51
|
Stereological Investigation of Regional Brain Volumes after Acute and Chronic Cuprizone-Induced Demyelination. Cells 2019; 8:cells8091024. [PMID: 31484353 PMCID: PMC6770802 DOI: 10.3390/cells8091024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/19/2019] [Accepted: 08/30/2019] [Indexed: 02/03/2023] Open
Abstract
Brain volume measurement is one of the most frequently used biomarkers to establish neuroprotective effects during pre-clinical multiple sclerosis (MS) studies. Furthermore, whole-brain atrophy estimates in MS correlate more robustly with clinical disability than traditional, lesion-based metrics. However, the underlying mechanisms leading to brain atrophy are poorly understood, partly due to the lack of appropriate animal models to study this aspect of the disease. The purpose of this study was to assess brain volumes and neuro-axonal degeneration after acute and chronic cuprizone-induced demyelination. C57BL/6 male mice were intoxicated with cuprizone for up to 12 weeks. Brain volume, as well as total numbers and densities of neurons, were determined using design-based stereology. After five weeks of cuprizone intoxication, despite severe demyelination, brain volumes were not altered at this time point. After 12 weeks of cuprizone intoxication, a significant volume reduction was found in the corpus callosum and diverse subcortical areas, particularly the internal capsule and the thalamus. Thalamic volume loss was accompanied by glucose hypermetabolism, analyzed by [18F]-fluoro-2-deoxy-d-glucose (18F-FDG) positron-emission tomography. This study demonstrates region-specific brain atrophy of different subcortical brain regions after chronic cuprizone-induced demyelination. The chronic cuprizone demyelination model in male mice is, thus, a useful tool to study the underlying mechanisms of subcortical brain atrophy and to investigate the effectiveness of therapeutic interventions.
Collapse
|
52
|
Mandolesi G, Bullitta S, Fresegna D, De Vito F, Rizzo FR, Musella A, Guadalupi L, Vanni V, Stampanoni Bassi M, Buttari F, Viscomi MT, Centonze D, Gentile A. Voluntary running wheel attenuates motor deterioration and brain damage in cuprizone-induced demyelination. Neurobiol Dis 2019; 129:102-117. [DOI: 10.1016/j.nbd.2019.05.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/23/2018] [Accepted: 05/13/2019] [Indexed: 12/27/2022] Open
|
53
|
Zhang J, Fox H, Xiong H. Severer nodular lesion in white matter than in gray matter in simian immunodeficiency virus-infected monkey, but not closely correlated with viral infection. J Biomed Res 2019; 34:292-300. [PMID: 32525497 PMCID: PMC7386408 DOI: 10.7555/jbr.33.20180047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Immune cell accumulation and white matter anomaly are common features of HIV (human immunodeficiency virus) -infected patients in combination antiretroviral therapy (cART) era. Neuroimaging tests on cART treated patients displayed prominent diffuse white matter lesions. Notably, immune cell nodular lesion (NL) was a conspicuous type of pathological change in HIV/SIV (simian immunodeficiency virus) infected brain before cART. Therefore, we used SIV infected brain to investigate the distribution of those NLs in gray and white matters. We found a significant higher number of NLs in white matter than that in gray matter. However, virus infection correlated with macrophage NLs but not with microglia NLs, especially in white matter. In addition, NLs interrupted white matter integrity more severely, since even tiny nodules could disconnect nerve fibers in white matter tracts. In the gray matter with dense myelinated axons, NLs obviously encroached those fibers; in the area of few myelinated axons, small nodules well co-localized with extracellular matrix between neurons.
Collapse
Affiliation(s)
- Jingdong Zhang
- Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Howard Fox
- Department of Pharmacology and Experiment Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Huangui Xiong
- Department of Pharmacology and Experiment Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
54
|
Oligodendrocyte degeneration and concomitant microglia activation directs peripheral immune cells into the forebrain. Neurochem Int 2019; 126:139-153. [PMID: 30867127 DOI: 10.1016/j.neuint.2019.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/21/2019] [Accepted: 03/06/2019] [Indexed: 11/22/2022]
Abstract
Brain-intrinsic degenerative cascades are a proposed factor driving inflammatory lesion formation in multiple sclerosis (MS) patients. We recently showed that encephalitogenic lymphocytes are recruited to the sites of active demyelination induced by cuprizone. Here, we investigated whether cuprizone-induced oligodendrocyte and myelin pathology is sufficient to trigger peripheral immune cell recruitment into the forebrain. We show that early cuprizone-induced white matter lesions display a striking similarity to early MS lesions, i.e., oligodendrocyte degeneration, microglia activation and absence of severe lymphocyte infiltration. Such early cuprizone lesions are sufficient to trigger peripheral immune cell recruitment secondary to subsequent EAE (experimental autoimmune encephalomyelitis) induction. The lesions are characterized by discontinuation of the perivascular glia limitans, focal axonal damage, and perivascular astrocyte pathology. Time course studies showed that the severity of cuprizone-induced lesions positively correlates with the extent of peripheral immune cell recruitment. Furthermore, results of genome-wide array analyses suggest that moesin is integral for early microglia activation in cuprizone and MS lesions. This study underpins the significance of brain-intrinsic degenerative cascades for immune cell recruitment and, in consequence, MS lesion formation.
Collapse
|
55
|
Kramann N, Menken L, Pförtner R, Schmid SN, Stadelmann C, Wegner C, Brück W. Glial fibrillary acidic protein expression alters astrocytic chemokine release and protects mice from cuprizone-induced demyelination. Glia 2019; 67:1308-1319. [DOI: 10.1002/glia.23605] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Nadine Kramann
- Institute of Neuropathology, University Medical Center Göttingen; Göttingen Germany
| | - Lena Menken
- Institute of Neuropathology, University Medical Center Göttingen; Göttingen Germany
| | - Ramona Pförtner
- Institute of Neuropathology, University Medical Center Göttingen; Göttingen Germany
| | - Susanne N. Schmid
- Institute of Neuropathology, University Medical Center Göttingen; Göttingen Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen; Göttingen Germany
| | - Christiane Wegner
- Institute of Neuropathology, University Medical Center Göttingen; Göttingen Germany
- Institute of Pathology, University Medical Center Göttingen; Göttingen Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Göttingen; Göttingen Germany
| |
Collapse
|
56
|
Wlodarczyk A, Benmamar-Badel A, Cédile O, Jensen KN, Kramer I, Elsborg NB, Owens T. CSF1R Stimulation Promotes Increased Neuroprotection by CD11c+ Microglia in EAE. Front Cell Neurosci 2019; 12:523. [PMID: 30687013 PMCID: PMC6335250 DOI: 10.3389/fncel.2018.00523] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/14/2018] [Indexed: 12/27/2022] Open
Abstract
Microglia are resident immune cells of the central nervous system. Their development and maintenance depend on stimulation of Colony Stimulating Factor-1 receptor (CSF1R). Microglia play an important role in neurodevelopment and a population of microglia that expresses the complement receptor CD11c is critical for primary myelination. This population is virtually absent in the healthy adult brain but increases dramatically upon neuroinflammatory conditions, and these microglia are suggested to play a protective role in central nervous system (CNS) diseases. To date, the molecular trigger for their expansion is unknown. Here we showed that stimulation of CSF1R by either of its ligands, CSF1 and interleukin (IL)-34, can induce expansion of CD11c+ microglia. In addition, such stimulation resulted in amelioration of EAE symptoms and decreased demyelination. Treatment with CSF1R ligands also induced expression of the chemokine CCL2, and we showed that experimental overexpression of CCL2 in the brain led to a dramatic increase of CD11c+ microglia, independent of CCR2. Moreover, this led to elevated CSF1 expression, suggesting a positive feedback loop between CSF1R and CCL2. These data provide new insights to microglia biology and open new perspectives for modulating microglial activity in neuroinflammatory diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Oriane Cédile
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Kirstine Nolling Jensen
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Isabella Kramer
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nick Boe Elsborg
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| |
Collapse
|
57
|
Visualization of the Breakdown of the Axonal Transport Machinery: a Comparative Ultrastructural and Immunohistochemical Approach. Mol Neurobiol 2018; 56:3984-3998. [PMID: 30238390 DOI: 10.1007/s12035-018-1353-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/13/2018] [Indexed: 12/22/2022]
Abstract
Axonal damage is a major factor contributing to disease progression in multiple sclerosis (MS) patients. On the histological level, acute axonal injury is most frequently analyzed by anti-amyloid precursor protein immunohistochemistry. To what extent this method truly detects axonal injury, and whether other proteins and organelles are as well subjected to axonal transport deficits in demyelinated tissues is not known. The aim of this study was to correlate ultrastructural morphology with the immunohistochemical appearance of acute axonal injury in a model of toxin-induced oligodendrocyte degeneration. C57BL/6J mice were intoxicated with 0.25% cuprizone to induce demyelination. The corpus callosum was investigated by serial block-face scanning electron microscopy (i.e., 3D EM), immunohistochemistry, and immunofluorescence microscopy. Brain tissues of progressive MS patients were included to test the relevance of our findings in mice for MS. Volumes of axonal swellings, determined by 3D EM, were comparable to volumes of axonal spheroids, determined by anti-APP immunofluorescence stains. Axonal swellings were present at myelinated and non-myelinated axonal internodes. Densities of amyloid precursor protein (APP)+ spheroids were highest during active demyelination. Besides APP, vesicular glutamate transporter 1 and mitochondrial proteins accumulated at sites of axonal spheroids. Such accumulations were found as well in lesions of progressive MS patients. In this correlative ultrastructural-immunohistochemical study, we provide strong evidence that breakdown of the axonal transport machinery results in focal accumulations of mitochondria and different synaptic proteins. We provide new marker proteins to visualize acute axonal injury, which helps to further understand the complex nature of axonal damage in progressive MS.
