1
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
2
|
Xie L, Cen LP, Li Y, Gilbert HY, Strelko O, Berlinicke C, Stavarache MA, Ma M, Wang Y, Cui Q, Kaplitt MG, Zack DJ, Benowitz LI, Yin Y. Monocyte-derived SDF1 supports optic nerve regeneration and alters retinal ganglion cells' response to Pten deletion. Proc Natl Acad Sci U S A 2022; 119:e2113751119. [PMID: 35394873 PMCID: PMC9169637 DOI: 10.1073/pnas.2113751119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/23/2022] [Indexed: 12/22/2022] Open
Abstract
Although mammalian retinal ganglion cells (RGCs) normally cannot regenerate axons nor survive after optic nerve injury, this failure is partially reversed by inducing sterile inflammation in the eye. Infiltrative myeloid cells express the axogenic protein oncomodulin (Ocm) but additional, as-yet-unidentified, factors are also required. We show here that infiltrative macrophages express stromal cell–derived factor 1 (SDF1, CXCL12), which plays a central role in this regard. Among many growth factors tested in culture, only SDF1 enhances Ocm activity, an effect mediated through intracellular cyclic AMP (cAMP) elevation and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) activation. SDF1 deficiency in myeloid cells (CXCL12flx/flxLysM-Cre−/+ mice) or deletion of the SDF1 receptor CXCR4 in RGCs (intraocular AAV2-Cre in CXCR4flx/flx mice) or SDF1 antagonist AMD3100 greatly suppresses inflammation-induced regeneration and decreases RGC survival to baseline levels. Conversely, SDF1 induces optic nerve regeneration and RGC survival, and, when combined with Ocm/cAMP, SDF1 increases axon regeneration to levels similar to those induced by intraocular inflammation. In contrast to deletion of phosphatase and tensin homolog (Pten), which promotes regeneration selectively from αRGCs, SDF1 promotes regeneration from non-αRGCs and enables the latter cells to respond robustly to Pten deletion; however, SDF1 surprisingly diminishes the response of αRGCs to Pten deletion. When combined with inflammation and Pten deletion, SDF1 enables many RGCs to regenerate axons the entire length of the optic nerve. Thus, SDF1 complements the effects of Ocm in mediating inflammation-induced regeneration and enables different RGC subtypes to respond to Pten deletion.
Collapse
Affiliation(s)
- Lili Xie
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Ling-Ping Cen
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Yiqing Li
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510085, China
| | - Hui-Ya Gilbert
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Oleksandr Strelko
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Mihaela A. Stavarache
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Madeline Ma
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Yongting Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Cui
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Larry I. Benowitz
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115
| | - Yuqin Yin
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| |
Collapse
|
3
|
Göttle P, Schichel K, Reiche L, Werner L, Zink A, Prigione A, Küry P. TLR4 Associated Signaling Disrupters as a New Means to Overcome HERV-W Envelope-Mediated Myelination Deficits. Front Cell Neurosci 2021; 15:777542. [PMID: 34887730 PMCID: PMC8650005 DOI: 10.3389/fncel.2021.777542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023] Open
Abstract
Myelin repair in the adult central nervous system (CNS) is driven by successful differentiation of resident oligodendroglial precursor cells (OPCs) and thus constitutes a neurodegenerative process capable to compensate for functional deficits upon loss of oligodendrocytes and myelin sheaths as it is observed in multiple sclerosis (MS). The human endogenous retrovirus type W (HERV-W) represents an MS-specific pathogenic entity, and its envelope (ENV) protein was previously identified as a negative regulator of OPC maturation—hence, it is of relevance in the context of diminished myelin repair. We here focused on the activity of the ENV protein and investigated how it can be neutralized for improved remyelination. ENV-mediated activation of toll like receptor 4 (TLR4) increases inducible nitric oxide synthase (iNOS) expression, prompts nitrosative stress, and results in myelin-associated deficits, such as decreased levels of oligodendroglial maturation marker expression and morphological alterations. The intervention of TLR4 surface expression represents a potential means to rescue such ENV-dependent deficits. To this end, the rescue capacity of specific substances, either modulating V-ATPase activity or myeloid differentiation 2 (MD2)-mediated TLR4 glycosylation status, such as compound 20 (C20), L48H437, or folimycin, was analyzed, as these processes were demonstrated to be relevant for TLR4 surface expression. We found that pharmacological treatment can rescue the maturation arrest of oligodendroglial cells and their myelination capacity and can prevent iNOS induction in the presence of the ENV protein. In addition, downregulation of TLR4 surface expression was observed. Furthermore, mitochondrial integrity crucial for oligodendroglial cell differentiation was affected in the presence of ENV and ameliorated upon pharmacological treatment. Our study, therefore, provides novel insights into possible means to overcome myelination deficits associated with HERV-W ENV-mediated myelin deficits.
