1
|
Pilar Matud M, del Pino MJ, Bethencourt JM, Estefanía Lorenzo D. Stressful Events, Psychological Distress and Well-Being during the Second Wave of COVID-19 Pandemic in Spain: A Gender Analysis. APPLIED RESEARCH IN QUALITY OF LIFE 2022; 18:1-29. [PMID: 36619208 PMCID: PMC9803894 DOI: 10.1007/s11482-022-10140-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/25/2022] [Indexed: 06/17/2023]
Abstract
The present study investigates gender differences in stressful events, psychological distress and well-being during the second wave of COVID-19 in Spain, analyzing women's and men's risk and resilience factors for psychological distress and for well-being. Participants were 1758 individuals from the general population, 50.8% women, aged between 18 and 79 years. Women and men did not differ in age, number of children, educational level, occupation or marital status. The participants were assessed by seven self-report questionnaires and scales. The results revealed that women experienced more psychological distress, more negative feelings, more stressful events related to the COVID-19 pandemic, more social support, and lower thriving and self-esteem than men. Multiple regression analyses showed that, in the case of women and men as well, self-esteem was the most important predictor of higher well-being and lower psychological distress and negative feelings while more COVID-19 pandemic-related stressful events were associated with higher psychological distress and lower well-being. Another important predictor of greater well-being for either gender was social support while unemployment was associated with lower well-being. In women and men, a higher educational level was associated with greater psychological distress and negative feelings; the male sample revealed that psychological distress was also connected to younger age while in women it was associated with lower instrumental social support. The results suggest that gender plays an important role in the mental health effects of the COVID-19 pandemic, with the risk being higher for women than for men.
Collapse
Affiliation(s)
- M. Pilar Matud
- Department of Clinical Psychology, Psychobiology and Methodology, Facultad de Psicología y Logopedia, Universidad de La Laguna, Apartado 456, 38200 La Laguna, Spain
| | - Mª José del Pino
- Department of Sociology, University Pablo de Olavide, Seville, Spain
| | - Juan Manuel Bethencourt
- Department of Clinical Psychology, Psychobiology and Methodology, Facultad de Psicología y Logopedia, Universidad de La Laguna, Apartado 456, 38200 La Laguna, Spain
| | - D. Estefanía Lorenzo
- Department of Clinical Psychology, Psychobiology and Methodology, Facultad de Psicología y Logopedia, Universidad de La Laguna, Apartado 456, 38200 La Laguna, Spain
| |
Collapse
|
2
|
Voskuhl RR, MacKenzie-Graham A. Chronic experimental autoimmune encephalomyelitis is an excellent model to study neuroaxonal degeneration in multiple sclerosis. Front Mol Neurosci 2022; 15:1024058. [PMID: 36340686 PMCID: PMC9629273 DOI: 10.3389/fnmol.2022.1024058] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 09/30/2022] [Indexed: 08/19/2023] Open
Abstract
Animal models of multiple sclerosis (MS), specifically experimental autoimmune encephalomyelitis (EAE), have been used extensively to develop anti-inflammatory treatments. However, the similarity between MS and one particular EAE model does not end at inflammation. MS and chronic EAE induced in C57BL/6 mice using myelin oligodendrocyte glycoprotein (MOG) peptide 35-55 share many neuropathologies. Beyond both having white matter lesions in spinal cord, both also have widespread neuropathology in the cerebral cortex, hippocampus, thalamus, striatum, cerebellum, and retina/optic nerve. In this review, we compare neuropathologies in each of these structures in MS with chronic EAE in C57BL/6 mice, and find evidence that this EAE model is well suited to study neuroaxonal degeneration in MS.
Collapse
Affiliation(s)
- Rhonda R. Voskuhl
- UCLA MS Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | |
Collapse
|
3
|
Sun T, Zeng H, Fan L, Fei J, Chen G. Semaphorin 6D regulate corralling, hematoma compaction and white matter injury in mice after intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2022; 31:106803. [PMID: 36174325 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106803] [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: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES The Semaphorin 6D (SEMA6D) shows important roles in cell guidance and lipid metabolism, but the effects and mechanisms of SEMA6D on tissue repair, white matter injury and the recovery of neurological function after intracerebral hemorrhage have not been well studied. MATERIALS AND METHODS In this study, the autologous whole blood injection model of intracerebral hemorrhage was established in C57 male mice. SEMA6D knockout CRISPR utilized in the study. Assessments included neurological score evaluation and immunofluorescence. RESULTS SEMA6D increased and peaked at 7d after intracerebral hemorrhage, and mainly located in neurons, microglia and astrocytes. SEMA6D knockout CRISPR aggravated neurological function and showed signs of poorer corralling and hematoma resolution, with more compartments of well-established physical barrier and more extensive GFAP positive astrocytic border. Furthermore, SEMA6D can prevent the decrease of NF-H in the peri-hematoma region, while SEMA6D knockout aggravated WMI. CONCLUSIONS Our study suggested that SEMA6D could influence the recovery of neurological function by regulating the corralling, hematoma compaction and WMI in mice after intracerebral hemorrhage.
Collapse
Affiliation(s)
- Ting Sun
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China.
| | - Hanhai Zeng
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| | - Linfeng Fan
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| | - Jing Fei
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China.
| | - Gao Chen
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| |
Collapse
|
4
|
Xie S, Yang J, Huang S, Fan Y, Xu T, He J, Guo J, Ji X, Wang Z, Li P, Chen J, Zhang Y. Disrupted myelination network in the cingulate cortex of Parkinson's disease. IET Syst Biol 2022; 16:98-119. [PMID: 35394697 PMCID: PMC9290774 DOI: 10.1049/syb2.12043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/31/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022] Open
Abstract
The cingulate cortex is part of the conserved limbic system, which is considered as a hub of emotional and cognitive control. Accumulating evidence suggested that involvement of the cingulate cortex is significant for cognitive impairment of Parkinson's disease (PD). However, mechanistic studies of the cingulate cortex in PD pathogenesis are limited. Here, transcriptomic and regulatory network analyses were conducted for the cingulate cortex in PD. Enrichment and clustering analyses showed that genes involved in regulation of membrane potential and glutamate receptor signalling pathway were upregulated. Importantly, myelin genes and the oligodendrocyte development pathways were markedly downregulated, indicating disrupted myelination in PD cingulate cortex. Cell‐type‐specific signatures revealed that myelinating oligodendrocytes were the major cell type damaged in the PD cingulate cortex. Furthermore, downregulation of myelination pathways in the cingulate cortex were shared and validated in another independent RNAseq cohort of dementia with Lewy bodies (DLB). In combination with ATACseq data, gene regulatory networks (GRNs) were further constructed for 32 transcription factors (TFs) and 466 target genes among differentially expressed genes (DEGs) using a tree‐based machine learning algorithm. Several transcription factors, including Olig2, Sox8, Sox10, E2F1, and NKX6‐2, were highlighted as key nodes in a sub‐network, which control many overlapping downstream targets associated with myelin formation and gliogenesis. In addition, the authors have validated a subset of DEGs by qPCRs in two PD mouse models. Notably, seven of these genes,TOX3, NECAB2 NOS1, CAPN3, NR4A2, E2F1 and FOXP2, have been implicated previously in PD or neurodegeneration and are worthy of further studies as novel candidate genes. Together, our findings provide new insights into the role of remyelination as a promising new approach to treat PD after demyelination.
Collapse
Affiliation(s)
- Song Xie
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiajun Yang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shenghui Huang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yuanlan Fan
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tao Xu
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China
| | - Jiangshuang He
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiahao Guo
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiang Ji
- Department of Mathematics, School of Science & Engineering, Tulane University, New Orleans, Louisiana, USA
| | - Zhibo Wang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiangfan Chen
- Molecular Neuropharmacology Lab, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China
| | - Yi Zhang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,The Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, Zhejiang Province, China.,Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
5
|
Lee A, Kwon OW, Jung KR, Song GJ, Yang HJ. The effects of Korean Red Ginseng-derived components on oligodendrocyte lineage cells: Distinct facilitatory roles of the non-saponin and saponin fractions, and Rb1, in proliferation, differentiation and myelination. J Ginseng Res 2022; 46:104-114. [PMID: 35035243 PMCID: PMC8753459 DOI: 10.1016/j.jgr.2021.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/21/2021] [Accepted: 04/21/2021] [Indexed: 12/05/2022] Open
Abstract
Background Abnormalities of myelin, which increases the efficiency of action potential conduction, are found in neurological disorders. Korean Red Ginseng (KRG) demonstrates therapeutic efficacy against some of these conditions, however effects on oligodendrocyte (OL)s are not well known. Here, we examined the effects of KRG-derived components on development and protection of OL-lineage cells. Methods Primary OL precursor cell (OPC) cultures were prepared from neonatal mouse cortex. The protective efficacies of the KRG components were examined against inhibitors of mitochondrial respiratory chain activity. For in vivo function of Rb1 on myelination, after 10 days of oral gavage into adult male mice, forebrains were collected. OPC proliferation were assessed by BrdU incorporation, and differentiation and myelination were examined by qPCR, western blot and immunocytochemistry. Results The non-saponin promoted OPC proliferation, while the saponin promoted differentiation. Both processes were mediated by AKT and extracellular regulated kinase (ERK) signaling. KRG extract, the saponin and non-saponin protected OPCs against oxidative stress, and both KRG extract and the saponin significantly increased the expression of the antioxidant enzyme. Among 11 major ginsenosides tested, Rb1 significantly increased OL membrane size in vitro. Moreover, Rb1 significantly increased myelin formation in adult mouse brain. Conclusion All KRG components prevented OPC deaths under oxidative stress. While non-saponin promoted proliferation, saponin fraction increased differentiation and OL membrane size. Furthermore, among all the tested ginsenosides, Rb1 showed the biggest increase in the membrane size and significantly enhanced myelination in vivo. These results imply therapeutic potentials of KRG and Rb1 for myelin-related disorders.