Collapse
|
58
|
Thomas L, Pasquini LA. Galectin-3-Mediated Glial Crosstalk Drives Oligodendrocyte Differentiation and (Re)myelination. Front Cell Neurosci 2018; 12:297. [PMID: 30258354 PMCID: PMC6143789 DOI: 10.3389/fncel.2018.00297] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
Galectin-3 (Gal-3) is the only chimeric protein in the galectin family. Gal-3 structure comprises unusual tandem repeats of proline and glycine-rich short stretches bound to a carbohydrate-recognition domain (CRD). The present review summarizes Gal-3 functions in the extracellular and intracellular space, its regulation and its internalization and secretion, with a focus on the current knowledge of Gal-3 role in central nervous system (CNS) health and disease, particularly oligodendrocyte (OLG) differentiation, myelination and remyelination in experimental models of multiple sclerosis (MS). During myelination, microglia-expressed Gal-3 promotes OLG differentiation by binding glycoconjugates present only on the cell surface of OLG precursor cells (OPC). During remyelination, microglia-expressed Gal-3 favors an M2 microglial phenotype, hence fostering myelin debris phagocytosis through TREM-2b phagocytic receptor and OLG differentiation. Gal-3 is necessary for myelin integrity and function, as evidenced by myelin ultrastructural and behavioral studies from LGALS3-/- mice. Mechanistically, Gal-3 enhances actin assembly and reduces Erk 1/2 activation, leading to early OLG branching. Gal-3 later induces Akt activation and increases MBP expression, promoting gelsolin release and actin disassembly and thus regulating OLG final differentiation. Altogether, findings indicate that Gal-3 mediates the glial crosstalk driving OLG differentiation and (re)myelination and may be regarded as a target in the design of future therapies for a variety of demyelinating diseases.
Collapse
Affiliation(s)
- Laura Thomas
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Laura Andrea Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
59
|
Aust V, Kress E, Abraham S, Schröder N, Kipp M, Stope MB, Pufe T, Tauber SC, Brandenburg LO. Lack of chemokine (C-C motif) ligand 3 leads to decreased survival and reduced immune response after bacterial meningitis. Cytokine 2018; 111:246-254. [PMID: 30199766 DOI: 10.1016/j.cyto.2018.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/31/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022]
Abstract
Pneumococcal meningitis, caused by Streptococcus pneumoniae, is the most common type of bacterial meningitis. The clinical management of this disease has been challenged by the emergence of multidrug-resistant Streptococcus pneumoniae, requiring the urgent development of new therapeutic alternatives. Over the course of bacterial meningitis, pathogen invasion is accompanied by a massive recruitment of peripheral immune cells, especially neutrophil granulocytes, which are recruited under the coordination of several cytokines and chemokines. Here, we used chemokine (C-C motif) ligand 3 (Ccl3)-deficient mice to investigate the functional role of CCL3 in a mouse model of pneumococcal meningitis. Following intrathecal infection with Streptococcus pneumoniae Ccl3-deficient mice presented a significantly shorter survival and higher bacterial load than wildtype mice, paralleled by an ameliorated infiltration of neutrophil granulocytes into the CNS. Blood sample analysis revealed that infected Ccl3-deficient mice showed a significant decrease in erythrocytes, hemoglobin and hematocrit as well as in the number of banded neutrophils. Moreover, infected Ccl3-deficient mice showed an altered cytokine expression profile. Glial cell activation remained unchanged in both genotypes. In summary, this study demonstrates that CCL3 is beneficial in Streptococcus pneumoniae-induced meningitis. Pharmacological modulation of the CCL3 pathways might, therefore, represent a future therapeutic option to manage Streptococcus pneumoniae meningitis.
Collapse
Affiliation(s)
- Vanessa Aust
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Eugenia Kress
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Stephanie Abraham
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Nicole Schröder
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Markus Kipp
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany; Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Germany
| | - Lars-Ove Brandenburg
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
60
|
Trépanier MO, Hildebrand KD, Nyamoya SD, Amor S, Bazinet RP, Kipp M. Phosphatidylcholine 36:1 concentration decreases along with demyelination in the cuprizone animal model and in post-mortem multiple sclerosis brain tissue. J Neurochem 2018; 145:504-515. [DOI: 10.1111/jnc.14335] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/27/2018] [Accepted: 01/30/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Marc-Olivier Trépanier
- Department of Nutritional Sciences; Faculty of Medicine; University of Toronto; Toronto Ontario Canada
| | - Kayla D. Hildebrand
- Department of Nutritional Sciences; Faculty of Medicine; University of Toronto; Toronto Ontario Canada
| | - Stella D. Nyamoya
- Department of Neuroanatomy; Ludwig-Maximilians-University of Munich; Munich Germany
| | - Sandra Amor
- Department of Pathology; VU University Medical Centre; Amsterdam The Netherlands
- Blizard Institute; Barts and The London School of Medicine and Dentistry; Queen Mary University of London; London UK
| | - Richard P. Bazinet
- Department of Nutritional Sciences; Faculty of Medicine; University of Toronto; Toronto Ontario Canada
| | - Markus Kipp
- Department of Neuroanatomy; Ludwig-Maximilians-University of Munich; Munich Germany
| |
Collapse
|
61
|
Noumbissi ME, Galasso B, Stins MF. Brain vascular heterogeneity: implications for disease pathogenesis and design of in vitro blood-brain barrier models. Fluids Barriers CNS 2018; 15:12. [PMID: 29688865 PMCID: PMC5911972 DOI: 10.1186/s12987-018-0097-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/13/2018] [Indexed: 12/22/2022] Open
Abstract
The vertebrate blood–brain barrier (BBB) is composed of cerebral microvascular endothelial cells (CEC). The BBB acts as a semi-permeable cellular interface that tightly regulates bidirectional molecular transport between blood and the brain parenchyma in order to maintain cerebral homeostasis. The CEC phenotype is regulated by a variety of factors, including cells in its immediate environment and within functional neurovascular units. The cellular composition of the brain parenchyma surrounding the CEC varies between different brain regions; this difference is clearly visible in grey versus white matter. In this review, we discuss evidence for the existence of brain vascular heterogeneity, focusing on differences between the vessels of the grey and white matter. The region-specific differences in the vasculature of the brain are reflective of specific functions of those particular brain areas. This BBB-endothelial heterogeneity may have implications for the course of pathogenesis of cerebrovascular diseases and neurological disorders involving vascular activation and dysfunction. This heterogeneity should be taken into account when developing BBB-neuro-disease models representative of specific brain areas.
Collapse
Affiliation(s)
- Midrelle E Noumbissi
- Malaria Research Institute, Dept. Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, SPH East 4135, Baltimore, MD, 21205, USA
| | - Bianca Galasso
- Malaria Research Institute, Dept. Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, SPH East 4135, Baltimore, MD, 21205, USA
| | - Monique F Stins
- Malaria Research Institute, Dept. Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, SPH East 4135, Baltimore, MD, 21205, USA.
| |
Collapse
|
62
|
Yu Y, Fu P, Yu Z, Xie M, Wang W, Luo X. NKCC1 Inhibition Attenuates Chronic Cerebral Hypoperfusion-Induced White Matter Lesions by Enhancing Progenitor Cells of Oligodendrocyte Proliferation. J Mol Neurosci 2018; 64:449-458. [PMID: 29502291 DOI: 10.1007/s12031-018-1043-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/15/2018] [Indexed: 11/28/2022]
Abstract
Cerebral white matter is vulnerable to ischemic condition. However, no effective treatment to alleviate or restore the myelin damage caused by chronic cerebral hypoperfusion has been found. Na+-K+-Cl- cotransporter 1 (NKCC1), a Na+-K+-Cl- cotransporter widely expressed in the central nervous system (CNS), involves in regulation of cell swelling, EAA release, cell apoptosis, and proliferation. Nevertheless, the role of NKCC1 in chronic hypoperfusion-induced white matter lesions (WMLs) has not been explored. Here, mice subjected to bilateral common carotid artery stenosis (BCAS) were used as model of chronic cerebral hypoperfusion; density of progenitor cells of oligodendrocyte (OPCs), oligodendrocytes (OLs), astrocytes, and microglia was assessed by immunofluorescent staining and Western blot analysis; working memory was examined by eight-arm radial maze test; expression of MAPK signaling pathway was determined by Western blot analysis. After BCAS, white matter integrity disruption and working memory impairment were observed. NKCC1 inhibition by bumetanide administration enhanced OPC proliferation, attenuated chronic hypoperfusion-induced white matter damage, and promoted recovery of neurological function. However, NKCC1 inhibition caused no significant change in the densities of GFAP- and Iba-1-positive cells in the corpus callosum. Bumetanide administration significantly increased the expression of p-ERK and decreased the expression of p-JNK and p-p38 in comparison to vehicle-BCAS groups. In conclusion, NKCC1 inhibition might significantly ameliorate chronic cerebral hypoperfusion-induced WMLs and cognitive impairment by enhancing progenitor cells of oligodendrocyte proliferation, and this protective function of bumetanide might be mediated by modulation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Ying Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peicai Fu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
63
|
Low, but not high, dose triptolide controls neuroinflammation and improves behavioral deficits in toxic model of multiple sclerosis by dampening of NF-κB activation and acceleration of intrinsic myelin repair. Toxicol Appl Pharmacol 2018; 342:86-98. [DOI: 10.1016/j.taap.2018.01.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 01/15/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
|
64
|
Bjelobaba I, Begovic-Kupresanin V, Pekovic S, Lavrnja I. Animal models of multiple sclerosis: Focus on experimental autoimmune encephalomyelitis. J Neurosci Res 2018; 96:1021-1042. [PMID: 29446144 DOI: 10.1002/jnr.24224] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Multiple sclerosis (MS) is a chronic, progressive disorder of the central nervous system (CNS) that affects more than two million people worldwide. Several animal models resemble MS pathology; the most employed are experimental autoimmune encephalomyelitis (EAE) and toxin- and/or virus-induced demyelination. In this review we will summarize our knowledge on the utility of different animal models in MS research. Although animal models cannot replicate the complexity and heterogeneity of the MS pathology, they have proved to be useful for the development of several drugs approved for treatment of MS patients. This review focuses on EAE because it represents both clinical and pathological features of MS. During the past decades, EAE has been effective in illuminating various pathological processes that occur during MS, including inflammation, CNS penetration, demyelination, axonopathy, and neuron loss mediated by immune cells.
Collapse
Affiliation(s)
- Ivana Bjelobaba
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | | | - Sanja Pekovic
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic," Department of Neurobiology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
65
|
Lentferink DH, Jongsma JM, Werkman I, Baron W. Grey matter OPCs are less mature and less sensitive to IFNγ than white matter OPCs: consequences for remyelination. Sci Rep 2018; 8:2113. [PMID: 29391408 PMCID: PMC5794790 DOI: 10.1038/s41598-018-19934-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease characterized by the formation of demyelinated lesions in the central nervous system. At later stages of the disease repair in the form of remyelination often fails, which leads to axonal degeneration and neurological disability. For the regeneration of myelin, oligodendrocyte progenitor cells (OPCs) have to migrate, proliferate and differentiate into remyelinating oligodendrocytes. Remyelination occurs faster and is more extensive in grey matter (GM) lesions than in white matter (WM) lesions. Here, we examined differences in neonatal OPCs from GM (gmOPCs) and WM (wmOPCs), both intrinsically and in response to environmental (injury) signals. We show that gmOPCs are less mature than wmOPCs, both on morphological and on gene-expression level. Additionally, gmOPCs proliferate more and differentiate slower than wmOPCs. When exposed to astrocyte-secreted signals wmOPC, but not gmOPC, migration decreases. In addition, wmOPCs are more sensitive to the detrimental effects of IFNγ treatment on proliferation, differentiation, and process arborisation, which is potentiated by TNFα. Our results demonstrate that OPCs from GM and WM differ both intrinsically and in response to their environment, which may contribute to the difference in remyelination efficiency between GM and WM MS lesions.
