubation. Finally, our electron microscopy data show that inhibition of the S100B–RAGE axis prevents axonal damage and myelin loss, in parallel with enhanced functional remyelination, as observed by the presence of thinner myelin sheaths when compared with Control. Overall, our data implicate the S100B–RAGE axis in the extent of myelin and neuronal damage, as well as in the inflammatory response that follows a demyelinating insult. Thus, prevention of RAGE engagement may represent a novel strategy for promoting not only inflammatory reduction but also neuronal and myelin preservation and/or remyelination, improving recovery in a demyelinating condition as MS.
Collapse
Affiliation(s)
- Gisela Santos
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Andreia Barateiro
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Dora Brites
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Adelaide Fernandes
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
41
|
Koudriavtseva T, Stefanile A, Fiorelli M, Lapucci C, Lorenzano S, Zannino S, Conti L, D'Agosto G, Pimpinelli F, Di Domenico EG, Mandoj C, Giannarelli D, Donzelli S, Blandino G, Salvetti M, Inglese M. Coagulation/Complement Activation and Cerebral Hypoperfusion in Relapsing-Remitting Multiple Sclerosis. Front Immunol 2020; 11:548604. [PMID: 33193314 PMCID: PMC7655134 DOI: 10.3389/fimmu.2020.548604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Introduction Multiple sclerosis (MS) is a demyelinating disease of the central nervous system with an underlying immune-mediated and inflammatory pathogenesis. Innate immunity, in addition to the adaptive immune system, plays a relevant role in MS pathogenesis. It represents the immediate non-specific defense against infections through the intrinsic effector mechanism “immunothrombosis” linking inflammation and coagulation. Moreover, decreased cerebral blood volume (CBV), cerebral blood flow (CBF), and prolonged mean transit time (MTT) have been widely demonstrated by MRI in MS patients. We hypothesized that coagulation/complement and platelet activation during MS relapse, likely during viral infections, could be related to CBF decrease. Our specific aims are to evaluate whether there are differences in serum/plasma levels of coagulation/complement factors between relapsing-remitting (RR) MS patients (RRMS) in relapse and those in remission and healthy controls as well as to assess whether brain hemodynamic changes detected by MRI occur in relapse compared with remission. This will allow us to correlate coagulation status with perfusion and demographic/clinical features in MS patients. Materials and Methods This is a multi-center, prospective, controlled study. RRMS patients (1° group: 30 patients in relapse; 2° group: 30 patients in remission) and age/sex-matched controls (3° group: 30 subjects) will be enrolled in the study. Patients and controls will be tested for either coagulation/complement (C3, C4, C4a, C9, PT, aPTT, fibrinogen, factor II, VIII, and X, D-dimer, antithrombin, protein C, protein S, von-Willebrand factor), soluble markers of endothelial damage (thrombomodulin, Endothelial Protein C Receptor), antiphospholipid antibodies, lupus anticoagulant, complete blood count, viral serological assays, or microRNA microarray. Patients will undergo dynamic susceptibility contrast-enhanced MRI using a 3.0-T scanner to evaluate CBF, CBV, MTT, lesion number, and volume. Statistical Analysis ANOVA and unpaired t-tests will be used. The level of significance was set at p ≤ 0.05. Discussion Identifying a link between activation of coagulation/complement system and cerebral hypoperfusion could improve the identification of novel molecular and/or imaging biomarkers and targets, leading to the development of new effective therapeutic strategies in MS. Clinical Trial Registration Clinicaltrials.gov, identifier NCT04380220.