Collapse
Affiliation(s)
- Ahreum Lee
- Korea Institute of Brain Science, Seoul, Republic of Korea.,Department of Integrative Biosciences, University of Brain Education, Cheonan, Republic of Korea
| | - Oh Wook Kwon
- Department of Integrative Biosciences, University of Brain Education, Cheonan, Republic of Korea
| | - Kwi Ryun Jung
- Department of Integrative Biosciences, University of Brain Education, Cheonan, Republic of Korea
| | - Gyun Jee Song
- Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic of Korea
| | - Hyun-Jeong Yang
- Korea Institute of Brain Science, Seoul, Republic of Korea.,Department of Integrative Biosciences, University of Brain Education, Cheonan, Republic of Korea
| |
Collapse
|
6
|
Fu X, Zhou G, Wu X, Xu C, Zhou H, Zhuang J, Peng Y, Cao Y, Zeng H, Li Y, Li J, Gao L, Chen G, Wang L, Yan F. Inhibition of P2X4R attenuates white matter injury in mice after intracerebral hemorrhage by regulating microglial phenotypes. J Neuroinflammation 2021; 18:184. [PMID: 34425835 PMCID: PMC8383380 DOI: 10.1186/s12974-021-02239-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/12/2021] [Indexed: 12/28/2022] Open
Abstract
Background White matter injury (WMI) is a major neuropathological event associated with intracerebral hemorrhage (ICH). P2X purinoreceptor 4 (P2X4R) is a member of the P2X purine receptor family, which plays a crucial role in regulating WMI and neuroinflammation in central nervous system (CNS) diseases. Our study investigated the role of P2X4R in the WMI and the inflammatory response in mice, as well as the possible mechanism of action after ICH. Methods ICH was induced in mice via collagenase injection. Mice were treated with 5-BDBD and ANA-12 to inhibit P2X4R and tropomyosin-related kinase receptor B (TrkB), respectively. Immunostaining and quantitative polymerase chain reaction (qPCR) were performed to detect microglial phenotypes after the inhibition of P2X4R. Western blots (WB) and immunostaining were used to examine WMI and the underlying molecular mechanisms. Cylinder, corner turn, wire hanging, and forelimb placement tests were conducted to evaluate neurobehavioral function. Results After ICH, the protein levels of P2X4R were upregulated, especially on day 7 after ICH, and were mainly located in the microglia. The inhibition of P2X4R via 5-BDBD promoted neurofunctional recovery after ICH as well as the transformation of the pro-inflammatory microglia induced by ICH into an anti-inflammatory phenotype, and attenuated ICH-induced WMI. Furthermore, we found that TrkB blockage can reverse the protective effects of WMI as well as neuroprotection after 5-BDBD treatment. This result indicates that P2X4R plays a crucial role in regulating WMI and neuroinflammation and that P2X4R inhibition may benefit patients with ICH. Conclusions Our results demonstrated that P2X4R contributes to WMI by polarizing microglia into a pro-inflammatory phenotype after ICH. Furthermore, the inhibition of P2X4R promoted pro-inflammatory microglia polarization into an anti-inflammatory phenotype, enhanced brain-derived neurotrophic factor (BDNF) production, and through the BDNF/TrkB pathway, attenuated WMI and improved neurological function. Therefore, the regulation of P2X4R activation may be beneficial for the reducing of ICH-induced brain injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02239-3.
Collapse
Affiliation(s)
- Xiongjie Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Guoyang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Xinyan Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Chaoran Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Hang Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yang Cao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Hanhai Zeng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Yin Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| | - Lin Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| |
Collapse
|
7
|
Mangale V, Syage AR, Ekiz HA, Skinner DD, Cheng Y, Stone CL, Brown RM, O'Connell RM, Green KN, Lane TE. Microglia influence host defense, disease, and repair following murine coronavirus infection of the central nervous system. Glia 2020; 68:2345-2360. [PMID: 32449994 PMCID: PMC7280614 DOI: 10.1002/glia.23844] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022]
Abstract
The present study examines functional contributions of microglia in host defense, demyelination, and remyelination following infection of susceptible mice with a neurotropic coronavirus. Treatment with PLX5622, an inhibitor of colony stimulating factor 1 receptor (CSF1R) that efficiently depletes microglia, prior to infection of the central nervous system (CNS) with the neurotropic JHM strain of mouse hepatitis virus (JHMV) resulted in increased mortality compared with control mice that correlated with impaired control of viral replication. Single cell RNA sequencing (scRNASeq) of CD45+ cells isolated from the CNS revealed that PLX5622 treatment resulted in muted CD4+ T cell activation profile that was associated with decreased expression of transcripts encoding MHC class II and CD86 in macrophages but not dendritic cells. Evaluation of spinal cord demyelination revealed a marked increase in white matter damage in PLX5622-treated mice that corresponded with elevated expression of transcripts encoding disease-associated proteins Osteopontin (Spp1), Apolipoprotein E (Apoe), and Triggering receptor expressed on myeloid cells 2 (Trem2) that were enriched within macrophages. In addition, PLX5622 treatment dampened expression of Cystatin F (Cst7), Insulin growth factor 1 (Igf1), and lipoprotein lipase (Lpl) within macrophage populations which have been implicated in promoting repair of damaged nerve tissue and this was associated with impaired remyelination. Collectively, these findings argue that microglia tailor the CNS microenvironment to enhance control of coronavirus replication as well as dampen the severity of demyelination and influence repair.
Collapse
Affiliation(s)
- Vrushali Mangale
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Amber R. Syage
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - H. Atakan Ekiz
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Dominic D. Skinner
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Yuting Cheng
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Colleen L. Stone
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - R. Marshall Brown
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Ryan M. O'Connell
- Division of Microbiology & Immunology, Department of PathologyUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Kim N. Green
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological SciencesUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
8
|
Molecular Effects of FDA-Approved Multiple Sclerosis Drugs on Glial Cells and Neurons of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21124229. [PMID: 32545828 PMCID: PMC7352301 DOI: 10.3390/ijms21124229] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is characterized by peripheral and central inflammatory features, as well as demyelination and neurodegeneration. The available Food and Drug Administration (FDA)-approved drugs for MS have been designed to suppress the peripheral immune system. In addition, however, the effects of these drugs may be partially attributed to their influence on glial cells and neurons of the central nervous system (CNS). We here describe the molecular effects of the traditional and more recent FDA-approved MS drugs Fingolimod, Dimethyl Fumarate, Glatiramer Acetate, Interferon-β, Teriflunomide, Laquinimod, Natalizumab, Alemtuzumab and Ocrelizumab on microglia, astrocytes, neurons and oligodendrocytes. Furthermore, we point to a possible common molecular effect of these drugs, namely a key role for NFκB signaling, causing a switch from pro-inflammatory microglia and astrocytes to anti-inflammatory phenotypes of these CNS cell types that recently emerged as central players in MS pathogenesis. This notion argues for the need to further explore the molecular mechanisms underlying MS drug action.