Collapse
Affiliation(s)
- Dennis H Lentferink
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Jacomien M Jongsma
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Inge Werkman
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Wia Baron
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
66
|
Layé S, Nadjar A, Joffre C, Bazinet RP. Anti-Inflammatory Effects of Omega-3 Fatty Acids in the Brain: Physiological Mechanisms and Relevance to Pharmacology. Pharmacol Rev 2018; 70:12-38. [PMID: 29217656 DOI: 10.1124/pr.117.014092] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 09/05/2017] [Indexed: 12/17/2022] Open
Abstract
Classically, polyunsaturated fatty acids (PUFA) were largely thought to be relatively inert structural components of brain, largely important for the formation of cellular membranes. Over the past 10 years, a host of bioactive lipid mediators that are enzymatically derived from arachidonic acid, the main n-6 PUFA, and docosahexaenoic acid, the main n-3 PUFA in the brain, known to regulate peripheral immune function, have been detected in the brain and shown to regulate microglia activation. Recent advances have focused on how PUFA regulate the molecular signaling of microglia, especially in the context of neuroinflammation and behavior. Several active drugs regulate brain lipid signaling and provide proof of concept for targeting the brain. Because brain lipid metabolism relies on a complex integration of diet, peripheral metabolism, including the liver and blood, which supply the brain with PUFAs that can be altered by genetics, sex, and aging, there are many pathways that can be disrupted, leading to altered brain lipid homeostasis. Brain lipid signaling pathways are altered in neurologic disorders and may be viable targets for the development of novel therapeutics. In this study, we discuss in particular how n-3 PUFAs and their metabolites regulate microglia phenotype and function to exert their anti-inflammatory and proresolving activities in the brain.
Collapse
Affiliation(s)
- Sophie Layé
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Agnès Nadjar
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Corinne Joffre
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Richard P Bazinet
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| |
Collapse
|
67
|
Toll-Like Receptor 2-Mediated Glial Cell Activation in a Mouse Model of Cuprizone-Induced Demyelination. Mol Neurobiol 2017; 55:6237-6249. [PMID: 29288338 DOI: 10.1007/s12035-017-0838-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/12/2017] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS) is a chronic degenerative disease of the central nervous system that is characterized by myelin abnormalities, oligodendrocyte pathology, and concomitant glia activation. The factors triggering gliosis and demyelination are currently not well characterized. New findings suggest an important role of the innate immune response in the initiation and progression of active demyelinating lesions. Especially during progressive disease, aberrant glia activation rather than the invasion of peripheral immune cells is accountable for progressive neuronal injury. The innate immune response can be induced by pathogen-associated or danger-associated molecular patterns, which are identified by pattern recognition receptors (PRRs), including the Toll-like receptors (TLRs). In this study, we used the cuprizone model in mice to investigate the expression of TLR2 during the course of cuprizone-induced demyelination. In addition, we used TLR2-deficient mice to analyze the functional role of TLR2 activation during cuprizone-induced demyelination and reactive gliosis. We show a significantly increased expression of TLR2 in the corpus callosum and hippocampus of cuprizone-intoxicated mice. The absence of receptor signaling in TLR2-deficient mice resulted in less severe reactive astrogliosis in the corpus callosum and cortex. In addition, microglia activation was ameliorated in the corpus callosum of TLR2-deficient mice, but augmented in the cortex compared to wild-type littermates. Extent of demyelination and loss of mature oligodendrocytes was comparable in both genotypes. These results suggest that the TLR2 orchestrates glia activation during gray and white matter demyelination in the presence of an intact blood-brain barrier. Future studies now have to address the underlying mechanisms of the region-specific TLR2-mediated glia activation.
Collapse
|
68
|
Sanadgol N, Golab F, Tashakkor Z, Taki N, Moradi Kouchi S, Mostafaie A, Mehdizadeh M, Abdollahi M, Taghizadeh G, Sharifzadeh M. Neuroprotective effects of ellagic acid on cuprizone-induced acute demyelination through limitation of microgliosis, adjustment of CXCL12/IL-17/IL-11 axis and restriction of mature oligodendrocytes apoptosis. PHARMACEUTICAL BIOLOGY 2017; 55:1679-1687. [PMID: 28447514 PMCID: PMC6130560 DOI: 10.1080/13880209.2017.1319867] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 10/29/2016] [Accepted: 04/12/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT Ellagic acid (EA) is a natural phenol antioxidant with various therapeutic activities. However, the efficacy of EA has not been examined in neuropathologic conditions. OBJECTIVE In vivo neuroprotective effects of EA on cuprizone (cup)-induced demyelination were evaluated. MATERIAL AND METHODS C57BL/6 J mice were fed with chow containing 0.2% cup for 4 weeks to induce oligodendrocytes (OLGs) depletion predominantly in the corpus callosum (CC). EA was administered at different doses (40 or 80 mg/kg body weight/day/i.p.) from the first day of cup diet. Oligodendrocytes apoptosis [TUNEL assay and myelin oligodendrocyte glycoprotein (MOG+)/caspase-3+ cells), gliosis (H&E staining, glial fibrillary acidic protein (GFAP+) and macrophage-3 (Mac-3+) cells) and inflammatory markers (interleukin 17 (IL-17), interleukin 11 (IL-11) and stromal cell-derived factor 1 α (SDF-1α) or CXCL12] during cup intoxication were examined. RESULTS High dose of EA (EA-80) increased mature oligodendrocytes population (MOG+ cells, p < 0.001), and decreased apoptosis (p < 0.05) compared with the cup mice. Treatment with both EA doses did not show any considerable effects on the expression of CXCL12, but significantly down-regulated the expression of IL-17 and up-regulated the expression of IL-11 in mRNA levels compared with the cup mice. Only treatment with EA-80 significantly decreased the population of active macrophage (MAC-3+ cells, p < 0.001) but not reactive astrocytes (GFAP+ cells) compared with the cup mice. DISCUSSION AND CONCLUSION In this model, EA-80 effectively reduces lesions via reduction of neuroinflammation and toxic effects of cup on mature OLGs. EA is a suitable therapeutic agent for moderate brain damage in neurodegenerative diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Nima Sanadgol
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Science, Tehran, Iran
| | - Zakiyeh Tashakkor
- MSc in Cell and Developmental Biology, Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Nooshin Taki
- MSc in Cell and Developmental Biology, Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Samira Moradi Kouchi
- MSc in Cell and Developmental Biology, Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ali Mostafaie
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Mehdizadeh
- Cellular and Molecular Research Center, Iran University of Medical Science, Tehran, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghorban Taghizadeh
- Department of Occupational Therapy, Faculty of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
69
|
Berghoff SA, Düking T, Spieth L, Winchenbach J, Stumpf SK, Gerndt N, Kusch K, Ruhwedel T, Möbius W, Saher G. Blood-brain barrier hyperpermeability precedes demyelination in the cuprizone model. Acta Neuropathol Commun 2017; 5:94. [PMID: 29195512 PMCID: PMC5710130 DOI: 10.1186/s40478-017-0497-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 11/10/2022] Open
Abstract
In neuroinflammatory disorders such as multiple sclerosis, the physiological function of the blood-brain barrier (BBB) is perturbed, particularly in demyelinating lesions and supposedly secondary to acute demyelinating pathology. Using the toxic non-inflammatory cuprizone model of demyelination, we demonstrate, however, that the onset of persistent BBB impairment precedes demyelination. In addition to a direct effect of cuprizone on endothelial cells, a plethora of inflammatory mediators, which are mainly of astroglial origin during the initial disease phase, likely contribute to the destabilization of endothelial barrier function in vivo. Our study reveals that, at different time points of pathology and in different CNS regions, the level of gliosis correlates with the extent of BBB hyperpermeability and edema. Furthermore, in mutant mice with abolished type 3 CXC chemokine receptor (CXCR3) signaling, inflammatory responses are dampened and BBB dysfunction ameliorated. Together, these data have implications for understanding the role of BBB permeability in the pathogenesis of demyelinating disease.
Collapse
|
70
|
Baxi EG, DeBruin J, Jin J, Strasburger HJ, Smith MD, Orthmann-Murphy JL, Schott JT, Fairchild AN, Bergles DE, Calabresi PA. Lineage tracing reveals dynamic changes in oligodendrocyte precursor cells following cuprizone-induced demyelination. Glia 2017; 65:2087-2098. [PMID: 28940645 DOI: 10.1002/glia.23229] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 08/20/2017] [Accepted: 08/23/2017] [Indexed: 11/10/2022]
Abstract
The regeneration of oligodendrocytes is a crucial step in recovery from demyelination, as surviving oligodendrocytes exhibit limited structural plasticity and rarely form additional myelin sheaths. New oligodendrocytes arise through the differentiation of platelet-derived growth factor receptor α (PDGFRα) expressing oligodendrocyte progenitor cells (OPCs) that are widely distributed throughout the CNS. Although there has been detailed investigation of the behavior of these progenitors in white matter, recent studies suggest that disease burden in multiple sclerosis (MS) is more strongly correlated with gray matter atrophy. The timing and efficiency of remyelination in gray matter is distinct from white matter, but the dynamics of OPCs that contribute to these differences have not been defined. Here, we used in vivo genetic fate tracing to determine the behavior of OPCs in gray and white matter regions in response to cuprizone-induced demyelination. Our studies indicate that the temporal dynamics of OPC differentiation varies significantly between white and gray matter. While OPCs rapidly repopulate the corpus callosum and mature into CC1 expressing mature oligodendrocytes, OPC differentiation in the cingulate cortex and hippocampus occurs much more slowly, resulting in a delay in remyelination relative to the corpus callosum. The protracted maturation of OPCs in gray matter may contribute to greater axonal pathology and disease burden in MS.