Collapse
Affiliation(s)
- Tatiana Koudriavtseva
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Annunziata Stefanile
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Fiorelli
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Caterina Lapucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Svetlana Lorenzano
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Silvana Zannino
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Conti
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanna D'Agosto
- Clinical Pathology and Microbiology Unit, IRCC San Gallicano Institute, Rome, Italy
| | - Fulvia Pimpinelli
- Clinical Pathology and Microbiology Unit, IRCC San Gallicano Institute, Rome, Italy
| | | | - Chiara Mandoj
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Diana Giannarelli
- Biostatistics, Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sara Donzelli
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Salvetti
- Department of Neuroscience Mental Health and Sensory Organs (NEMOS), Sapienza University, Sant'Andrea Hospital, Rome, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Department of Neurology, Radiology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
42
|
Cheli VT, Correale J, Paez PM, Pasquini JM. Iron Metabolism in Oligodendrocytes and Astrocytes, Implications for Myelination and Remyelination. ASN Neuro 2020; 12:1759091420962681. [PMID: 32993319 PMCID: PMC7545512 DOI: 10.1177/1759091420962681] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Iron is a key nutrient for normal central nervous system (CNS) development and function; thus, iron deficiency as well as iron excess may result in harmful effects in the CNS. Oligodendrocytes and astrocytes are crucial players in brain iron equilibrium. However, the mechanisms of iron uptake, storage, and efflux in oligodendrocytes and astrocytes during CNS development or under pathological situations such as demyelination are not completely understood. In the CNS, iron is directly required for myelin production as a cofactor for enzymes involved in ATP, cholesterol and lipid synthesis, and oligodendrocytes are the cells with the highest iron levels in the brain which is linked to their elevated metabolic needs associated with the process of myelination. Unlike oligodendrocytes, astrocytes do not have a high metabolic requirement for iron. However, these cells are in close contact with blood vessel and have a strong iron transport capacity. In several pathological situations, changes in iron homoeostasis result in altered cellular iron distribution and accumulation and oxidative stress. In inflammatory demyelinating diseases such as multiple sclerosis, reactive astrocytes accumulate iron and upregulate iron efflux and influx molecules, which suggest that they are outfitted to take up and safely recycle iron. In this review, we will discuss the participation of oligodendrocytes and astrocytes in CNS iron homeostasis. Understanding the molecular mechanisms of iron uptake, storage, and efflux in oligodendrocytes and astrocytes is necessary for planning effective strategies for iron management during CNS development as well as for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Veronica T Cheli
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, The State University of New York, University at Buffalo, Buffalo, New York, United States
| | | | - Pablo M Paez
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, Hunter James Kelly Research Institute, The State University of New York, University at Buffalo, Buffalo, New York, United States
| | - Juana M Pasquini
- CONICET, Instituto de Química y Fisicoquímica Biológicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
43
|
Goldfarb S, Fainstein N, Ben-Hur T. Electroconvulsive stimulation attenuates chronic neuroinflammation. JCI Insight 2020; 5:137028. [PMID: 32780728 PMCID: PMC7526446 DOI: 10.1172/jci.insight.137028] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Electroconvulsive therapy is highly effective in resistant depression by unknown mechanisms. Microglial toxicity was suggested to mediate depression and plays key roles in neuroinflammatory and degenerative diseases, where there is critical shortage in therapies. We examined the effects of electroconvulsive seizures (ECS) on chronic neuroinflammation and microglial neurotoxicity. Electric brain stimulation inducing full tonic-clonic seizures during chronic relapsing-progressive experimental autoimmune encephalomyelitis (EAE) reduced spinal immune cell infiltration, reduced myelin and axonal loss, and prevented clinical deterioration. Using the transfer EAE model, we examined the effect of ECS on systemic immune response in donor mice versus ECS effect on CNS innate immune activity in recipient mice. ECS did not affect encephalitogenicity of systemic T cells, but it targeted the CNS directly to inhibit T cell-induced neuroinflammation. In vivo and ex vivo assays indicated that ECS suppressed microglial neurotoxicity by reducing inducible NOS expression, nitric oxide, and reactive oxygen species (ROS) production, and by reducing CNS oxidative stress. Microglia from ECS-treated EAE mice expressed less T cell stimulatory and chemoattractant factors. Our findings indicate that electroconvulsive therapy targets the CNS innate immune system to reduce neuroinflammation by attenuating microglial neurotoxicity. These findings signify a potentially novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric, and neurodegenerative diseases.
Collapse
|
44
|
Moreno-García Á, Bernal-Chico A, Colomer T, Rodríguez-Antigüedad A, Matute C, Mato S. Gene Expression Analysis of Astrocyte and Microglia Endocannabinoid Signaling during Autoimmune Demyelination. Biomolecules 2020; 10:biom10091228. [PMID: 32846891 PMCID: PMC7563448 DOI: 10.3390/biom10091228] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
The endocannabinoid system is associated with protective effects in multiple sclerosis (MS) that involve attenuated innate immune cell responses. Astrocytes and microglia are modulated by endocannabinoids and participate in the biosynthesis and metabolism of these compounds. However, the role of neuroglial cells as targets and mediators of endocannabinoid signaling in MS is poorly understood. Here we used a microfluidic RT-qPCR screen to assess changes in the expression of the main endocannabinoid signaling genes in astrocytes and microglia purified from female mice during the time-course of experimental autoimmune encephalomyelitis (EAE). We show that astrocytes and microglia upregulate the expression of genes encoding neurotoxic A1 and pro-inflammatory molecules at the acute disease with many of these transcripts remaining elevated during the recovery phase. Both cell populations exhibited an early onset decrease in the gene expression levels of 2-arachidonoylglycerol (2-AG) hydrolytic enzymes that persisted during EAE progression as well as cell-type-specific changes in the transcript levels for genes encoding cannabinoid receptors and molecules involved in anandamide (AEA) signaling. Our results demonstrate that astrocytes and microglia responses to autoimmune demyelination involve alterations in the expression of multiple endocannabinoid signaling-associated genes and suggest that this system may regulate the induction of neurotoxic and pro-inflammatory transcriptional programs in both cell types during MS.