Collapse
|
9
|
Jiang T, Zhang W, Wang Z. Laquinimod Protects Against TNF-α-Induced Attachment of Monocytes to Human Aortic Endothelial Cells (HAECs) by Increasing the Expression of KLF2. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1683-1691. [PMID: 32440094 PMCID: PMC7222522 DOI: 10.2147/dddt.s243666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/06/2020] [Indexed: 01/01/2023]
Abstract
Introduction As a worldwide health issue, the treatment and prevention of atherosclerosis present an important goal. Increased levels of proinflammatory cytokines such as TNF-α-associated chronic inflammatory response cause endothelial cells to lose their ability to regulate vascular function. Lipid-laden immune cells are recruited to the endothelium where they adhere to the endothelial wall and invade the intimal space, thereby leading to the development of atherosclerotic lesions, fatty plaques, and thickening of the arterial wall. In the present study, for the first time, we investigated the effects of laquinimod, an immunomodulatory agent used for the treatment of multiple sclerosis, on human aortic endothelial in a TNF-α-induced atherosclerotic microenvironment. At present, the mechanism of action of laquinimod is not well defined. Methods The effects of laquinimod on the gene expression of IL-6, MCP-1, VCAM-1, E-selectin, and KLF2 were measured by real-time PCR. ELISA assay was used to determine protein secretion and expression. Phosphorylation of ERK5 and the protein level of KLF2 were measured by Western blot analysis. The attachment of monocytes to endothelial cells was assayed by calcein-AM staining and fluorescent microscopy. Results Our findings demonstrate that laquinimod reduced the expression of key inflammatory cytokines and chemokines, including IL-6, MCP-1, and HMGB1. We further demonstrate that laquinimod significantly reduced the attachment of monocytes to endothelial cells, which is mediated through reduced expression of the cellular adhesion molecules VCAM-1 and E-selectin. Here, we found that laquinimod could significantly increase the expression of KLF2 through activation of ERK5 signaling. The results of our KLF2 knockdown experiment confirm that the effects of laquinimod observed in vitro are dependent on KLF2 expression. Conclusion Together, these findings suggest a potential antiatherosclerotic capacity of laquinimod. Further research will elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Tiechao Jiang
- Department of Cardiovascular Medicine, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Wenhao Zhang
- Department of Cardiovascular Medicine, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Zhongyu Wang
- Department of Cardiovascular Medicine, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China
| |
Collapse
|
10
|
Laquinimod ameliorates secondary brain inflammation. Neurobiol Dis 2019; 134:104675. [PMID: 31731041 DOI: 10.1016/j.nbd.2019.104675] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/21/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence suggests that a degenerative processes within the brain can trigger the formation of new, focal inflammatory lesions in Multiple Sclerosis (MS). Here, we used a novel pre-clinical MS animal model to test whether the amelioration of degenerative brain events reduces the secondary recruitment of peripheral immune cells and, in consequence, inflammatory lesion development. Neural degeneration was induced by a 3 weeks cuprizone intoxication period. To mitigate the cuprizone-induced pathology, animals were treated with Laquinimod (25 mg/kg) during the cuprizone-intoxication period. At the beginning of week 6, encephalitogenic T cell development in peripheral lymphoid organs was induced by the immunization with myelin oligodendrocyte glycoprotein 35-55 peptide (i.e., Cup/EAE). Demyelination, axonal injury and reactive gliosis were determined by immunohistochemistry. Positron emission tomography (PET) imaging was performed to analyze glia activation in vivo. Vehicle-treated cuprizone mice displayed extensive callosal demyelination, glia activation and enhanced TSPO-ligand binding. This cuprizone-induced pathology was profoundly ameliorated in mice treated with Laquinimod. In vehicle-treated Cup/EAE mice, the cuprizone-induced pathology triggered massive peripheral immune cell recruitment into the forebrain, evidenced by multifocal perivascular inflammation, glia activation and neuro-axonal injury. While anti myelin oligodendrocyte glycoprotein 35-55 peptide immune responses were comparable in vehicle- and Laquinimod-treated Cup/EAE mice, the cuprizone-triggered immune cell recruitment was ameliorated by the Laquinimod treatment. This study clearly illustrates that amelioration of a primary brain-intrinsic degenerative process secondary halts peripheral immune cell recruitment and, in consequence, inflammatory lesion development. These findings have important consequences for the interpretation of the results of clinical studies.
Collapse
|
11
|
Nyamoya S, Steinle J, Chrzanowski U, Kaye J, Schmitz C, Beyer C, Kipp M. Laquinimod Supports Remyelination in Non-Supportive Environments. Cells 2019; 8:cells8111363. [PMID: 31683658 PMCID: PMC6912710 DOI: 10.3390/cells8111363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 01/20/2023] Open
Abstract
Inflammatory demyelination, which is a characteristic of multiple sclerosis lesions, leads to acute functional deficits and, in the long term, to progressive axonal degeneration. While remyelination is believed to protect axons, the endogenous-regenerative processes are often incomplete or even completely fail in many multiple sclerosis patients. Although it is currently unknown why remyelination fails, recurrent demyelination of previously demyelinated white matter areas is one contributing factor. In this study, we investigated whether laquinimod, which has demonstrated protective effects in active multiple sclerosis patients, protects against recurrent demyelination. To address this, male mice were intoxicated with cuprizone for up to eight weeks and treated with either a vehicle solution or laquinimod at the beginning of week 5, where remyelination was ongoing. The brains were harvested and analyzed by immunohistochemistry. At the time-point of laquinimod treatment initiation, oligodendrocyte progenitor cells proliferated and maturated despite ongoing demyelination activity. In the following weeks, myelination recovered in the laquinimod- but not vehicle-treated mice, despite continued cuprizone intoxication. Myelin recovery was paralleled by less severe microgliosis and acute axonal injury. In this study, we were able to demonstrate that laquinimod, which has previously been shown to protect against cuprizone-induced oligodendrocyte degeneration, exerts protective effects during oligodendrocyte progenitor differentiation as well. By this mechanism, laquinimod allows remyelination in non-supportive environments. These results should encourage further clinical studies in progressive multiple sclerosis patients.
Collapse
Affiliation(s)
- Stella Nyamoya
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany.
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
| | - Julia Steinle
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
| | - Uta Chrzanowski
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
| | - Joel Kaye
- AyalaPharma, VP Research & Nonclinical Development, Rehovot 7670104, Israel.
| | - Christoph Schmitz
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany.
| | - Cordian Beyer
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
| | - Markus Kipp
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany.
- Centre for Transdisciplinary Neurosciences, Rostock University Medical Center, 18057 Rostock, Germany.
| |
Collapse
|
12
|
Liu X, Deng J, Li R, Tan C, Li H, Yang Z, Chen L, Chen Y, Tan X. ERβ-selective agonist alleviates inflammation in a multiple sclerosis model via regulation of MHC II in microglia. Am J Transl Res 2019; 11:4411-4424. [PMID: 31396345 PMCID: PMC6684890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 06/13/2019] [Indexed: 06/10/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune, demyelinating, and neurodegenerative disease of the central nervous system (CNS) that affects 2-2.5 million people worldwide. Although the etiology of MS is not well known, MS is widely considered to be an autoimmune disease. Currently approved MS drugs reduce relapse rates but fail to reverse or prevent neurodegeneration and disability progression. Increasing evidence indicates that microglia and major histocompatibility complex class II (MHC II) expression in these cells play important roles in the pathophysiology of MS. For a T cell to contribute to CNS pathogenesis, it must be reactivated by antigen-presenting cells within the CNS parenchyma. Susceptibility to MS is associated with MHC II genes, suggesting that presentation of antigens on MHC II plays an important role in CD4+ T-cell reactivation and disease initiation. An ERβ-selective agonist was previously reported to suppress reactivation of T cells invading the spinal cord, thereby reducing the severity of symptoms and decreasing mortality in the first 2 weeks after disease onset. However, the mechanism by which the expression of MHC II in microglia is regulated by ERβ-selective agonists is still unclear. Therefore, we hypothesize that ERβ-selective agonists inhibit MHC II expression in microglia via inhibition of class II trans-activator (CIITA) expression by a mechanism involving inhibition of the translocation of IFNγ regulatory factor (IRF-1) to the nucleus, thereby inhibiting the inflammatory response and symptoms in the MS model.
Collapse
Affiliation(s)
- Xi Liu
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| | - Jing Deng
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| | - Rong Li
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| | - Changhong Tan
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| | - Hongli Li
- Department of Histology and Embryology, Third Military Medical UniversityChongqing 400038, China
| | - Zhong Yang
- Department of Clinical Hematology, Southwest Hospital, Third Military Medical University (Army Medical University)Chongqing 400038, China
| | - Lifen Chen
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| | - Yangmei Chen
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| | - Xinjie Tan
- Department of Neurology, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| |
Collapse
|
13
|
Karim H, Kim SH, Lauderdale K, Lapato AS, Atkinson K, Yasui N, Yamate-Morgan H, Sekyi M, Katzenellenbogen JA, Tiwari-Woodruff SK. Analogues of ERβ ligand chloroindazole exert immunomodulatory and remyelinating effects in a mouse model of multiple sclerosis. Sci Rep 2019; 9:503. [PMID: 30679747 PMCID: PMC6345788 DOI: 10.1038/s41598-018-37420-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/27/2018] [Indexed: 01/06/2023] Open
Abstract
Pharmaceutical agents currently approved for the treatment of multiple sclerosis reduce relapse rates, but do not reverse or prevent neurodegeneration nor initiate myelin repair. The highly selective estrogen receptor (ER) β ligand chloroindazole (IndCl) shows particular promise promoting both remyelination while reducing inflammatory cytokines in the central nervous system of mice with experimental autoimmune encephalomyelitis. To optimize these benefits, we developed and screened seven novel IndCl analogues for their efficacy in promoting primary oligodendrocyte (OL) progenitor cell survival, proliferation, and differentiation in vitro by immunohistochemistry. Two analogues, IndCl-o-chloro and IndCl-o-methyl, induced proliferation and differentiation equivalent to IndCl and were selected for subsequent in vivo evaluation for their impact on clinical disease course, white matter pathology, and inflammation. Both compounds ameliorated disease severity, increased mature OLs, and improved overall myelination in the corpus callosum and white matter tracts of the spinal cord. These effects were accompanied by reduced production of the OL toxic molecules interferon-γ and chemokine (C-X-C motif) ligand, CXCL10 by splenocytes with no discernable effect on central nervous system-infiltrating leukocyte numbers, while IndCl-o-methyl also reduced peripheral interleukin (IL)−17. In addition, expression of the chemokine CXCL1, which is associated with developmental oligodendrogenesis, was upregulated by IndCl and both analogues. Furthermore, callosal compound action potential recordings from analogue-treated mice demonstrated a larger N1 component amplitude compared to vehicle, suggesting more functionally myelinated fibers. Thus, the o-Methyl and o-Chloro IndCl analogues represent a class of ERβ ligands that offer significant remyelination and neuroprotection as well as modulation of the immune system; hence, they appear appropriate to consider further for therapeutic development in multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Hawra Karim
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Sung Hoon Kim
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Kelli Lauderdale
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Andrew S Lapato
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Kelley Atkinson
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Norio Yasui
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hana Yamate-Morgan
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | - Maria Sekyi
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA
| | | | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA, 92521, USA. .,Center for Glia Neuronal Interaction, UCR School of Medicine, Riverside, CA, 92521, USA.