Collapse
Affiliation(s)
- Emily G Baxi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph DeBruin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jing Jin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hayley J Strasburger
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer L Orthmann-Murphy
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University Medical School, Baltimore, Maryland
| | - Jason T Schott
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amanda N Fairchild
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University Medical School, Baltimore, Maryland
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University Medical School, Baltimore, Maryland
| |
Collapse
|
71
|
CRAMP deficiency leads to a pro-inflammatory phenotype and impaired phagocytosis after exposure to bacterial meningitis pathogens. Cell Commun Signal 2017; 15:32. [PMID: 28915816 PMCID: PMC5602852 DOI: 10.1186/s12964-017-0190-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/12/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Antimicrobial peptides are important components of the host defence with a broad range of functions including direct antimicrobial activity and modulation of inflammation. Lack of cathelin-related antimicrobial peptide (CRAMP) was associated with higher mortality and bacterial burden and impaired neutrophil granulocyte infiltration in a model of pneumococcal meningitis. The present study was designed to characterize the effects of CRAMP deficiency on glial response and phagocytosis after exposure to bacterial stimuli. METHODS CRAMP-knock out and wildtype glial cells were exposed to bacterial supernatants from Streptococcus pneumoniae and Neisseria meningitides or the bacterial cell wall components lipopolysaccharide and peptidoglycan. Cell viability, expression of pro- and anti-inflammatory mediators and activation of signal transduction pathways, phagocytosis rate and glial cell phenotype were investigated by means of cell viability assays, immunohistochemistry, real-time RT-PCR and Western blot. RESULTS CRAMP-deficiency was associated with stronger expression of pro-inflammatory and weakened expression of anti-inflammatory cytokines indicating a higher degree of glial cell activation even under resting-state conditions. Furthermore, increased translocation of nuclear factor 'kappa-light-chain-enhancer' of activated B-cells was observed and phagocytosis of S. pneumoniae was reduced in CRAMP-deficient microglia indicating impaired antimicrobial activity. CONCLUSIONS In conclusion, the present study detected severe alterations of the glial immune response due to lack of CRAMP. The results indicate the importance of CRAMP to maintain and regulate the delicate balance between beneficial and harmful immune response in the brain.
Collapse
|
72
|
Bihler K, Kress E, Esser S, Nyamoya S, Tauber SC, Clarner T, Stope MB, Pufe T, Brandenburg LO. Formyl Peptide Receptor 1-Mediated Glial Cell Activation in a Mouse Model of Cuprizone-Induced Demyelination. J Mol Neurosci 2017; 62:232-243. [PMID: 28466255 DOI: 10.1007/s12031-017-0924-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/17/2017] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is a chronic degenerative disease of the central nervous system that is characterized by myelin abnormalities, oligodendrocyte pathology, and concomitant glia activation. Unclear are the factors triggering gliosis and demyelination. New findings suggest an important role of the innate immune response in the initiation and progression of active demyelinating lesions. The innate immune response is induced by pathogen-associated or danger-associated molecular patterns, which are identified by pattern recognition receptors (PRRs), including the G-protein coupled with formyl peptide receptors (FPRs). Glial cells, the immune cells of the central nervous system, also express the PRRs. In this study, we used the cuprizone mice model to investigate the expression of the FPR1 in the course of cuprizone-induced demyelination In addition, we used FPR1-deficient mice to analyze glial cell activation through immunohistochemistry and real-time RT-PCR in cuprizone model. Our results revealed a significantly increased expression of FPR1 in the cortex of cuprizone-treated mice. FPR1-deficient mice showed a slight but significant decrease of demyelination in the corpus callosum compared to the wild-type mice. Furthermore, FPR1 deficiency resulted in reduced glial cell activation and mRNA expression of microglia/macrophages markers, as well as pro- and anti-inflammatory cytokines in the cortex, compared to wild-type mice after cuprizone-induced demyelination. Combined together, these results suggest that the FPR1 is an important part of the innate immune response in the course of cuprizone-induced demyelination.
Collapse
Affiliation(s)
- Kai Bihler
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Eugenia Kress
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Stefan Esser
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Stella Nyamoya
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Tim Clarner
- Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Lars-Ove Brandenburg
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
73
|
Zhang Y, Yin L, Zheng N, Zhang L, Liu J, Liang W, Wang Q. Icariin enhances remyelination process after acute demyelination induced by cuprizone exposure. Brain Res Bull 2017; 130:180-187. [DOI: 10.1016/j.brainresbull.2017.01.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 01/30/2017] [Indexed: 10/25/2022]
|
74
|
Taraboletti A, Walker T, Avila R, Huang H, Caporoso J, Manandhar E, Leeper TC, Modarelli DA, Medicetty S, Shriver LP. Cuprizone Intoxication Induces Cell Intrinsic Alterations in Oligodendrocyte Metabolism Independent of Copper Chelation. Biochemistry 2017; 56:1518-1528. [PMID: 28186720 PMCID: PMC6145805 DOI: 10.1021/acs.biochem.6b01072] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cuprizone intoxication is a common animal model used to test myelin regenerative therapies for the treatment of diseases such as multiple sclerosis. Mice fed this copper chelator develop reversible, region-specific oligodendrocyte loss and demyelination. While the cellular changes influencing the demyelinating process have been explored in this model, there is no consensus about the biochemical mechanisms of toxicity in oligodendrocytes and about whether this damage arises from the chelation of copper in vivo. Here we have identified an oligodendroglial cell line that displays sensitivity to cuprizone toxicity and performed global metabolomic profiling to determine biochemical pathways altered by this treatment. We link these changes with alterations in brain metabolism in mice fed cuprizone for 2 and 6 weeks. We find that cuprizone induces widespread changes in one-carbon and amino acid metabolism as well as alterations in small molecules that are important for energy generation. We used mass spectrometry to examine chemical interactions that are important for copper chelation and toxicity. Our results indicate that cuprizone induces global perturbations in cellular metabolism that may be independent of its copper chelating ability and potentially related to its interactions with pyridoxal 5'-phosphate, a coenzyme essential for amino acid metabolism.
Collapse
Affiliation(s)
| | - Tia Walker
- Department of Chemistry, Indiana University Northwest, Gary, Indiana 46408, United States
| | - Robin Avila
- Renovo Neural, Inc., Cleveland, Ohio 44106, United States
| | - He Huang
- Department of Chemistry, University of Akron, Akron, Ohio 44325, United States
| | - Joel Caporoso
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260, United States
| | - Erendra Manandhar
- Department of Chemistry, University of Akron, Akron, Ohio 44325, United States
| | - Thomas C. Leeper
- Department of Chemistry, College of Wooster, Wooster, Ohio 44691, United States
| | - David A. Modarelli
- Department of Chemistry, University of Akron, Akron, Ohio 44325, United States
| | | | - Leah P. Shriver
- Department of Chemistry, University of Akron, Akron, Ohio 44325, United States
- Department of Biology, University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
75
|
Heckers S, Held N, Kronenberg J, Skripuletz T, Bleich A, Gudi V, Stangel M. Investigation of Cuprizone Inactivation by Temperature. Neurotox Res 2017; 31:570-577. [PMID: 28124768 DOI: 10.1007/s12640-017-9704-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 12/25/2022]
Abstract
Animal models, such as cuprizone (bis-cyclohexanone oxaldihydrazone) feeding, are helpful to study experimental demyelination and remyelination in the context of diseases like multiple sclerosis. Cuprizone is a copper chelator, which when supplemented to the normal food of C57BL/6J mice in a concentration of 0.2% leads to oligodendroglial loss, subsequent microglia and astrocyte activation, resulting in demyelination. Termination of the cuprizone diet results in remyelination, promoted by newly formed mature oligodendrocytes. The exact mode of cuprizone's action is not well understood, and information about its inactivation and cleavage are still not available. The knowledge of these processes could lead to a better understanding of cuprizone's mode of action, as well as a safer handling of this toxin. We therefore performed experiments with the aim to inactivate cuprizone by thermal heating, since it was suggested in the past that cuprizone is heat sensitive. C57BL/6J mice were fed for 4 weeks with 0.2% cuprizone, either thermally pretreated (60, 80, 105, 121 °C) or not heated. In addition, primary rat oligodendrocytes, as a known selective toxic target of cuprizone, were incubated with 350 μM cuprizone solutions, which were either thermally pretreated or not. Our results demonstrate that none of the tested thermal pretreatment conditions could abrogate or restrict the toxic and demyelinating effects of cuprizone, neither in vitro nor in vivo. In conclusion, the current study rebuts the hypothesis of cuprizone as a heat-sensitive compound, as well as the assumption that heat exposure is a reason for an insufficient demyelination of cuprizone-containing pellets.
Collapse
Affiliation(s)
- Sandra Heckers
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Nadine Held
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Jessica Kronenberg
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Thomas Skripuletz
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany. .,Center for Systems Neuroscience, Hannover, Germany. .,Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
76
|
Kipp M, Nyamoya S, Hochstrasser T, Amor S. Multiple sclerosis animal models: a clinical and histopathological perspective. Brain Pathol 2017; 27:123-137. [PMID: 27792289 DOI: 10.1111/bpa.12454] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022] Open
Abstract
There is a broad consensus that multiple sclerosis (MS) represents more than an inflammatory disease: it harbors several characteristic aspects of a classical neurodegenerative disorder, that is, damage to axons, synapses and nerve cell bodies. While we are equipped with appropriate therapeutic options to prevent immune-cell driven relapses, effective therapeutic options to prevent the progressing neurodegeneration are still missing. In this review article, we will discuss to what extent pathology of the progressive disease stage can be modeled in MS animal models. While acute and relapsing-remitting forms of experimental autoimmune encephalomyelitis (EAE), which are T cell dependent, are aptly suited to model relapsing-remitting phases of MS, other EAE models, especially the secondary progressive EAE stage in Biozzi ABH mice is better representing the secondary progressive phase of MS, which is refractory to many immune therapies. Besides EAE, the cuprizone model is rapidly gaining popularity to study the formation and progression of demyelinating CNS lesions without T cell involvement. Here, we discuss these two non-popular MS models. It is our aim to point out the pathological hallmarks of MS, and discuss which pathological aspects of the disease can be best studied in the various animal models available.