Collapse
Affiliation(s)
- Álvaro Moreno-García
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
| | - Ana Bernal-Chico
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
| | - Teresa Colomer
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
| | - Alfredo Rodríguez-Antigüedad
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
- Biocruces, Bizkaia, E-48903 Barakaldo, Spain
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
| | - Susana Mato
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
- Biocruces, Bizkaia, E-48903 Barakaldo, Spain
- Correspondence:
| |
Collapse
|
45
|
Bijelić DD, Milićević KD, Lazarević MN, Miljković DM, Bogdanović Pristov JJ, Savić DZ, Petković BB, Andjus PR, Momčilović MB, Nikolić LM. Central nervous system-infiltrated immune cells induce calcium increase in astrocytes via astroglial purinergic signaling. J Neurosci Res 2020; 98:2317-2332. [PMID: 32799373 DOI: 10.1002/jnr.24699] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/22/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
Interaction between autoreactive immune cells and astroglia is an important part of the pathologic processes that fuel neurodegeneration in multiple sclerosis. In this inflammatory disease, immune cells enter into the central nervous system (CNS) and they spread through CNS parenchyma, but the impact of these autoreactive immune cells on the activity pattern of astrocytes has not been defined. By exploiting naïve astrocytes in culture and CNS-infiltrated immune cells (CNS IICs) isolated from rat with experimental autoimmune encephalomyelitis (EAE), here we demonstrate previously unrecognized properties of immune cell-astrocyte interaction. We show that CNS IICs but not the peripheral immune cell application, evokes a rapid and vigorous intracellular Ca2+ increase in astrocytes by promoting glial release of ATP. ATP propagated Ca2+ elevation through glial purinergic P2X7 receptor activation by the hemichannel-dependent nucleotide release mechanism. Astrocyte Ca2+ increase is specifically triggered by the autoreactive CD4+ T-cell application and these two cell types exhibit close spatial interaction in EAE. Therefore, Ca2+ signals may mediate a rapid astroglial response to the autoreactive immune cells in their local environment. This property of immune cell-astrocyte interaction may be important to consider in studies interrogating CNS autoimmune disease.
Collapse
Affiliation(s)
- Dunja D Bijelić
- Faculty of Biology, Center for Laser Microscopy, University of Belgrade, Belgrade, Serbia
| | - Katarina D Milićević
- Faculty of Biology, Center for Laser Microscopy, University of Belgrade, Belgrade, Serbia
| | - Milica N Lazarević
- Department of Immunology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Djordje M Miljković
- Department of Immunology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena J Bogdanović Pristov
- Department of Life Sciences, Institute for Multidisciplinary Research, University of Belgrade, Belgrade, Serbia
| | - Danijela Z Savić
- Department of Neurobiology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Branka B Petković
- Department of Neurophysiology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Pavle R Andjus
- Faculty of Biology, Center for Laser Microscopy, University of Belgrade, Belgrade, Serbia
| | - Miljana B Momčilović
- Department of Immunology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ljiljana M Nikolić
- Department of Neurophysiology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
46
|
Morris AH, Hughes KR, Oakes RS, Cai MM, Miller SD, Irani DN, Shea LD. Engineered immunological niches to monitor disease activity and treatment efficacy in relapsing multiple sclerosis. Nat Commun 2020; 11:3871. [PMID: 32747712 PMCID: PMC7398910 DOI: 10.1038/s41467-020-17629-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 07/09/2020] [Indexed: 12/19/2022] Open
Abstract
Relapses in multiple sclerosis can result in irreversible nervous system tissue injury. If these events could be detected early, targeted immunotherapy could potentially slow disease progression. We describe the use of engineered biomaterial-based immunological niches amenable to biopsy to provide insights into the phenotype of innate immune cells that control disease activity in a mouse model of multiple sclerosis. Differential gene expression in cells from these niches allow monitoring of disease dynamics and gauging the effectiveness of treatment. A proactive treatment regimen, given in response to signal within the niche but before symptoms appeared, substantially reduced disease. This technology offers a new approach to monitor organ-specific autoimmunity, and represents a platform to analyze immune dysfunction within otherwise inaccessible target tissues.