| |
Collapse
|
14
|
Pepper RE, Pitman KA, Cullen CL, Young KM. How Do Cells of the Oligodendrocyte Lineage Affect Neuronal Circuits to Influence Motor Function, Memory and Mood? Front Cell Neurosci 2018; 12:399. [PMID: 30524235 PMCID: PMC6262292 DOI: 10.3389/fncel.2018.00399] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are immature cells in the central nervous system (CNS) that can rapidly respond to changes within their environment by modulating their proliferation, motility and differentiation. OPCs differentiate into myelinating oligodendrocytes throughout life, and both cell types have been implicated in maintaining and modulating neuronal function to affect motor performance, cognition and emotional state. However, questions remain about the mechanisms employed by OPCs and oligodendrocytes to regulate circuit function, including whether OPCs can only influence circuits through their generation of new oligodendrocytes, or can play other regulatory roles within the CNS. In this review, we detail the molecular and cellular mechanisms that allow OPCs, newborn oligodendrocytes and pre-existing oligodendrocytes to regulate circuit function and ultimately influence behavioral outcomes.
Collapse
Affiliation(s)
- Renee E Pepper
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
15
|
Wilmes AT, Reinehr S, Kühn S, Pedreiturria X, Petrikowski L, Faissner S, Ayzenberg I, Stute G, Gold R, Dick HB, Kleiter I, Joachim SC. Laquinimod protects the optic nerve and retina in an experimental autoimmune encephalomyelitis model. J Neuroinflammation 2018; 15:183. [PMID: 29903027 PMCID: PMC6002998 DOI: 10.1186/s12974-018-1208-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 05/20/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The oral immunomodulatory agent laquinimod is currently evaluated for multiple sclerosis (MS) treatment. Phase II and III studies demonstrated a reduction of degenerative processes. In addition to anti-inflammatory effects, laquinimod might have neuroprotective properties, but its impact on the visual system, which is often affected by MS, is unknown. The aim of our study was to investigate potential protective effects of laquinimod on the optic nerve and retina in an experimental autoimmune encephalomyelitis (EAE) model. METHODS We induced EAE in C57/BL6 mice via MOG35-55 immunization. Animals were divided into an untreated EAE group, three EAE groups receiving laquinimod (1, 5, or 25 mg/kg daily), starting the day post-immunization, and a non-immunized control group. Thirty days post-immunization, scotopic electroretinograms were carried out, and mice were sacrificed for histopathology (HE, LFB), immunohistochemistry (MBP, Iba1, Tmem119, F4/80, GFAP, vimentin, Brn-3a, cleaved caspase 3) of the optic nerve and retina, and retinal qRT-PCR analyses (Brn-3a, Iba1, Tmem119, AMWAP, CD68, GFAP). To evaluate the effect of a therapeutic approach, EAE animals were treated with 25 mg/kg laquinimod from day 16 when 60% of the animals had developed clinical signs of EAE. RESULTS Laquinimod reduced neurological EAE symptoms and improved the neuronal electrical output of the inner nuclear layer compared to untreated EAE mice. Furthermore, cellular infiltration, especially recruited phagocytes, and demyelination in the optic nerve were reduced. Microglia were diminished in optic nerve and retina. Retinal macroglial signal was reduced under treatment, whereas in the optic nerve macroglia were not affected. Additionally, laquinimod preserved retinal ganglion cells and reduced apoptosis. A later treatment with laquinimod in a therapeutic approach led to a reduction of clinical signs and to an improved b-wave amplitude. However, no changes in cellular infiltration and demyelination of the optic nerves were observed. Also, the number of retinal ganglion cells remained unaltered. CONCLUSION From our study, we deduce neuroprotective and anti-inflammatory effects of laquinimod on the optic nerve and retina in EAE mice, when animals were treated before any clinical signs were noted. Given the fact that the visual system is frequently affected by MS, the agent might be an interesting subject of further neuro-ophthalmic investigations.
Collapse
Affiliation(s)
- Anna T Wilmes
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Sandra Kühn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Xiomara Pedreiturria
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Laura Petrikowski
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Ilya Ayzenberg
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Ingo Kleiter
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, 44791, Bochum, Germany.
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany.
| |
Collapse
|
16
|
Increase in chemokine CXCL1 by ERβ ligand treatment is a key mediator in promoting axon myelination. Proc Natl Acad Sci U S A 2018; 115:6291-6296. [PMID: 29844175 PMCID: PMC6004485 DOI: 10.1073/pnas.1721732115] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Estrogen receptor β (ERβ) ligands promote remyelination in mouse models of multiple sclerosis. Recent work using experimental autoimmune encephalomyelitis (EAE) has shown that ERβ ligands induce axon remyelination, but impact peripheral inflammation to varying degrees. To identify if ERβ ligands initiate a common immune mechanism in remyelination, central and peripheral immunity and pathology in mice given ERβ ligands at peak EAE were assessed. All ERβ ligands induced differential expression of cytokines and chemokines, but increased levels of CXCL1 in the periphery and in astrocytes. Oligodendrocyte CXCR2 binds CXCL1 and has been implicated in normal myelination. In addition, despite extensive immune cell accumulation in the CNS, all ERβ ligands promoted extensive remyelination in mice at peak EAE. This finding highlights a component of the mechanism by which ERβ ligands mediate remyelination. Hence, interplay between the immune system and central nervous system may be responsible for the remyelinating effects of ERβ ligands. Our findings of potential neuroprotective benefits arising from the presence of CXCL1 could have implications for improved therapies for multiple sclerosis.
Collapse
|
17
|
Laquinimod ameliorates excitotoxic damage by regulating glutamate re-uptake. J Neuroinflammation 2018; 15:5. [PMID: 29304807 PMCID: PMC5756343 DOI: 10.1186/s12974-017-1048-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/21/2017] [Indexed: 11/10/2022] Open
Abstract
Background Laquinimod is an immunomodulatory drug under clinical investigation for the treatment of the progressive form of multiple sclerosis (MS) with both anti-inflammatory and neuroprotective effects. Excitotoxicity, a prominent pathophysiological feature of MS and of its animal model, experimental autoimmune encephalomyelitis (EAE), involves glutamate transporter (GluT) dysfunction in glial cells. The aim of this study was to assess whether laquinimod might exert direct neuroprotective effects by interfering with the mechanisms of excitotoxicity linked to GluT function impairments in EAE. Methods Osmotic minipumps allowing continuous intracerebroventricular (icv) infusion of laquinimod for 4 weeks were implanted into C57BL/6 mice before EAE induction. EAE cerebella were taken to perform western blot and qPCR experiments. For ex vivo experiments, EAE cerebellar slices were incubated with laquinimod before performing electrophysiology, western blot, and qPCR. Results In vivo treatment with laquinimod attenuated EAE clinical score at the peak of the disease, without remarkable effects on inflammatory markers. In vitro application of laquinimod to EAE cerebellar slices prevented EAE-linked glutamatergic alterations without mitigating astrogliosis and inflammation. Moreover, such treatment induced an increase of Slcla3 mRNA coding for the glial glutamate–aspartate transporter (GLAST) without affecting the protein content. Concomitantly, laquinimod significantly increased the levels of the glial glutamate transporter 1 (GLT-1) protein and pharmacological blockade of GLT-1 function fully abolished laquinimod anti-excitotoxic effect. Conclusions Overall, our results suggest that laquinimod protects against glutamate excitotoxicity of the cerebellum of EAE mice by bursting the expression of glial glutamate transporters, independently of its anti-inflammatory effects.