Collapse
Affiliation(s)
- Markus Kipp
- Department of Neuroanatomy, Faculty of Medicine, LMU München University, München, 80336, Germany
| | - Stella Nyamoya
- Department of Neuroanatomy, Faculty of Medicine, LMU München University, München, 80336, Germany.,Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, D-52074, Germany
| | - Tanja Hochstrasser
- Department of Neuroanatomy, Faculty of Medicine, LMU München University, München, 80336, Germany
| | - Sandra Amor
- Department of Pathology, VU University Medical Centre, Amsterdam, The Netherlands.,Barts and The London School of Medicine and Dentistry, Neuroimmunology Unit, , Queen Mary University of London, Neuroscience Centre, Blizard Institute of Cell and Molecular Science, London, UK
| |
Collapse
|
77
|
Böhland M, Kress E, Stope MB, Pufe T, Tauber SC, Brandenburg LO. Lack of Toll-like receptor 2 results in higher mortality of bacterial meningitis by impaired host resistance. J Neuroimmunol 2016; 299:90-97. [PMID: 27725130 DOI: 10.1016/j.jneuroim.2016.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 12/17/2022]
Abstract
Bacterial meningitis is - despite therapeutical progress during the last decades - still characterized by high mortality and severe permanent neurogical sequelae. The brain is protected from penetrating pathogens by both the blood-brain barrier and the innate immune system. Invading pathogens are recognized by so-called pattern recognition receptors including the Toll-like receptors (TLR) which are expressed by glial immune cells in the central nervous system. Among these, TLR2 is responsible for the detection of Gram-positive bacteria such as the meningitis-causing pathogen Streptococcus pneumoniae. Here, we used TLR2-deficient mice to investigate the effects on mortality, bacterial growth and inflammation in a mouse model of pneumococcal meningitis. Our results revealed a significantly increased mortality rate and higher bacterial burden in TLR2-deficient mice with pneumococcal meningitis. Furthermore, infected TLR2-deficient mice suffered from a significantly increased pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) and Chemokine (C-C motif) ligand 2 (CCL2) or CCL3 chemokine expression and decreased expression of anti-inflammatory cytokines and antimicrobial peptides. In contrast, glial cell activation assessed by glial cell marker expression was comparable to wildtype mice. Taken together, the results suggest that TLR2 is essential for an efficient immune response against Streptococcus pneumoniae meningitis since lack of the receptor led to a worse outcome by higher mortality due to increased bacterial burden, weakened innate immune response and reduced expression of antimicrobial peptides.
Collapse
Affiliation(s)
- Martin Böhland
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Eugenia Kress
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Germany
| | | |
Collapse
|
78
|
Kipp M, Hochstrasser T, Schmitz C, Beyer C. Female sex steroids and glia cells: Impact on multiple sclerosis lesion formation and fine tuning of the local neurodegenerative cellular network. Neurosci Biobehav Rev 2016; 67:125-36. [DOI: 10.1016/j.neubiorev.2015.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/30/2015] [Accepted: 11/04/2015] [Indexed: 01/01/2023]
|
79
|
Ofengeim D, Ito Y, Najafov A, Zhang Y, Shan B, DeWitt JP, Ye J, Zhang X, Chang A, Vakifahmetoglu-Norberg H, Geng J, Py B, Zhou W, Amin P, Berlink Lima J, Qi C, Yu Q, Trapp B, Yuan J. Activation of necroptosis in multiple sclerosis. Cell Rep 2015; 10:1836-49. [PMID: 25801023 DOI: 10.1016/j.celrep.2015.02.051] [Citation(s) in RCA: 418] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 01/14/2015] [Accepted: 02/21/2015] [Indexed: 11/20/2022] Open
Abstract
Multiple sclerosis (MS), a common neurodegenerative disease of the CNS, is characterized by the loss of oligodendrocytes and demyelination. Tumor necrosis factor α (TNF-α), a proinflammatory cytokine implicated in MS, can activate necroptosis, a necrotic cell death pathway regulated by RIPK1 and RIPK3 under caspase-8-deficient conditions. Here, we demonstrate defective caspase-8 activation, as well as activation of RIPK1, RIPK3, and MLKL, the hallmark mediators of necroptosis, in the cortical lesions of human MS pathological samples. Furthermore, we show that MS pathological samples are characterized by an increased insoluble proteome in common with other neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson’s disease (PD), and Huntington's disease (HD). Finally, we show that necroptosis mediates oligodendrocyte degeneration induced by TNF-α and that inhibition of RIPK1 protects against oligodendrocyte cell death in two animal models of MS and in culture. Our findings demonstrate that necroptosis is involved in MS and suggest that targeting RIPK1 may represent a therapeutic strategy for MS.
Collapse
|
80
|
Guglielmetti C, Veraart J, Roelant E, Mai Z, Daans J, Van Audekerke J, Naeyaert M, Vanhoutte G, Delgado Y Palacios R, Praet J, Fieremans E, Ponsaerts P, Sijbers J, Van der Linden A, Verhoye M. Diffusion kurtosis imaging probes cortical alterations and white matter pathology following cuprizone induced demyelination and spontaneous remyelination. Neuroimage 2015; 125:363-377. [PMID: 26525654 DOI: 10.1016/j.neuroimage.2015.10.052] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/15/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022] Open
Abstract
Although MRI is the gold standard for the diagnosis and monitoring of multiple sclerosis (MS), current conventional MRI techniques often fail to detect cortical alterations and provide little information about gliosis, axonal damage and myelin status of lesioned areas. Diffusion tensor imaging (DTI) and diffusion kurtosis imaging (DKI) provide sensitive and complementary measures of the neural tissue microstructure. Additionally, specific white matter tract integrity (WMTI) metrics modelling the diffusion in white matter were recently derived. In the current study we used the well-characterized cuprizone mouse model of central nervous system demyelination to assess the temporal evolution of diffusion tensor (DT), diffusion kurtosis tensor (DK) and WMTI-derived metrics following acute inflammatory demyelination and spontaneous remyelination. While DT-derived metrics were unable to detect cuprizone induced cortical alterations, the mean kurtosis (MK) and radial kurtosis (RK) were found decreased under cuprizone administration, as compared to age-matched controls, in both the motor and somatosensory cortices. The MK remained decreased in the motor cortices at the end of the recovery period, reflecting long lasting impairment of myelination. In white matter, DT, DK and WMTI-derived metrics enabled the detection of cuprizone induced changes differentially according to the stage and the severity of the lesion. More specifically, the MK, the RK and the axonal water fraction (AWF) were the most sensitive for the detection of cuprizone induced changes in the genu of the corpus callosum, a region less affected by cuprizone administration. Additionally, microgliosis was associated with an increase of MK and RK during the acute inflammatory demyelination phase. In regions undergoing severe demyelination, namely the body and splenium of the corpus callosum, DT-derived metrics, notably the mean diffusion (MD) and radial diffusion (RD), were among the best discriminators between cuprizone and control groups, hence highlighting their ability to detect both acute and long lasting changes. Interestingly, WMTI-derived metrics showed the aptitude to distinguish between the different stages of the disease. Both the intra-axonal diffusivity (Da) and the AWF were found to be decreased in the cuprizone treated group, Da specifically decreased during the acute inflammatory demyelinating phase whereas the AWF decrease was associated to the spontaneous remyelination and the recovery period. Altogether our results demonstrate that DKI is sensitive to alterations of cortical areas and provides, along with WMTI metrics, information that is complementary to DT-derived metrics for the characterization of demyelination in both white and grey matter and subsequent inflammatory processes associated with a demyelinating event.
Collapse
Affiliation(s)
- C Guglielmetti
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
| | - J Veraart
- iMinds - Vision Lab, Department of Physics, University of Antwerp, Antwerp, Belgium; Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - E Roelant
- StatUa Center for Statistics, University of Antwerp, Antwerp, Belgium
| | - Z Mai
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
| | - J Daans
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | - M Naeyaert
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
| | - G Vanhoutte
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
| | | | - J Praet
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
| | - E Fieremans
- Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - P Ponsaerts
- Experimental Cell Transplantation Group, Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - J Sijbers
- iMinds - Vision Lab, Department of Physics, University of Antwerp, Antwerp, Belgium
| | | | - M Verhoye
- Bio-Imaging Lab, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
81
|
Große-Veldmann R, Becker B, Amor S, van der Valk P, Beyer C, Kipp M. Lesion Expansion in Experimental Demyelination Animal Models and Multiple Sclerosis Lesions. Mol Neurobiol 2015; 53:4905-17. [DOI: 10.1007/s12035-015-9420-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/02/2015] [Indexed: 01/03/2023]
|
82
|
Stuart MJ, Singhal G, Baune BT. Systematic Review of the Neurobiological Relevance of Chemokines to Psychiatric Disorders. Front Cell Neurosci 2015; 9:357. [PMID: 26441528 PMCID: PMC4564736 DOI: 10.3389/fncel.2015.00357] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/26/2015] [Indexed: 12/13/2022] Open
Abstract
Psychiatric disorders are highly prevalent and disabling conditions of increasing public health relevance. Much recent research has focused on the role of cytokines in the pathophysiology of psychiatric disorders; however, the related family of immune proteins designated chemokines has been relatively neglected. Chemokines were originally identified as having chemotactic function on immune cells; however, recent evidence has begun to elucidate novel, brain-specific functions of these proteins of relevance to the mechanisms of psychiatric disorders. A systematic review of both human and animal literature in the PubMed and Google Scholar databases was undertaken. After application of all inclusion and exclusion criteria, 157 references were remained for the review. Some early mechanistic evidence does associate select chemokines with the neurobiological processes, including neurogenesis, modulation of the neuroinflammatory response, regulation of the hypothalamus–pituitary–adrenal axis, and modulation of neurotransmitter systems. This early evidence however does not clearly demonstrate any specificity for a certain psychiatric disorder, but is primarily relevant to mechanisms which are shared across disorders. Notable exceptions include CCL11 that has recently been shown to impair hippocampal function in aging – of distinct relevance to Alzheimer’s disease and depression in the elderly, and pre-natal exposure to CXCL8 that may disrupt early neurodevelopmental periods predisposing to schizophrenia. Pro-inflammatory chemokines, such as CCL2, CCL7, CCL8, CCL12, and CCL13, have been shown to drive chemotaxis of pro-inflammatory cells to the inflamed or injured CNS. Likewise, CX3CL has been implicated in promoting glial cells activation, pro-inflammatory cytokines secretion, expression of ICAM-1, and recruitment of CD4+ T-cells into the CNS during neuroinflammatory processes. With further translational research, chemokines may present novel diagnostic and/or therapeutic targets in psychiatric disorders.