Collapse
Affiliation(s)
- Aaron H Morris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kevin R Hughes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Robert S Oakes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michelle M Cai
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David N Irani
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Jørgensen LØ, Hyrlov KH, Elkjaer ML, Weber AB, Pedersen AE, Svenningsen ÅF, Illes Z. Cladribine modifies functional properties of microglia. Clin Exp Immunol 2020; 201:328-340. [PMID: 32492189 PMCID: PMC7419928 DOI: 10.1111/cei.13473] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023] Open
Abstract
Cladribine (CdA), an oral prodrug approved for the treatment of relapsing multiple sclerosis, selectively depletes lymphocytes. CdA passes the blood–brain barrier, suggesting a potential effect on central nervous system (CNS) resident cells. We examined if CdA modifies the phenotype and function of naive and activated primary mouse microglia, when applied in the concentrations 0·1–1 μM that putatively overlap human cerebrospinal fluid (CSF) concentrations. Primary microglia cultures without stimulation or in the presence of proinflammatory lipopolysaccharide (LPS) or anti‐inflammatory interleukin (IL)‐4 were treated with different concentrations of CdA for 24 h. Viability was assessed by MTT [3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide] assay. Phagocytotic ability and morphology were examined by flow cytometry and random migration using IncuCyte Zoom and TrackMate. Change in gene expression was examined by quantitative polymerase chain reaction (qPCR) and protein secretion by Meso Scale Discovery. We found that LPS and IL‐4 up‐regulated deoxycytidine kinase (DCK) expression. Only activated microglia were affected by CdA, and this was unrelated to viability. CdA 0·1–1 μM significantly reduced granularity, phagocytotic ability and random migration of activated microglia. CdA 10 μM increased the IL‐4‐induced gene expression of arginase 1 (Arg1) and LPS‐induced expression of IL‐1β, tumor necrosis factor (TNF), inducible nitric oxide synthase (iNOS) and Arg1, but protein secretion remained unaffected. CdA 10 μM potentiated the increased expression of anti‐inflammatory TNF receptor 2 (TNF‐R2) but not TNF‐R1 induced by LPS. This suggests that microglia acquire a less activated phenotype when treated with 0·1–1 μM CdA that putatively overlaps human CSF concentrations. This may be related to the up‐regulated gene expression of DCK upon activation, and suggests a potential alternative mechanism of CdA with direct effect on CNS resident cells.
Collapse
Affiliation(s)
- L Ø Jørgensen
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - K H Hyrlov
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - M L Elkjaer
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - A B Weber
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - A E Pedersen
- Merck A/S, Søborg, Denmark.,Department of Odontology, University of Copenhagen, Copenhagen, Denmark
| | - Å Fex Svenningsen
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Z Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
48
|
El Waly B, Buttigieg E, Karakus C, Brustlein S, Debarbieux F. Longitudinal Intravital Microscopy Reveals Axon Degeneration Concomitant With Inflammatory Cell Infiltration in an LPC Model of Demyelination. Front Cell Neurosci 2020; 14:165. [PMID: 32655371 PMCID: PMC7324938 DOI: 10.3389/fncel.2020.00165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/15/2020] [Indexed: 11/13/2022] Open
Abstract
Demyelination and axon degeneration are major events in all neurodegenerative diseases, including multiple sclerosis. Intoxication of oligodendrocytes with lysophosphatidylcholine (LPC) is often used as a selective model of focal and reversible demyelination thought to have no incidence for neurons. To characterize the cascade of cellular events involved in LPC-induced demyelination, we have combined intravital coherent antistoke Raman scattering microscopy with intravital two-photon fluorescence microscopy in multicolor transgenic reporter mice. Moreover, taking advantage of a unique technique of spinal glass window implantation, we here provide the first longitudinal description of cell dynamics in the same volume of interest over weeks after insults. We have detected several patterns of axon–myelin interactions and classified them in early and advanced events. Unexpectedly, we have found that oligodendrocyte damages are followed by axon degeneration within 2 days after LPC incubation, and this degeneration is amplified after the recruitment of the peripheral proinflammatory cells at day 4. Beyond day 7, the recovery of axon number and myelin takes 3 more weeks postlesion and involves a new wave of anti-inflammatory innate immune cells at day 14. Therefore, recurrent imaging over several weeks suggests an important role of peripheral immune cells in regulating both the axonal and oligodendroglial fates and thereby the remyelination status. Better understanding the recruitment of peripheral immune cells during demyelinating events should help to improve diagnosis and therapy.