Collapse
|
18
|
Teske N, Liessem A, Fischbach F, Clarner T, Beyer C, Wruck C, Fragoulis A, Tauber SC, Victor M, Kipp M. Chemical hypoxia-induced integrated stress response activation in oligodendrocytes is mediated by the transcription factor nuclear factor (erythroid-derived 2)-like 2 (NRF2). J Neurochem 2018; 144:285-301. [PMID: 29210072 DOI: 10.1111/jnc.14270] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 11/18/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023]
Abstract
The extent of remyelination in multiple sclerosis lesions is often incomplete. Injury to oligodendrocyte progenitor cells can be a contributing factor for such incomplete remyelination. The precise mechanisms underlying insufficient repair remain to be defined, but oxidative stress appears to be involved. Here, we used immortalized oligodendrocyte cell lines as model systems to investigate a causal relation of oxidative stress and endoplasmic reticulum stress signaling cascades. OLN93 and OliNeu cells were subjected to chemical hypoxia by blocking the respiratory chain at various levels. Mitochondrial membrane potential and oxidative stress levels were quantified by flow cytometry. Endoplasmic reticulum stress was monitored by the expression induction of activating transcription factor 3 and 4 (Atf3, Atf4), DNA damage-inducible transcript 3 protein (Ddit3), and glucose-regulated protein 94. Lentiviral silencing of nuclear factor (erythroid-derived 2)-like 2 or kelch-like ECH-associated protein 1 was applied to study the relevance of NRF2 for endoplasmic reticulum stress responses. We demonstrate that inhibition of the respiratory chain induces oxidative stress in cultured oligodendrocytes which is paralleled by the expression induction of distinct mediators of the endoplasmic reticulum stress response, namely Atf3, Atf4, and Ddit3. Atf3 and Ddit3 expression induction is potentiated in kelch-like ECH-associated protein 1-deficient cells and absent in cells lacking the oxidative stress-related transcription factor NRF2. This study provides strong evidence that oxidative stress in oligodendrocytes activates endoplasmic reticulum stress response in a NRF2-dependent manner and, in consequence, might regulate oligodendrocyte degeneration in multiple sclerosis and other neurological disorders.
Collapse
Affiliation(s)
- Nico Teske
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Annette Liessem
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Felix Fischbach
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Tim Clarner
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy and JARA-BRAIN, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Christoph Wruck
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | | | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Marion Victor
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Markus Kipp
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
19
|
Ziemssen T, Tumani H, Sehr T, Thomas K, Paul F, Richter N, Samara E, Spiegelstein O, Sorani E, Bar-Ilan O, Mimrod D, Hayardeny L. Safety and in vivo immune assessment of escalating doses of oral laquinimod in patients with RRMS. J Neuroinflammation 2017; 14:172. [PMID: 28859672 PMCID: PMC5577769 DOI: 10.1186/s12974-017-0945-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/16/2017] [Indexed: 11/18/2022] Open
Abstract
Background Laquinimod is an oral immunomodulator in clinical development to treat relapsing-remitting multiple sclerosis (RRMS). Laquinimod is in clinical development for the treatment of multiple sclerosis and Huntington Disease (HD). The objective of this study is to assess the safety, tolerability, pharmacokinetics (PK) and cytoimmunologic effects following escalating doses of laquinimod in patients with RRMS. Methods One hundred twelve patients were randomly assigned to laquinimod/placebo in a series of separate dose-escalating cohorts starting from a daily oral dose of 0.9 mg/1.2 mg escalating to 2.7 mg, in 0.3 mg increments. Results Twenty-eight patients received placebo and 84 received laquinimod ranging from 0.9 to 2.7 mg. No deaths occurred. One serious adverse event (SAE) of perichondritis was reported, which was unrelated to laquinimod (0.9 mg). There was no increased incidence of adverse events (AEs) with escalating doses. Laquinimod-treated patients showed more abnormal laboratory levels in liver enzymes, P-amylase, C-reactive protein (CRP), and fibrinogen, but most shifts were clinically non-significant. The exposure of laquinimod was dose proportional and linear in the tested dose range. An immunological substudy showed significant dose-dependent decreases in 6-sulpho LacNAc + dendritic cell (slanDC) frequency following laquinimod compared to placebo. Conclusion Laquinimod doses up to 2.7 mg were safely administered to patients with RRMS. An in vivo effect of laquinimod on the innate immune system was demonstrated. Trial registration EudraCT Number: 2009-011234-99. Registered 23 June 2009. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0945-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tjalf Ziemssen
- Department of Neurology, MS Center Dresden, Center of Clinical Neuroscience, University Clinic Carl Gustav Carus Dresden, Dresden, Germany.
| | - Hayrettin Tumani
- Multiple Sclerosis Unit, Department of Neurology, University of Ulm, Ulm, Germany. .,Fachklinik für Neurologie Dietenbronn, Schwendi, Germany.
| | - Tony Sehr
- Department of Neurology, MS Center Dresden, Center of Clinical Neuroscience, University Clinic Carl Gustav Carus Dresden, Dresden, Germany
| | - Katja Thomas
- Department of Neurology, MS Center Dresden, Center of Clinical Neuroscience, University Clinic Carl Gustav Carus Dresden, Dresden, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center, Department of Neurology, Charité University Medicine, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité University Medicine Berlin, Berlin, Germany
| | - Nils Richter
- Neurologische Gemeinschaftspraxis, Düsseldorf, Germany
| | | | | | - Ella Sorani
- Teva Pharmaceutical Industries, Netanya, Israel
| | | | | | - Liat Hayardeny
- Teva Pharmaceutical Industries, Netanya, Israel.,Galmed Pharmaceuticals, Tel Aviv, Israel
| |
Collapse
|
20
|
Hussain RZ, Miller-Little WA, Lambracht-Washington D, Jaramillo TC, Takahashi M, Zhang S, Fu M, Cutter GR, Hayardeny L, Powell CM, Rosenberg RN, Stüve O. Laquinimod has no effects on brain volume or cellular CNS composition in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. J Neuroimmunol 2017; 309:100-110. [PMID: 28601278 DOI: 10.1016/j.jneuroim.2017.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Laquinimod is an anti-inflammatory agent with good central nervous system (CNS) bioavailability, and neuroprotective and myelorestorative properties. A clinical trial in patients with multiple sclerosis demonstrated that laquinimod significantly reduced loss of brain volume. The cellular substrate or molecular events underlying that treatment effect are unknown. In this study, we aimed to explore laquinimod's potential effects on brain volume, animal behavior, cellular numbers and composition of CNS-intrinsic cells and mononuclear cells within the CNS, amyloid beta (Aβ) accumulation and tau phosphorylation in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. METHODS Utilizing a dose response study design, four months old F1 3xTg-AD/C3H mice were treated for 10months between ages 4 and 14months with laquinimod (5, 10, or 25mg/kg), or PBS administered by oral gavage. Brain volumes were measured in a 7 Tesla magnetic resonance imager (MRI) at ages 4 and 14months. Behavioral testing included locomotor and rearing activity and the Morris water maze task. Cell numbers and immunophenotypes were assessed by multiparameter flow cytometry. Aβ deposition and tau phosphorylation were determined by immunohistochemistry. RESULTS In the F1 3xTg-AD/C3H animal model of AD, there was no detectable reduction of brain volume over a period of 10months of treatment, as there was not brain atrophy in any of the placebo or treatment groups. Laquinimod had no detectable effects on most neurobehavioral outcomes. The number or composition of CNS intrinsic cells and mononuclear subsets isolated from the CNS were not altered by laquinimod. CONCLUSION This is the first demonstration that there are no age-associated brain volume changes in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. Consequently, laquinimod had no effect on that outcome of this study. Most secondary outcomes on the effects of laquinimod on behavior and the number and composition of CNS-intrinsic cells and mononuclear cells within the CNS were also negative.
Collapse
Affiliation(s)
- Rehana Z Hussain
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - William A Miller-Little
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Tom C Jaramillo
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Masaya Takahashi
- Department of Radiology, University of Texas Southwestern Medical Center at Dallas, USA; Advanced Imaging Center, University of Texas Southwestern Medical Center at Dallas, USA
| | - Shanrong Zhang
- Advanced Imaging Center, University of Texas Southwestern Medical Center at Dallas, USA
| | - Min Fu
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Gary R Cutter
- Department of Biostatistics, University of Alabama at Birmingham, USA
| | | | - Craig M Powell
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany; Neurology Section, VA North Texas Health Care System, Medical Service Dallas, VA Medical Center, USA.
| |
Collapse
|
21
|
Hasselmann JPC, Karim H, Khalaj AJ, Ghosh S, Tiwari-Woodruff SK. Consistent induction of chronic experimental autoimmune encephalomyelitis in C57BL/6 mice for the longitudinal study of pathology and repair. J Neurosci Methods 2017; 284:71-84. [PMID: 28396177 DOI: 10.1016/j.jneumeth.2017.04.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 03/15/2017] [Accepted: 04/04/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND While many groups use experimental autoimmune encephalomyelitis (EAE) as a model to uncover therapeutic targets and understand the pathology underlying multiple sclerosis (MS), EAE protocol variability introduces discrepancies in central nervous system (CNS) pathogenesis and clinical disease, limiting the comparability between studies and slowing much-needed translational research. OPTIMIZED METHOD Here we describe a detailed, reliable protocol for chronic EAE induction in C57BL/6 mice utilizing two injections of myelin oligodendrocyte glycoprotein (35-55) peptide mixed with complete Freund's adjuvant and paired with pertussis toxin. RESULTS The active MOG35-55-EAE protocol presented here induces ascending paralysis in 80-100% of immunized mice. We observe: (1) consistent T cell immune activation, (2) robust CNS infiltration by peripheral immune cells, and (3) perivascular demyelinating lesions concurrent with axon damage in the spinal cord and various brain regions, including the optic nerve, cortex, hippocampus, internal capsule, and cerebellum. COMPARISON WITH EXISTING METHOD(S) Lack of detailed protocols, combined with variability between laboratories, make EAE results difficult to compare and hinder the use of this model for therapeutic development. We provide the most detailed active MOG35-55-EAE protocol to date. With this protocol, we observe high disease incidence and a consistent, reliable disease course. The resulting pathology is MS-like and includes optic neuritis, perivascular mononuclear infiltration, CNS axon demyelination, and axon damage in both infiltrating lesions and otherwise normal-appearing white matter. CONCLUSIONS By providing a detailed active MOG35-55-EAE protocol that yields consistent and robust pathology, we aim to foster comparability between pre-clinical studies and facilitate the discovery of MS therapeutics.