Collapse
Affiliation(s)
- Michael J Stuart
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, University of Adelaide , Adelaide, SA , Australia ; School of Medicine, University of Queensland , Brisbane, QLD , Australia
| | - Gaurav Singhal
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, University of Adelaide , Adelaide, SA , Australia
| | - Bernhard T Baune
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, University of Adelaide , Adelaide, SA , Australia
| |
Collapse
|
83
|
Prins M, Schul E, Geurts J, van der Valk P, Drukarch B, van Dam AM. Pathological differences between white and grey matter multiple sclerosis lesions. Ann N Y Acad Sci 2015. [PMID: 26200258 DOI: 10.1111/nyas.12841] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a debilitating disease characterized by demyelination of the central nervous system (CNS), resulting in widespread formation of white matter lesions (WMLs) and grey matter lesions (GMLs). WMLs are pathologically characterized by the presence of immune cells that infiltrate the CNS, whereas these immune cells are barely present in GMLs. This striking pathological difference between WMLs and GMLs raises questions about the underlying mechanism. It is known that infiltrating leukocytes contribute to the generation of WMLs; however, since GMLs show a paucity of infiltrating immune cells, their importance in GML formation remains to be determined. Here, we review pathological characteristics of WMLs and GMLs, and suggest some possible explanations for the observed pathological differences. In our view, cellular and molecular characteristics of WM and GM, and local differences within WMLs and GMLs (in particular, in glial cell populations and the molecules they express), determine the pathway to demyelination. Further understanding of GML pathogenesis, considered to contribute to chronic MS, may have a direct impact on the development of novel therapeutic targets to counteract this progressive neurological disorder.
Collapse
Affiliation(s)
| | | | | | - Paul van der Valk
- Department of Pathology, VU University Medical Center, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
84
|
Cheng T, Yang B, Li D, Ma S, Tian Y, Qu R, Zhang W, Zhang Y, Hu K, Guan F, Wang J. Wharton's Jelly Transplantation Improves Neurologic Function in a Rat Model of Traumatic Brain Injury. Cell Mol Neurobiol 2015; 35:641-9. [PMID: 25638565 PMCID: PMC4481175 DOI: 10.1007/s10571-015-0159-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/22/2015] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI), which can lead to disability, dysfunction, and even death, is a prominent health problem worldwide. Effective therapy for this serious and debilitating condition is needed. Human umbilical cord matrix, known as Wharton's jelly (WJ), provides a natural, interface scaffold that is enriched in mesenchymal stem cells. In this study, we tested the efficacy of WJ tissue transplantation in a weight-drop model of TBI in rats. WJ tissue was cultured and transplanted into the injury site 24 h after TBI. The modified neurologic severity score, body weight, brain edema, and lesion volume were evaluated at various time points after TBI. Cognitive behavior was assessed by the novel object recognition test and the Morris water maze test. Expression of brain-derived neurotrophic factor (BDNF) in the perilesional brain area was measured at day 14 after TBI. We found that WJ tissue transplantation lessened TBI-induced brain edema (day 3), reduced lesion volume (day 28), improved neurologic function (days 21-28), and promoted memory and cognitive recovery. Additionally, expression of BDNF mRNA and protein was higher in WJ tissue-treated rats than in sham-operated or vehicle-treated rats. These data suggest that WJ tissue transplantation can reduce TBI-induced brain injury and may have therapeutic potential for the treatment of TBI.
Collapse
Affiliation(s)
- Tian Cheng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan People’s Republic of China
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD 21205 USA
| | - Bo Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan People’s Republic of China
| | - Dongpeng Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan People’s Republic of China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Yi Tian
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan People’s Republic of China
| | - Ruina Qu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Wenjin Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan People’s Republic of China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Kai Hu
- Henan Academy of Medical Sciences, Zhengzhou, 450003 Henan People’s Republic of China
| | - Fangxia Guan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan People’s Republic of China
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan People’s Republic of China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD 21205 USA
| |
Collapse
|
85
|
Anatomical Distribution of Cuprizone-Induced Lesions in C57BL6 Mice. J Mol Neurosci 2015; 57:166-75. [DOI: 10.1007/s12031-015-0595-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/02/2015] [Indexed: 12/19/2022]
|
86
|
Dörr A, Kress E, Podschun R, Pufe T, Tauber SC, Brandenburg LO. Intrathecal application of the antimicrobial peptide CRAMP reduced mortality and neuroinflammation in an experimental model of pneumococcal meningitis. J Infect 2015; 71:188-99. [PMID: 25896094 DOI: 10.1016/j.jinf.2015.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/23/2015] [Accepted: 04/10/2015] [Indexed: 12/23/2022]
Abstract
Antimicrobial peptides (AP) are important components of the innate immune system. Our previous work revealed a higher mortality rate and up-regulation of proinflammatory gene expression as well as glial cell activation in cathelicidin-related antimicrobial peptide (CRAMP)-deficient mice after bacterial meningitis. However, the influence of CRAMP application on the progression of inflammation and its impact on mortality after bacterial meningitis remains unknown. To assess the effects of continuous CRAMP exposure in the brain, C57BL/6 wildtype mice were given intracerebroventricular infusion of CRAMP to investigate the effects on mortality, glial cell activation and inflammation in a mouse model of pneumococcal meningitis using immunohistochemistry and realtime RT-PCR. Our results revealed a decrease of mortality after CRAMP infusion. The intrathecal CRAMP infusion after pneumococcal meningitis resulted in a decreased mRNA expression of pro-inflammatory cytokines, whereas the immune responses including the expression of pattern recognition receptors and chemokines were increased in bacterial meningitis. Taken together, the results support the important role of CRAMP as part of the innate immune response against pathogens in bacterial CNS infections. The APs may be a promising approach for the development of an adjuvant therapy for bacterial meningitis.
Collapse
Affiliation(s)
- Arndt Dörr
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Eugenia Kress
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Rainer Podschun
- Institute for Infection Medicine, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Germany
| | | |
Collapse
|
87
|
Doorn KJ, Brevé JJP, Drukarch B, Boddeke HW, Huitinga I, Lucassen PJ, van Dam AM. Brain region-specific gene expression profiles in freshly isolated rat microglia. Front Cell Neurosci 2015; 9:84. [PMID: 25814934 PMCID: PMC4357261 DOI: 10.3389/fncel.2015.00084] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/23/2015] [Indexed: 12/30/2022] Open
Abstract
Microglia are important cells in the brain that can acquire different morphological and functional phenotypes dependent on the local situation they encounter. Knowledge on the region-specific gene signature of microglia may hold valuable clues for microglial functioning in health and disease, e.g., Parkinson's disease (PD) in which microglial phenotypes differ between affected brain regions. Therefore, we here investigated whether regional differences exist in gene expression profiles of microglia that are isolated from healthy rat brain regions relevant for PD. We used an optimized isolation protocol based on a rapid isolation of microglia from discrete rat gray matter regions using density gradients and fluorescent-activated cell sorting. Application of the present protocol followed by gene expression analysis enabled us to identify subtle differences in region-specific microglial expression profiles and show that the genetic profile of microglia already differs between different brain regions when studied under control conditions. As such, these novel findings imply that brain region-specific microglial gene expression profiles exist that may contribute to the region-specific differences in microglia responsivity during disease conditions, such as seen in, e.g., PD.
Collapse
Affiliation(s)
- Karlijn J Doorn
- Department Structural and Functional Plasticity of the Nervous System, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands ; Neuroscience Campus Amsterdam, Department Anatomy and Neurosciences, VU University Medical Center Amsterdam, Netherlands
| | - John J P Brevé
- Neuroscience Campus Amsterdam, Department Anatomy and Neurosciences, VU University Medical Center Amsterdam, Netherlands
| | - Benjamin Drukarch
- Neuroscience Campus Amsterdam, Department Anatomy and Neurosciences, VU University Medical Center Amsterdam, Netherlands
| | - Hendrikus W Boddeke
- Section Medical Physiology, Department of Neuroscience, University Medical Centre Groningen Groningen, Netherlands
| | - Inge Huitinga
- Neuroimmunology Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences Amsterdam, Netherlands
| | - Paul J Lucassen
- Department Structural and Functional Plasticity of the Nervous System, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Anne-Marie van Dam
- Department Structural and Functional Plasticity of the Nervous System, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
88
|
Clarner T, Janssen K, Nellessen L, Stangel M, Skripuletz T, Krauspe B, Hess FM, Denecke B, Beutner C, Linnartz-Gerlach B, Neumann H, Vallières L, Amor S, Ohl K, Tenbrock K, Beyer C, Kipp M. CXCL10 Triggers Early Microglial Activation in the Cuprizone Model. THE JOURNAL OF IMMUNOLOGY 2015; 194:3400-13. [DOI: 10.4049/jimmunol.1401459] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
89
|
Janssen K, Rickert M, Clarner T, Beyer C, Kipp M. Absence of CCL2 and CCL3 Ameliorates Central Nervous System Grey Matter But Not White Matter Demyelination in the Presence of an Intact Blood-Brain Barrier. Mol Neurobiol 2015; 53:1551-1564. [PMID: 25663168 DOI: 10.1007/s12035-015-9113-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/22/2015] [Indexed: 11/25/2022]
Abstract
A broad spectrum of diseases is characterized by myelin abnormalities, oligodendrocyte pathology, and concomitant glia activation, among multiple sclerosis (MS). Our knowledge regarding the factors triggering gliosis and demyelination is scanty. Chemokines are pivotal for microglia and astrocyte activation and orchestrate critical steps during the formation of central nervous system (CNS) demyelinating lesions. Redundant functions of chemokines complicate, however, the study of their functional relevance. We used the cuprizone model to study redundant functions of two chemokines, CCL2/MCP1 and CCL3/MIP1α, which are critically involved in the pathological process of cuprizone-induced demyelination. First, we generated a mutant mouse strain lacking functional genes of both chemokines and demonstrated that double-mutant animals are viable, fertile, and do not present with gross abnormalities. Astrocytes and peritoneal macrophages, cultured form tissues of these animals did neither express CCL2 nor CCL3. Exposure to cuprizone resulted in increased CCL2 and CCL3 brain levels in wild-type but not mutant animals. Cuprizone-induced demyelination, oligodendrocyte loss, and astrogliosis were significantly ameliorated in the cortex but not corpus callosum of chemokine-deficient animals. In summary, we provide a novel powerful model to study the redundant function of two important chemokines. Our study reveals that chemokine function in the CNS redounds to region-specific pathophysiological events.