Collapse
Affiliation(s)
- Bilal El Waly
- Institut des Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France.,Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| | - Emeline Buttigieg
- Institut des Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France.,Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Cem Karakus
- Institut des Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France.,Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| | - Sophie Brustlein
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France.,CNRS, IBDM, Turing Center for Living System, Aix-Marseille Université, Marseille, France
| | - Franck Debarbieux
- Institut des Neurosciences de la Timone, UMR 7289, CNRS, Aix-Marseille Université, Marseille, France.,Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| |
Collapse
|
49
|
Development of the first in vivo GPR17 ligand through an iterative drug discovery pipeline: A novel disease-modifying strategy for multiple sclerosis. PLoS One 2020; 15:e0231483. [PMID: 32320409 PMCID: PMC7176092 DOI: 10.1371/journal.pone.0231483] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/24/2020] [Indexed: 01/09/2023] Open
Abstract
The GPR17 receptor, expressed on oligodendroglial precursors (OPCs, the myelin producing cells), has emerged as an attractive target for a pro-myelinating strategy in multiple sclerosis (MS). However, the proof-of-concept that selective GPR17 ligands actually exert protective activity in vivo is still missing. Here, we exploited an iterative drug discovery pipeline to prioritize novel and selective GPR17 pro-myelinating agents out of more than 1,000,000 compounds. We first performed an in silico high-throughput screening on GPR17 structural model to identify three chemically-diverse ligand families that were then combinatorially exploded and refined. Top-scoring compounds were sequentially tested on reference pharmacological in vitro assays with increasing complexity, ending with myelinating OPC-neuron co-cultures. Successful ligands were filtered through in silico simulations of metabolism and pharmacokinetics, to select the most promising hits, whose dose and ability to target the central nervous system were then determined in vivo. Finally, we show that, when administered according to a preventive protocol, one of them (named by us as galinex) is able to significantly delay the onset of experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. This outcome validates the predictivity of our pipeline to identify novel MS-modifying agents.
Collapse
|
50
|
Namekata K, Guo X, Kimura A, Azuchi Y, Kitamura Y, Harada C, Harada T. Roles of the DOCK-D family proteins in a mouse model of neuroinflammation. J Biol Chem 2020; 295:6710-6720. [PMID: 32241915 DOI: 10.1074/jbc.ra119.010438] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 03/16/2020] [Indexed: 02/01/2023] Open
Abstract
The DOCK-D (dedicator of cytokinesis D) family proteins are atypical guanine nucleotide exchange factors that regulate Rho GTPase activity. The family consists of Zizimin1 (DOCK9), Zizimin2 (DOCK11), and Zizimin3 (DOCK10). Functions of the DOCK-D family proteins are presently not well-explored, and the role of the DOCK-D family in neuroinflammation is unknown. In this study, we generated three mouse lines in which DOCK9 (DOCK9 -/-), DOCK10 (DOCK10 -/-), or DOCK11 (DOCK11 -/-) had been deleted and examined the phenotypic effects of these gene deletions in MOG35-55 peptide-induced experimental autoimmune encephalomyelitis, an animal model of the neuroinflammatory disorder multiple sclerosis. We found that all the gene knockout lines were healthy and viable. The only phenotype observed under normal conditions was a slightly smaller proportion of B cells in splenocytes in DOCK10 -/- mice than in the other mouse lines. We also found that the migration ability of macrophages is impaired in DOCK10 -/- and DOCK11 -/- mice and that the severity of experimental autoimmune encephalomyelitis was ameliorated only in DOCK10 -/- mice. No apparent phenotype was observed for DOCK9 -/- mice. Further investigations indicated that lipopolysaccharide stimulation up-regulates DOCK10 expression in microglia and that microglial migration is decreased in DOCK10 -/- mice. Up-regulation of C-C motif chemokine ligand 2 (CCL2) expression induced by activation of Toll-like receptor 4 or 9 signaling was reduced in DOCK10 -/- astrocytes compared with WT astrocytes. Taken together, our findings suggest that DOCK10 plays a role in innate immunity and neuroinflammation and might represent a potential therapeutic target for managing multiple sclerosis.
Collapse
Affiliation(s)
- Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yuriko Azuchi
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yuta Kitamura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|