Collapse
Affiliation(s)
| | - Hawra Karim
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA 92521, USA
| | - Anna J Khalaj
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA 92521, USA
| | - Subir Ghosh
- Department of Statistics, UCR-CNAS, Riverside, CA 92521, USA
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, CA 92521, USA; Department of Neuroscience, UCR School of Medicine, Riverside, CA 92521, USA; Center for Glial-Neuronal Interactions, UCR School of Medicine, CA 92506, USA.
| |
Collapse
|
22
|
Matrine promotes NT3 expression in CNS cells in experimental autoimmune encephalomyelitis. Neurosci Lett 2017; 649:100-106. [PMID: 28392360 DOI: 10.1016/j.neulet.2017.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/30/2017] [Accepted: 04/02/2017] [Indexed: 12/12/2022]
Abstract
Neurotrophin 3 (NT3) is a potent neurotrophic factor for promoting remyelination and recovery of neuronal function; upregulation of its expression in the central nervous system (CNS) is thus of major therapeutic importance for neurological deficits. Matrine (MAT), a quinolizidine alkaloid derived from the herb Radix Sophorae Flavescent, has been recently reported to effectively ameliorate clinical signs in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), by secreting antiinflammatory cytokines. In the present study, our goal was to investigate whether MAT could affect NT3 expression of glial cells in the CNS, the major cell populations in the CNS foci of MS/EAE. We found that MAT markedly upregulated NT3 expression in the CNS not only by microglia/macrophages and astrocytes, but also by oligodendrocyte precursor cells, indicative of both paracrine and autocrine effects on myelinating cells. While MAT treatment reduced the numbers of iNOS+ M1, but increased Arg1+ M2 microglia/macrophage phenotypes, NT3 expression was upregulated in both phenotypes. These results indicate that MAT therapy for EAE acts, at least in part, by stimulating local production of NT3 by glial cells in the CNS, which protects neural cells from CNS inflammation-induced tissue damage.
Collapse
|
23
|
Dulamea AO. Role of Oligodendrocyte Dysfunction in Demyelination, Remyelination and Neurodegeneration in Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 958:91-127. [PMID: 28093710 DOI: 10.1007/978-3-319-47861-6_7] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS) during development and throughout adulthood. They result from a complex and well controlled process of activation, proliferation, migration and differentiation of oligodendrocyte progenitor cells (OPCs) from the germinative niches of the CNS. In multiple sclerosis (MS), the complex pathological process produces dysfunction and apoptosis of OLs leading to demyelination and neurodegeneration. This review attempts to describe the patterns of demyelination in MS, the steps involved in oligodendrogenesis and myelination in healthy CNS, the different pathways leading to OLs and myelin loss in MS, as well as principles involved in restoration of myelin sheaths. Environmental factors and their impact on OLs and pathological mechanisms of MS are also discussed. Finally, we will present evidence about the potential therapeutic targets in re-myelination processes that can be accessed in order to develop regenerative therapies for MS.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- Neurology Clinic, University of Medicine and Pharmacy "Carol Davila", Fundeni Clinical Institute, Building A, Neurology Clinic, Room 201, 022328, Bucharest, Romania.
| |
Collapse
|
24
|
Matías-Guiu J, Gomez-Pinedo U, Matias-Guiu JA. News in multiple sclerosis: Remyelination as a therapeutic target. Med Clin (Barc) 2016; 148:377-380. [PMID: 27923464 DOI: 10.1016/j.medcli.2016.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 01/26/2023]
Affiliation(s)
- Jorge Matías-Guiu
- Servicio de Neurología, Instituto de Neurociencias, Hospital Clínico San Carlos, Universidad Complutense, Instituto de Investigación Sanitaria San Carlos (idiSSC), Madrid, España.
| | - Ulises Gomez-Pinedo
- Servicio de Neurología, Instituto de Neurociencias, Hospital Clínico San Carlos, Universidad Complutense, Instituto de Investigación Sanitaria San Carlos (idiSSC), Madrid, España
| | - Jordi A Matias-Guiu
- Servicio de Neurología, Instituto de Neurociencias, Hospital Clínico San Carlos, Universidad Complutense, Instituto de Investigación Sanitaria San Carlos (idiSSC), Madrid, España
| |
Collapse
|
25
|
Laquinimod arrests experimental autoimmune encephalomyelitis by activating the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 2016; 113:E6145-E6152. [PMID: 27671624 DOI: 10.1073/pnas.1607843113] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Laquinimod is an oral drug currently being evaluated for the treatment of relapsing, remitting, and primary progressive multiple sclerosis and Huntington's disease. Laquinimod exerts beneficial activities on both the peripheral immune system and the CNS with distinctive changes in CNS resident cell populations, especially astrocytes and microglia. Analysis of genome-wide expression data revealed activation of the aryl hydrocarbon receptor (AhR) pathway in laquinimod-treated mice. The AhR pathway modulates the differentiation and function of several cell populations, many of which play an important role in neuroinflammation. We therefore tested the consequences of AhR activation in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) using AhR knockout mice. We demonstrate that the pronounced effect of laquinimod on clinical score, CNS inflammation, and demyelination in EAE was abolished in AhR-/- mice. Furthermore, using bone marrow chimeras we show that deletion of AhR in the immune system fully abrogates, whereas deletion within the CNS partially abrogates the effect of laquinimod in EAE. These data strongly support the idea that AhR is necessary for the efficacy of laquinimod in EAE and that laquinimod may represent a first-in-class drug targeting AhR for the treatment of multiple sclerosis and other neurodegenerative diseases.
Collapse
|
26
|
Berg J, Mahmoudjanlou Y, Duscha A, Massa MG, Thöne J, Esser C, Gold R, Haghikia A. The immunomodulatory effect of laquinimod in CNS autoimmunity is mediated by the aryl hydrocarbon receptor. J Neuroimmunol 2016; 298:9-15. [DOI: 10.1016/j.jneuroim.2016.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/08/2016] [Indexed: 01/16/2023]
|
27
|
Laquinimod rescues striatal, cortical and white matter pathology and results in modest behavioural improvements in the YAC128 model of Huntington disease. Sci Rep 2016; 6:31652. [PMID: 27528441 PMCID: PMC4985819 DOI: 10.1038/srep31652] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022] Open
Abstract
Increasing evidence supports a role for abnormal immune activation and inflammatory responses in Huntington disease (HD). In this study, we evaluated the therapeutic potential of laquinimod (1 and 10 mg/kg), a novel immunomodulatory agent shown to be protective in a number of neuroinflammatory conditions, in the YAC128 mouse model of HD. Treatment with laquinimod for 6 months rescued atrophy in the striatum, in certain cortical regions, and in the corpus callosum of YAC128 HD mice. Diffusion tensor imaging showed that white matter microstructural abnormalities in the posterior corpus callosum were improved following treatment with low dose (1 mg/kg) laquinimod, and were paralleled by reduced levels of interleukin-6 in the periphery of YAC128 HD mice. Functionally, treatment with laquinimod (1 and 10 mg/kg) led to modest improvements in motor function and in depressive-like behaviour. Taken together, these results suggest that laquinimod may improve some features of pathology in HD, and provides support for the role of immune activation in the pathogenesis of HD.
Collapse
|
28
|
Hainke U, Thomas K, Ziemssen T. Laquinimod in the treatment of relapsing remitting multiple sclerosis. Expert Opin Drug Metab Toxicol 2016; 12:701-9. [PMID: 27089834 DOI: 10.1080/17425255.2016.1179279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Laquinimod is a new once-daily oral administrable agent, which is under investigation in a phase 3 clinical trial for relapsing remitting multiple sclerosis (RRMS) and in a phase 2 clinical trial for primary progressive MS (PPMS). AREAS COVERED The pharmacokinetic, pharmacodynamic and the safety profiles of laquinimod are covered in this review. In preclinical studies, the ability to prevent both experimental autoimmune encephalomyelitis and experimental autoimmune neuritis has been demonstrated. Reduced cell infiltration, demyelination, axonal damage and a shift of T-helper cell responses have been shown. Accordingly, in human studies, a decrease of pro-inflammatory and an increase of anti-inflammatory cytokines have been measured and a significant reduction of disease progression and a decrease in brain volume loss has been demonstrated. During all clinical studies a favorable safety profile was observed for 0.6mg laquinimod. New information about cardiovascular events is prompting the discontinuation of higher dosing regimens in both ongoing trials. EXPERT OPINION Laquinimod is a first in class oral agent with high potential to reduce disease progression in RRMS and PPMS. Owing to its favorable safety profile, a combination with 0.6mg laquinimod and other disease modifying therapies could be an option in future MS therapy.