Collapse
Affiliation(s)
- Katharina Janssen
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mira Rickert
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Tim Clarner
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Markus Kipp
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
- Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany.
| |
Collapse
|
90
|
Anastasiadou S, Liebenehm S, Sinske D, Meyer zu Reckendorf C, Moepps B, Nordheim A, Knöll B. Neuronal expression of the transcription factor serum response factor modulates myelination in a mouse multiple sclerosis model. Glia 2015; 63:958-76. [PMID: 25639799 DOI: 10.1002/glia.22794] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/08/2015] [Accepted: 01/08/2015] [Indexed: 12/14/2022]
Abstract
In multiple sclerosis (MS), neurons in addition to inflammatory cells are now considered to mediate disease origin and progression. So far, molecular and cellular mechanisms of neuronal MS contributions are poorly understood. Herein we analyzed whether neuron-restricted signaling by the neuroprotective transcription factor serum response factor (SRF) modulates de- and remyelination in a rodent MS model. In the mouse cuprizone model, neuron- (Srf (flox/flox;CaMKCreERT2)) but not glia-specific (Srf (flox/flox;PlpCreERT2)) SRF depletion impaired demyelination suggesting impaired debris clearance by astrocytes and microglia. This supports an important role of SRF expression in neurons but not oligodendrocytes in de- and remyelination. During remyelination, NG2- and OLIG2-positive cells of the oligodendrocyte lineage as well as de novo mRNA synthesis of myelin genes were also reduced in neuron-specific Srf mutants. Using the stripe assay, we demonstrate that cortices of cuprizone-fed wild-type mice elicited astrocyte and microglia activation whereas this was abrogated in cuprizone-fed neuron-specific Srf mutants. We identified CCL chemokines (e.g. CCL2) as neuron-derived SRF-regulated paracrine signals rescuing immune cell activation upon neuronal SRF deletion. In summary, we uncovered important roles of neurons and neuronally expressed SRF in MS associated de- and remyelination.
Collapse
Affiliation(s)
- Sofia Anastasiadou
- Institute of Physiological Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | | | | | | | | | | | | |
Collapse
|
91
|
Slowik A, Schmidt T, Beyer C, Amor S, Clarner T, Kipp M. The sphingosine 1-phosphate receptor agonist FTY720 is neuroprotective after cuprizone-induced CNS demyelination. Br J Pharmacol 2014; 172:80-92. [PMID: 25220526 DOI: 10.1111/bph.12938] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/28/2014] [Accepted: 08/23/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Modulation of the sphingosine 1-phosphate receptor is an approved treatment for relapsing multiple sclerosis because of its anti-inflammatory effect of retaining lymphocytes within the lymph nodes. Here, we evaluated the potential of an agonist at this receptor, FTY720 (fingolimod), to activate the promyelinating pathways within the brain to encourage remyelination and neuroprotection. EXPERIMENTAL APPROACH In this study, we used the cuprizone model in male C57BL/6 mice and tested the promyelinating and neuroprotective effects of FTY720 after acute and chronic toxin-induced experimental demyelination. We used histological, immunohistochemical and gene expression methods. KEY RESULTS The midline of the corpus callosum was severely demyelinated after acute and chronic cuprizone-induced demyelination. Robust endogenous remyelination was evident after acute, but impaired after chronic, demyelination. FTY720 treatment modestly accelerated myelin recovery after acute but not chronic cuprizone exposure. Markers of gliosis (astrocyte and microglia activation) were not affected by FTY720 treatment. Remarkably, the accumulation of amyloid precursor protein-positive spheroids in axons was less distinct in FTY720-treated animals, indicating that this compound alleviated ongoing axonal damage. CONCLUSIONS AND IMPLICATIONS We show that even during endogenous remyelination, axonal degeneration continued at a low level, accumulating over time. This continuous neurodegenerative process was ameliorated by FTY720 treatment. FTY720 preserved CNS integrity by direct interaction with brain resident cells, the actions of which are still to be defined.
Collapse
Affiliation(s)
- A Slowik
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | |
Collapse
|
92
|
Oldekamp S, Pscheidl S, Kress E, Soehnlein O, Jansen S, Pufe T, Wang JM, Tauber SC, Brandenburg LO. Lack of formyl peptide receptor 1 and 2 leads to more severe inflammation and higher mortality in mice with of pneumococcal meningitis. Immunology 2014; 143:447-61. [PMID: 24863484 DOI: 10.1111/imm.12324] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 12/22/2022] Open
Abstract
Bacterial meningitis is, despite progress in research and the development of new treatment strategies, still a cause of severe neuronal sequelae. The brain is protected from penetrating pathogens by both the blood-brain barrier and the innate immune system. The invading pathogens are recognized by pattern recognition receptors including the G-protein coupled formyl peptide receptors (FPRs), which are expressed by immune cells of the central nervous system. The expression of FPRs is up-regulated during bacterial meningitis, but the consequence on the progression of inflammation and impact on mortality are far from clear. Therefore, we used mFPR1 and mFPR2-deficient mice to investigate the effects on inflammation, bacterial growth and mortality in a mouse model of pneumococcal meningitis. Our results revealed increased bacterial burden, increased neutrophil infiltration and higher mortality in mFPR1/2-deficient mice in comparison to wild-type mice. The mFPR1- or mFPR2-deficient mice also showed significantly increased glial cell density, whereas the immune responses including the expression of anti-inflammatory cytokines and antimicrobial peptides were decreased in bacterial meningitis. Taken together, the results suggest that FPR1 and FPR2 play an important role in the innate immune responses against Streptococcus pneumoniae within the central nervous system and the lack of the receptors leads to a dysregulation of the inflammatory response compared with wild-type mice.
Collapse
Affiliation(s)
- Sandra Oldekamp
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Krauspe BM, Dreher W, Beyer C, Baumgartner W, Denecke B, Janssen K, Langhans CD, Clarner T, Kipp M. Short-term cuprizone feeding verifies N-acetylaspartate quantification as a marker of neurodegeneration. J Mol Neurosci 2014; 55:733-48. [PMID: 25189319 DOI: 10.1007/s12031-014-0412-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 08/20/2014] [Indexed: 01/27/2023]
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) is a quantitative MR imaging technique often used to complement conventional MR imaging with specific metabolic information. A key metabolite is the amino acid derivative N-Acetylaspartate (NAA) which is an accepted marker to measure the extent of neurodegeneration in multiple sclerosis (MS) patients. NAA is catabolized by the enzyme aspartoacylase (ASPA) which is predominantly expressed in oligodendrocytes. Since the formation of MS lesions is paralleled by oligodendrocyte loss, NAA might accumulate in the brain, and therefore, the extent of neurodegeneration might be underestimated. In the present study, we used the well-characterized cuprizone model. There, the loss of oligodendrocytes is paralleled by a reduction in ASPA expression and activity as demonstrated by genome-wide gene expression analysis and enzymatic activity assays. Notably, brain levels of NAA were not increased as determined by gas chromatography-mass spectrometry and 1H-MRS. These important findings underpin the reliability of NAA quantification as a valid marker for the paraclinical determination of the extent of neurodegeneration, even under conditions of oligodendrocyte loss in which impaired metabolization of NAA is expected. Future studies have to reveal whether other enzymes are able to metabolize NAA or whether an excess of NAA is cleared by other mechanisms rather than enzymatic metabolism.
Collapse
Affiliation(s)
- Barbara Maria Krauspe
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Prins M, Dutta R, Baselmans B, Brevé JJP, Bol JGJM, Deckard SA, van der Valk P, Amor S, Trapp BD, de Vries HE, Drukarch B, van Dam AM. Discrepancy in CCL2 and CCR2 expression in white versus grey matter hippocampal lesions of Multiple Sclerosis patients. Acta Neuropathol Commun 2014; 2:98. [PMID: 25149422 PMCID: PMC4158064 DOI: 10.1186/s40478-014-0098-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 08/10/2014] [Indexed: 12/21/2022] Open
Abstract
A remarkable pathological difference between grey matter lesions (GML) and white matter lesions (WML) in Multiple Sclerosis (MS) patients is the paucity of infiltrating leukocytes in GML. To better understand these pathological differences, we hypothesize that the chemokine monocyte chemotactic protein-1 (MCP-1 or CCL2), of importance for leukocyte migration, and its receptor CCR2 are more abundantly expressed in WML than in GML of MS patients. To this end, we analyzed CCL2 and CCR2 expression in the hippocampus, comprising WML and GML,of post-mortem MS patients, and of control subjects. CCL2 and CCR2 mRNA were significantly increased in demyelinated MS hippocampus. Semi-quantification of CCL2 and CCR2 immunoreactivity showed that CCL2 is present in astrocytes only in active WML. CCR2 is upregulated in monocytes/macrophages or amoeboid microglia in active WML, and in ramified microglia in active GML, although to a lesser extent. As a follow-up, we observed a significantly increased CCL2 production by WM-, but not GM-derived astrocytes upon stimulation with bz-ATP in vitro. Finally, upon CCL2 stimulation, GM-derived microglia significantly increased their proliferation rate. We conclude that within hippocampal lesions, CCL2 expression is mainly restricted to WML, whereas the receptor CCR2 is upregulated in both WML and GML. The relative absence of CCL2 in GML may explain the lack of infiltrating immune cells in this type of lesions. We propose that the divergent expression of CCL2 and CCR2 in WML and GML explains or contributes to the differences in WML and GML formation in MS.
Collapse
Affiliation(s)
- Marloes Prins
- />Department of Anatomy and Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, Van der Boechorststraat 7, 1081 Amsterdam, BT The Netherlands
| | - Ranjan Dutta
- />Department of Neurosciences, Cleveland Clinic, Lerner Research Institute, VU University Medical Center, Cleveland, OH USA
| | - Bart Baselmans
- />Department of Anatomy and Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, Van der Boechorststraat 7, 1081 Amsterdam, BT The Netherlands
| | - John J P Brevé
- />Department of Anatomy and Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, Van der Boechorststraat 7, 1081 Amsterdam, BT The Netherlands
| | - John G J M Bol
- />Department of Anatomy and Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, Van der Boechorststraat 7, 1081 Amsterdam, BT The Netherlands
| | - Sadie A Deckard
- />Department of Neurosciences, Cleveland Clinic, Lerner Research Institute, VU University Medical Center, Cleveland, OH USA
| | - Paul van der Valk
- />Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Sandra Amor
- />Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- />Neuroimmunology Unit, Blizard Institute of Cell and Molecular Science, Barts and The London, School of Medicine and Dentistry, VU University Medical Center, London, UK
| | - Bruce D Trapp
- />Department of Neurosciences, Cleveland Clinic, Lerner Research Institute, VU University Medical Center, Cleveland, OH USA
| | - Helga E de Vries
- />Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Benjamin Drukarch
- />Department of Anatomy and Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, Van der Boechorststraat 7, 1081 Amsterdam, BT The Netherlands
| | - Anne-Marie van Dam
- />Department of Anatomy and Neurosciences, VU University Medical Center, Neuroscience Campus Amsterdam, Van der Boechorststraat 7, 1081 Amsterdam, BT The Netherlands
| |
Collapse
|
95
|
Protective Effect of a cAMP Analogue on Behavioral Deficits and Neuropathological Changes in Cuprizone Model of Demyelination. Mol Neurobiol 2014; 52:130-41. [PMID: 25128030 DOI: 10.1007/s12035-014-8857-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/06/2014] [Indexed: 01/06/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease that leads to neuronal cell loss. Cyclic AMP and its analogs are well known to decrease inflammation and apoptosis. In the present study, we examined the effects of bucladesine, a cell-permeable analogue of cyclic adenosine monophosphate (cAMP), on myelin proteins (PLP, PMP-22), inflammation, and apoptotic, as well as anti-apoptotic factors in cuprizone model of demyelination. C57BL/6J mice were fed with chow containing 0.2% copper chelator cuprizone or vehicle by daily oral gavage for 5 weeks to induce reversible demyelination predominantly of the corpus callosum. Bucladesine was administered intraperitoneally at different doses (0.24, 0.48, or 0.7 μg/kg body weight) during the last 7 days of 5-week cuprizone treatment. Bucladesine exhibited a protective effect on myelination. Furthermore, bucladesine significantly decreased the production of interleukin-6 pro-inflammatory mediator as well as nuclear factor-κB activation and reduced the mean number of apoptotic cells compared to cuprizone-treated mice. Bucladesine also decreased production of caspase-3 as well as Bax and increased Bcl-2 levels. Our data revealed that enhancement of intracellular cAMP prevents demyelination and plays anti-inflammatory and anti-apoptotic properties in mice cuprizone model of demyelination. This suggests the modulation of intracellular cAMP as a potential target for treatment of MS.