Collapse
Affiliation(s)
- Undine Hainke
- a Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus , Dresden University of Technology , Dresden , Germany
| | - Katja Thomas
- a Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus , Dresden University of Technology , Dresden , Germany
| | - Tjalf Ziemssen
- a Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus , Dresden University of Technology , Dresden , Germany
| |
Collapse
|
29
|
Harlow DE, Honce JM, Miravalle AA. Remyelination Therapy in Multiple Sclerosis. Front Neurol 2015; 6:257. [PMID: 26696956 PMCID: PMC4674562 DOI: 10.3389/fneur.2015.00257] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/23/2015] [Indexed: 01/10/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disorder of the central nervous system that results in destruction of the myelin sheath that surrounds axons and eventual neurodegeneration. Current treatments approved for the treatment of relapsing forms of MS target the aberrant immune response and successfully reduce the severity of attacks and frequency of relapses. Therapies are still needed that can repair damage particularly for the treatment of progressive forms of MS for which current therapies are relatively ineffective. Remyelination can restore neuronal function and prevent further neuronal loss and clinical disability. Recent advancements in our understanding of the molecular and cellular mechanisms regulating myelination, as well as the development of high-throughput screens to identify agents that enhance myelination, have lead to the identification of many potential remyelination therapies currently in preclinical and early clinical development. One problem that has plagued the development of treatments to promote remyelination is the difficulty in assessing remyelination in patients with current imaging techniques. Powerful new imaging technologies are making it easier to discern remyelination in patients, which is critical for the assessment of these new therapeutic strategies during clinical trials. This review will summarize what is currently known about remyelination failure in MS, strategies to overcome this failure, new therapeutic treatments in the pipeline for promoting remyelination in MS patients, and new imaging technologies for measuring remyelination in patients.
Collapse
Affiliation(s)
- Danielle E Harlow
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus , Aurora, CO , USA
| | - Justin M Honce
- Department of Radiology, University of Colorado Anschutz Medical Campus , Aurora, CO , USA
| | - Augusto A Miravalle
- Department of Neurology, University of Colorado Anschutz Medical Campus , Aurora, CO , USA
| |
Collapse
|
30
|
Mandolesi G, Gentile A, Musella A, Fresegna D, De Vito F, Bullitta S, Sepman H, Marfia GA, Centonze D. Synaptopathy connects inflammation and neurodegeneration in multiple sclerosis. Nat Rev Neurol 2015; 11:711-24. [PMID: 26585978 DOI: 10.1038/nrneurol.2015.222] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis (MS) has long been regarded as a chronic inflammatory disease of the white matter that leads to demyelination and eventually to neurodegeneration. In the past decade, several aspects of MS pathogenesis have been challenged, and degenerative changes of the grey matter, which are independent of demyelination, have become a topic of interest. CNS inflammation in MS and experimental autoimmune encephalomyelitis (EAE; a disease model used to study MS in rodents) causes a marked imbalance between GABAergic and glutamatergic transmission, and a loss of synapses, all of which leads to a diffuse 'synaptopathy'. Altered synaptic transmission can occur early in MS and EAE, independently of demyelination and axonal loss, and subsequently causes excitotoxic damage. Inflammation-driven synaptic abnormalities are emerging as a prominent pathogenic mechanism in MS-importantly, they are potentially reversible and, therefore, represent attractive therapeutic targets. In this Review, we focus on the connection between inflammation and synaptopathy in MS and EAE, which sheds light not only on the pathophysiology of MS but also on that of primary neurodegenerative disorders in which inflammatory processes contribute to disease progression.
Collapse
Affiliation(s)
- Georgia Mandolesi
- IRCCS Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Antonietta Gentile
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Alessandra Musella
- IRCCS Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Diego Fresegna
- IRCCS Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Francesca De Vito
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Silvia Bullitta
- IRCCS Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Helena Sepman
- IRCCS Fondazione Santa Lucia/Centro Europeo per la Ricerca sul Cervello (CERC), Via del Fosso di Fiorano 64, 00143 Rome, Italy.,Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Girolama A Marfia
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Diego Centonze
- IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| |
Collapse
|
31
|
Stasiolek M, Linker RA, Hayardeny L, Bar Ilan O, Gold R. Immune parameters of patients treated with laquinimod, a novel oral therapy for the treatment of multiple sclerosis: results from a double-blind placebo-controlled study. IMMUNITY INFLAMMATION AND DISEASE 2015; 3:45-55. [PMID: 26029365 PMCID: PMC4444148 DOI: 10.1002/iid3.42] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 11/11/2022]
Abstract
Laquinimod is a novel orally administered drug for the treatment of relapsing remitting multiple sclerosis (RRMS). In this immunological substudy of the phase III Assessment of Oral Laquinimod in Preventing Progression of MS (ALLEGRO) trial, we performed an ex vivo and in vitro analysis of effects exerted by laquinimod on peripheral blood immune cell populations from RRMS patients with a special focus on monocyte phenotype and function. Approximately 100 patients were enrolled following a standardized protocol. Half of the patients received laquinimod and the other half received placebo. Peripheral blood samples were collected prior to commencement of therapy and after 1, 3, 6, 12, and 24 months of continuous therapy. Main lymphocytic and antigen presenting cell fractions were analyzed in peripheral blood mononuclear cells (PBMCs) ex vivo by flow cytometry. The proliferative response of PBMCs to mitogen or recall antigen was assessed in culture experiments. Untouched monocytes were sorted magnetically and cultured under pro-inflammatory conditions. PBMC analysis showed no significant differences of investigated lymphocytic and antigen presenting cell populations over time within each group, or between the two groups. However, the detailed in vitro analysis of monocytes demonstrated a lower level of CD86 expression on monocytes stimulated with LPS in laquinimod patients beginning from the 1st month of treatment. Upon pro-inflammatory stimulation, monocytes obtained from laquinimod treated patients tended to secrete lower levels of the proinflammatory chemokines CCL2 or CCL5. Taken together, in this prospective study, we demonstrate immune modulation but no immunosuppressive biological activity of laquinimod in a large group of MS patients.
Collapse
Affiliation(s)
- Mariusz Stasiolek
- Department of Neurology, Polish Mother's Memorial Hospital-Research Institute Lodz, Poland
| | - Ralf A Linker
- Department of Neurology, Ruhr-University Bochum St. Josef-Hospital, Bochum, Germany
| | - Liat Hayardeny
- Teva Innovative Research and Development Group, Teva Pharmaceutical Industries Ltd 5 Bazel Street, Petah, Tiqva, 49131, Israel
| | - Oren Bar Ilan
- Teva Innovative Research and Development Group, Teva Pharmaceutical Industries Ltd 5 Bazel Street, Petah, Tiqva, 49131, Israel
| | - Ralf Gold
- Department of Neurology, Ruhr-University Bochum St. Josef-Hospital, Bochum, Germany
| |
Collapse
|
32
|
Williams A. Remyelination in multiple sclerosis: what do we know and where are we going? Neurodegener Dis Manag 2015; 5:49-59. [PMID: 25711454 DOI: 10.2217/nmt.14.40] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) patients today have more hope of a good disease outcome with an ever-increasing choice of immunomodulatory therapies to reduce disease relapses, thought to be caused by inflammation within the CNS, leading to axonal demyelination. However, although there has been much progress in this disease phase, there has been little impact on the progressive phase of MS, when neurodegeneration dominates and patients accumulate disability over years. This failure of prevention of progressive disease has led to a frame-shift in research thinking, focusing on neuroprotective strategies such as promotion of remyelination, to be used alongside immunomodulatory therapies. This review discusses this unmet need in MS, in terms of pathology and current knowledge of remyelination and proremyelinating therapies.
Collapse
|
33
|
Multiple functional therapeutic effects of the estrogen receptor β agonist indazole-Cl in a mouse model of multiple sclerosis. Proc Natl Acad Sci U S A 2014; 111:18061-6. [PMID: 25453074 DOI: 10.1073/pnas.1411294111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Currently available immunomodulatory therapies do not stop the pathogenesis underlying multiple sclerosis (MS) and are only partially effective in preventing the onset of permanent disability in patients with MS. Identifying a drug that stimulates endogenous remyelination and/or minimizes axonal degeneration would reduce the rate and degree of disease progression. Here, the effects of the highly selective estrogen receptor (ER) β agonist indazole chloride (Ind-Cl) on functional remyelination in chronic experimental autoimmune encephalomyelitis (EAE) mice were investigated by assessing pathologic, functional, and behavioral consequences of both prophylactic and therapeutic (peak EAE) treatment with Ind-Cl. Peripheral cytokines from autoantigen-stimulated splenocytes were measured, and central nervous system infiltration by immune cells, axon health, and myelination were assessed by immunohistochemistry and electron microscopy. Therapeutic Ind-Cl improved clinical disease and rotorod performance and also decreased peripheral Th1 cytokines and reactive astrocytes, activated microglia, and T cells in brains of EAE mice. Increased callosal myelination and mature oligodendrocytes correlated with improved callosal conduction and refractoriness. Therapeutic Ind-Cl-induced remyelination was independent of its effects on the immune system, as Ind-Cl increased remyelination within the cuprizone diet-induced demyelinating model. We conclude that Ind-Cl is a refined pharmacologic agent capable of stimulating functionally relevant endogenous myelination, with important implications for progressive MS treatment.
Collapse
|
34
|
Moore S, Khalaj AJ, Patel R, Yoon J, Ichwan D, Hayardeny L, Tiwari-Woodruff SK. Restoration of axon conduction and motor deficits by therapeutic treatment with glatiramer acetate. J Neurosci Res 2014; 92:1621-36. [PMID: 24989965 PMCID: PMC4305217 DOI: 10.1002/jnr.23440] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/25/2014] [Accepted: 05/27/2014] [Indexed: 12/20/2022]
Abstract
Glatiramer acetate (GA; Copaxone) is an approved drug for the treatment of multiple sclerosis (MS). The underlying multifactorial anti-inflammatory, neuroprotective effect of GA is in the induction of reactive T cells that release immunomodulatory cytokines and neurotrophic factors at the injury site. These GA-induced cytokines and growth factors may have a direct effect on axon function. Building on previous findings that suggest a neuroprotective effect of GA, we assessed the therapeutic effects of GA on brain and spinal cord pathology and functional correlates using the chronic experimental autoimmune encephalomyelitis (EAE) mouse model of MS. Therapeutic regimens were utilized based on promising prophylactic efficacy. More specifically, C57BL/6 mice were treated with 2 mg/mouse/day GA for 8 days beginning at various time points after EAE post-induction day 15, yielding a thorough, clinically relevant assessment of GA efficacy within the context of severe progressive disease. Therapeutic treatment with GA significantly decreased clinical scores and improved rotorod motor performance in EAE mice. These functional improvements were supported by an increase in myelinated axons and fewer amyloid precursor protein-positive axons in the spinal cords of GA-treated EAE mice. Furthermore, therapeutic GA decreased microglia/macrophage and T cell infiltrates and increased oligodendrocyte numbers in both the spinal cord and corpus callosum of EAE mice. Finally, GA improved callosal axon conduction and nodal protein organization in EAE. Our results demonstrate that therapeutic GA treatment has significant beneficial effects in a chronic mouse model of MS, in which its positive effects on both myelinated and non-myelinated axons results in improved axon function.
Collapse
Affiliation(s)
- Spencer Moore
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Anna J Khalaj
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Rhusheet Patel
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - JaeHee Yoon
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Daniel Ichwan
- Department of Neurology, UCLA School of MedicineLos Angeles, California
| | - Liat Hayardeny
- Pharmacology Unit, Global Innovative Research and Development, Teva Pharmaceutical IndustriesNetanya, Israel
| | - Seema K Tiwari-Woodruff
- Department of Neurology, UCLA School of MedicineLos Angeles, California
- Brain Research Institute, UCLA School of MedicineLos Angeles, California
| |
Collapse
|
35
|
Kelland EE, Gilmore W, Hayardeny L, Weiner LP, Lund BT. In vitro assessment of the direct effect of laquinimod on basic functions of human neural stem cells and oligodendrocyte progenitor cells. J Neurol Sci 2014; 346:66-74. [PMID: 25125045 DOI: 10.1016/j.jns.2014.07.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 06/19/2014] [Accepted: 07/28/2014] [Indexed: 12/21/2022]
Abstract
Laquinimod is an orally active immunomodulatory small molecule that has shown clear clinical benefit in trials for relapsing-remitting multiple sclerosis and in experimental rodent models that emulate multiple sclerosis (MS). Studies in healthy mice, and in mice with experimental autoimmune encephalomyelitis, have demonstrated that laquinimod is capable of entering the central nervous system. It is therefore important to determine if laquinimod is capable of a direct influence on basic functions of neural stem cells (NSC) or oligodendrocyte progenitor cells (OPC)--cells critical for myelin repair in MS. In order to address this question, a series of experiments was conducted to determine the effect of exogenous laquinimod on viability, proliferation, migration and differentiation of human NSC and OPC in vitro. These data show, for the first time in cells of human origin, that direct, short-term interaction between laquinimod and NSC or OPC, in an isolated in vitro setting, is not detrimental to the basic cellular function of these cells.
Collapse
Affiliation(s)
- Eve E Kelland
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Wendy Gilmore
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Liat Hayardeny
- Pharmacology Unit, Global Innovative R&D, Teva Pharmaceutical Industries, Netanya, Israel
| | - Leslie P Weiner
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brett T Lund
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
36
|
Mishra MK, Wang J, Keough MB, Fan Y, Silva C, Sloka S, Hayardeny L, Brück W, Yong VW. Laquinimod reduces neuroaxonal injury through inhibiting microglial activation. Ann Clin Transl Neurol 2014; 1:409-22. [PMID: 25356411 PMCID: PMC4184669 DOI: 10.1002/acn3.67] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/19/2014] [Accepted: 04/22/2014] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Laquinimod is an emerging oral medication for multiple sclerosis (MS) that reduces brain atrophy and progression of disability in two Phase III clinical trials. The mechanism of these effects is unclear. Persistent activation of microglia occurs in MS and contributes to injury. Thus, we investigated whether laquinimod alters properties of microglia in culture and in experimental autoimmune encephalomyelitis (EAE), and whether this reduces neurodegeneration. METHODS Microglia were cultured from human brains. EAE was induced in mice. RESULTS The activation of human microglia increased levels of several pro- and anti-inflammatory cytokines and these elevations were attenuated by pretreatment with laquinimod. Laquinimod prevented the decline in activated microglia of miR124a, a microRNA implicated in maintaining microglia quiescence, and reduced the activity of several signaling pathways (Jun-N-terminal kinase, ribosomal S6 kinase, and AKT/protein kinase B) in activated microglia. In EAE, axonal injury correlated with accumulation of microglia/macrophages in the spinal cord. EAE mice treated with laquinimod before onset of clinical signs subsequently had reduced microglia/macrophage density and axonal injury. Remarkably, when laquinimod treatment was initiated well into the disease course, the progressive demyelination, and axonal loss was halted. Besides inflammatory molecules associated with microglia, the level of inducible nitric oxide (NO) synthase capable of producing free radical toxicity was attenuated by laquinimod in EAE mice. Finally, in coculture where microglia activation caused neuronal death, laquinimod decreased NO levels, and neurotoxicity. INTERPRETATION Laquinimod is a novel inhibitor of microglial activation that lowers microglia-induced neuronal death in culture and axonal injury/loss in EAE.
Collapse
Affiliation(s)
- Manoj Kumar Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Janet Wang
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Michael B Keough
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Yan Fan
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Claudia Silva
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Scott Sloka
- Grand River Hospital Kitchener, Ontario, Canada
| | | | - Wolfgang Brück
- Department of Neuropathology, University Medical Center Göttingen, Germany
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| |
Collapse
|
37
|
Chen L, Coleman R, Leang R, Tran H, Kopf A, Walsh CM, Sears-Kraxberger I, Steward O, Macklin WB, Loring JF, Lane TE. Human neural precursor cells promote neurologic recovery in a viral model of multiple sclerosis. Stem Cell Reports 2014; 2:825-37. [PMID: 24936469 PMCID: PMC4050357 DOI: 10.1016/j.stemcr.2014.04.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 12/21/2022] Open
Abstract
Using a viral model of the demyelinating disease multiple sclerosis (MS), we show that intraspinal transplantation of human embryonic stem cell-derived neural precursor cells (hNPCs) results in sustained clinical recovery, although hNPCs were not detectable beyond day 8 posttransplantation. Improved motor skills were associated with a reduction in neuroinflammation, decreased demyelination, and enhanced remyelination. Evidence indicates that the reduced neuroinflammation is correlated with an increased number of CD4+CD25+FOXP3+ regulatory T cells (Tregs) within the spinal cords. Coculture of hNPCs with activated T cells resulted in reduced T cell proliferation and increased Treg numbers. The hNPCs acted, in part, through secretion of TGF-β1 and TGF-β2. These findings indicate that the transient presence of hNPCs transplanted in an animal model of MS has powerful immunomodulatory effects and mediates recovery. Further investigation of the restorative effects of hNPC transplantation may aid in the development of clinically relevant MS treatments. Spinal cord transplantation of hNPCs results in recovery in a viral model of MS hNPC-mediated recovery occurs in the absence of engrafted cells hNPCs are immunomodulatory through increasing the frequency of Tregs in the CNS hNPCs increase Treg frequency via a TGF-β1- and TGF-β2-dependent pathway
Collapse
Affiliation(s)
- Lu Chen
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Ronald Coleman
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ronika Leang
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Ha Tran
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alexandra Kopf
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Craig M Walsh
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Ilse Sears-Kraxberger
- Reeve-Irvine Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Oswald Steward
- Reeve-Irvine Research Center, Departments of Anatomy & Neurobiology, Neurobiology & Behavior, and Neurosurgery, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Jeanne F Loring
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas E Lane
- Department of Molecular Biology and Biochemistry, Sue and Bill Gross Stem Cell Center, Multiple Sclerosis Research Center, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|