Collapse
|
96
|
Eroglu B, Kimbler DE, Pang J, Choi J, Moskophidis D, Yanasak N, Dhandapani KM, Mivechi NF. Therapeutic inducers of the HSP70/HSP110 protect mice against traumatic brain injury. J Neurochem 2014; 130:626-41. [PMID: 24903326 DOI: 10.1111/jnc.12781] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/09/2014] [Accepted: 05/28/2014] [Indexed: 12/30/2022]
Abstract
Traumatic brain injury (TBI) induces severe harm and disability in many accident victims and combat-related activities. The heat-shock proteins Hsp70/Hsp110 protect cells against death and ischemic damage. In this study, we used mice deficient in Hsp110 or Hsp70 to examine their potential requirement following TBI. Data indicate that loss of Hsp110 or Hsp70 increases brain injury and death of neurons. One of the mechanisms underlying the increased cell death observed in the absence of Hsp110 and Hsp70 following TBI is the increased expression of reactive oxygen species-induced p53 target genes Pig1, Pig8, and Pig12. To examine whether drugs that increase the levels of Hsp70/Hsp110 can protect cells against TBI, we subjected mice to TBI and administered Celastrol or BGP-15. In contrast to Hsp110- or Hsp70i-deficient mice that were not protected following TBI and Celastrol treatment, there was a significant improvement of wild-type mice following administration of these drugs during the first week following TBI. In addition, assessment of neurological injury shows significant improvement in contextual and cued fear conditioning tests and beam balance in wild-type mice that were treated with Celastrol or BGP-15 following TBI compared to TBI-treated mice. These studies indicate a significant role of Hsp70/Hsp110 in neuronal survival following TBI and the beneficial effects of Hsp70/Hsp110 inducers toward reducing the pathological consequences of TBI. Our data indicate that loss of Hsp110 or Hsp70 in mice increases brain injury following TBI. (a) One of the mechanisms underlying the increased cell death observed in the absence of these Hsps following TBI is the increased expression of ROS-induced p53 target genes known as Pigs. In addition, (b) using drugs (Celastrol or BGP-15) to increase Hsp70/Hsp110 levels protect cells against TBI, suggesting the beneficial effects of Hsp70/Hsp110 inducers to reduce the pathological consequences of TBI.
Collapse
Affiliation(s)
- Binnur Eroglu
- Charlie Norwood VA Medical Center (CNVAMC), Augusta, Georgia, USA; Molecular Chaperone Biology, Georgia Regents University, Augusta, Georgia, USA; Cancer Center, Georgia Regents University, Augusta, Georgia, USA; Georgia Regents University (GRU) and Medical College of Georgia, Augusta, Georgia, USA
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Nystad AE, Wergeland S, Aksnes L, Myhr KM, Bø L, Torkildsen Ø. Effect of high-dose 1.25 dihydroxyvitamin D3on remyelination in the cuprizone model. APMIS 2014; 122:1178-86. [DOI: 10.1111/apm.12281] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 03/10/2014] [Indexed: 01/12/2023]
Affiliation(s)
- Agnes E. Nystad
- Department of Neurology; Norwegian Multiple Sclerosis Competence Centre; Haukeland University Hospital; Bergen Norway
- Kristian Gerhard Jebsen MS Research Centre; Department of Clinical Medicine; University of Bergen; Bergen Norway
| | - Stig Wergeland
- Department of Neurology; Norwegian Multiple Sclerosis Competence Centre; Haukeland University Hospital; Bergen Norway
- Kristian Gerhard Jebsen MS Research Centre; Department of Clinical Medicine; University of Bergen; Bergen Norway
| | - Lage Aksnes
- Department of Clinical Science; University of Bergen; Bergen Norway
- Department of Pediatrics; Haukeland University Hospital; Bergen Norway
| | - Kjell-Morten Myhr
- Department of Neurology; Norwegian Multiple Sclerosis Competence Centre; Haukeland University Hospital; Bergen Norway
- Kristian Gerhard Jebsen MS Research Centre; Department of Clinical Medicine; University of Bergen; Bergen Norway
| | - Lars Bø
- Department of Neurology; Norwegian Multiple Sclerosis Competence Centre; Haukeland University Hospital; Bergen Norway
- Kristian Gerhard Jebsen MS Research Centre; Department of Clinical Medicine; University of Bergen; Bergen Norway
| | - Øivind Torkildsen
- Department of Neurology; Norwegian Multiple Sclerosis Competence Centre; Haukeland University Hospital; Bergen Norway
- Kristian Gerhard Jebsen MS Research Centre; Department of Clinical Medicine; University of Bergen; Bergen Norway
| |
Collapse
|
98
|
Gudi V, Gingele S, Skripuletz T, Stangel M. Glial response during cuprizone-induced de- and remyelination in the CNS: lessons learned. Front Cell Neurosci 2014; 8:73. [PMID: 24659953 PMCID: PMC3952085 DOI: 10.3389/fncel.2014.00073] [Citation(s) in RCA: 273] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 02/19/2014] [Indexed: 12/21/2022] Open
Abstract
Although astrogliosis and microglia activation are characteristic features of multiple sclerosis (MS) and other central nervous system (CNS) lesions the exact functions of these events are not fully understood. Animal models help to understand the complex interplay between the different cell types of the CNS and uncover general mechanisms of damage and repair of myelin sheaths. The so called cuprizone model is a toxic model of demyelination in the CNS white and gray matter, which lacks an autoimmune component. Cuprizone induces apoptosis of mature oligodendrocytes that leads to a robust demyelination and profound activation of both astrocytes and microglia with regional heterogeneity between different white and gray matter regions. Although not suitable to study autoimmune mediated demyelination, this model is extremely helpful to elucidate basic cellular and molecular mechanisms during de- and particularly remyelination independently of interactions with peripheral immune cells. Phagocytosis and removal of damaged myelin seems to be one of the major roles of microglia in this model and it is well known that removal of myelin debris is a prerequisite of successful remyelination. Furthermore, microglia provide several signals that support remyelination. The role of astrocytes during de- and remyelination is not well defined. Both supportive and destructive functions have been suggested. Using the cuprizone model we could demonstrate that there is an important crosstalk between astrocytes and microglia. In this review we focus on the role of glial reactions and interaction in the cuprizone model. Advantages and limitations of as well as its potential therapeutic relevance for the human disease MS are critically discussed in comparison to other animal models.
Collapse
Affiliation(s)
- Viktoria Gudi
- Department of Neurology, Hannover Medical SchoolHannover, Germany
| | - Stefan Gingele
- Department of Neurology, Hannover Medical SchoolHannover, Germany
| | | | - Martin Stangel
- Department of Neurology, Hannover Medical SchoolHannover, Germany
- Center for Systems NeuroscienceHannover, Germany
| |
Collapse
|
99
|
Galectin-3 controls the response of microglial cells to limit cuprizone-induced demyelination. Neurobiol Dis 2014; 62:441-55. [DOI: 10.1016/j.nbd.2013.10.023] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/10/2013] [Accepted: 10/23/2013] [Indexed: 11/23/2022] Open
|
100
|
Goldberg J, Daniel M, van Heuvel Y, Victor M, Beyer C, Clarner T, Kipp M. Short-term cuprizone feeding induces selective amino acid deprivation with concomitant activation of an integrated stress response in oligodendrocytes. Cell Mol Neurobiol 2013; 33:1087-98. [PMID: 23979168 PMCID: PMC11497941 DOI: 10.1007/s10571-013-9975-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 08/12/2013] [Indexed: 12/20/2022]
Abstract
Cuprizone [bis(cyclohexylidenehydrazide)]-induced toxic demyelination is an experimental approach frequently used to study de- and re-myelination in the central nervous system. In this model, mice are fed with the copper chelator cuprizone which leads to oligodendrocyte apoptosis and subsequent microgliosis, astrocytosis, and demyelination. The underlying mechanisms of cuprizone-induced oligodendrocyte death are still unknown. We analysed differences in amino acid levels after short-term cuprizone exposure (i.e., 4 days). Furthermore, an amino acid response (AAR) pathway activated in oligodendrocytes after cuprizone intoxication was evaluated. Short-term cuprizone exposure resulted in a selective decrease of alanine, glycine, and proline plasma levels, which was paralleled by an increase of apoptotic cells in the liver and a decrease of alanine aminotransferase in the serum. These parameters were paralleled by oligodendrocyte apoptosis and the induction of an AAR with increased expression of the transcription factors ATF-3 and ATF-4 (activating transcription factor-3 and -4). Immunohistochemistry revealed that ATF-3 is exclusively expressed by oligodendrocytes and localized to the nuclear compartment. Our results suggest that cuprizone-induced liver dysfunction results in amino acid starvation and in consequence to the activation of an AAR. We propose that this stress response modulates oligodendrocyte viability in the cuprizone animal model.
Collapse
Affiliation(s)
- Johannes Goldberg
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Moritz Daniel
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Yasemin van Heuvel
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Marion Victor
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Cordian Beyer
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Tim Clarner
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Markus Kipp
- Faculty of Medicine, Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| |
Collapse
|