Collapse
Affiliation(s)
- Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Kira Schichel
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Luisa Werner
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Annika Zink
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
4
|
Guo YS, Yuan M, Han Y, Shen XY, Gao ZK, Bi X. Therapeutic Potential of Cytokines in Demyelinating Lesions After Stroke. J Mol Neurosci 2021; 71:2035-2052. [PMID: 33970426 DOI: 10.1007/s12031-021-01851-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022]
Abstract
White matter damage is a component of most human stroke and usually accounts for at least half of the lesion volume. Subcortical white matter stroke (WMS) accounts for 25% of all strokes and causes severe motor and cognitive dysfunction. The adult brain has a very limited ability to repair white matter damage. Pathological analysis shows that demyelination or myelin loss is the main feature of white matter injury and plays an important role in long-term sensorimotor and cognitive dysfunction. This suggests that demyelination is a major therapeutic target for ischemic stroke injury. An acute inflammatory reaction is triggered by brain ischemia, which is accompanied by cytokine production. The production of cytokines is an important factor affecting demyelination and myelin regeneration. Different cytokines have different effects on myelin damage and myelin regeneration. Exploring the role of cytokines in demyelination and remyelination after stroke and the underlying molecular mechanisms of demyelination and myelin regeneration after ischemic injury is very important for the development of rehabilitation treatment strategies. This review focuses on recent findings on the effects of cytokines on myelin damage and remyelination as well as the progress of research on the role of cytokines in ischemic stroke prognosis to provide a new treatment approach for amelioration of white matter damage after stroke.
Collapse
Affiliation(s)
- Yi-Sha Guo
- Shanghai University of Sport, Shanghai, 200438, China
| | - Mei Yuan
- Shanghai University of Sport, Shanghai, 200438, China
| | - Yu Han
- Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-Ya Shen
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Zhen-Kun Gao
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China.
| |
Collapse
|
5
|
Reiche L, Göttle P, Lane L, Duek P, Park M, Azim K, Schütte J, Manousi A, Schira-Heinen J, Küry P. C21orf91 Regulates Oligodendroglial Precursor Cell Fate-A Switch in the Glial Lineage? Front Cell Neurosci 2021; 15:653075. [PMID: 33796011 PMCID: PMC8008080 DOI: 10.3389/fncel.2021.653075] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
Neuropathological diseases of the central nervous system (CNS) are frequently associated with impaired differentiation of the oligodendroglial cell lineage and subsequent alterations in white matter structure and dynamics. Down syndrome (DS), or trisomy 21, is the most common genetic cause for cognitive impairments and intellectual disability (ID) and is associated with a reduction in the number of neurons and oligodendrocytes, as well as with hypomyelination and astrogliosis. Recent studies mainly focused on neuronal development in DS and underestimated the role of glial cells as pathogenic players. This also relates to C21ORF91, a protein considered a key modulator of aberrant CNS development in DS. We investigated the role of C21orf91 ortholog in terms of oligodendrogenesis and myelination using database information as well as through cultured primary oligodendroglial precursor cells (OPCs). Upon modulation of C21orf91 gene expression, we found this factor to be important for accurate oligodendroglial differentiation, influencing their capacity to mature and to myelinate axons. Interestingly, C21orf91 overexpression initiates a cell population coexpressing astroglial- and oligodendroglial markers indicating that elevated C21orf91 expression levels induce a gliogenic shift towards the astrocytic lineage reflecting non-equilibrated glial cell populations in DS brains.
Collapse
Affiliation(s)
- Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Lydie Lane
- CALIPHO Group, SIB Swiss Institute of Bioinformatics, Geneva, Switzerland.,Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Paula Duek
- CALIPHO Group, SIB Swiss Institute of Bioinformatics, Geneva, Switzerland.,Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mina Park
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Kasum Azim
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jana Schütte
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anastasia Manousi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jessica Schira-Heinen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
Pouzol L, Baumlin N, Sassi A, Tunis M, Marrie J, Vezzali E, Farine H, Mentzel U, Martinic MM. ACT-1004-1239, a first-in-class CXCR7 antagonist with both immunomodulatory and promyelinating effects for the treatment of inflammatory demyelinating diseases. FASEB J 2021; 35:e21431. [PMID: 33595155 DOI: 10.1096/fj.202002465r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 12/26/2022]
Abstract
Current strategies for the treatment of demyelinating diseases such as multiple sclerosis (MS) are based on anti-inflammatory or immunomodulatory drugs. Those drugs have the potential to reduce the frequency of new lesions but do not directly promote remyelination in the damaged central nervous system (CNS). Targeting CXCR7 (ACKR3) has been postulated as a potential therapeutic approach in demyelinating diseases, leading to both immunomodulation by reducing leukocyte infiltrates and promyelination by enhancing myelin repair. ACT-1004-1239 is a potent, selective, insurmountable, and orally available first-in-class CXCR7 receptor antagonist. The effect of ACT-1004-1239 was evaluated in the myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) and the cuprizone-induced demyelination mouse models. In addition, ACT-1004-1239 was assessed in a rat oligodendrocyte precursor cell (OPC) differentiation assay in vitro. In the MOG-induced EAE model, ACT-1004-1239 treatment (10-100 mg/kg, twice daily, orally) showed a significant dose-dependent reduction in disease clinical scores, resulting in increased survival. At the highest dose tested (100 mg/kg, twice daily), ACT-1004-1239 delayed disease onset and significantly reduced immune cell infiltrates into the CNS and plasma neurofilament light chain concentration. Treatment with ACT-1004-1239 dose-dependently increased plasma CXCL12 concentration, which correlated with a reduction of the cumulative disease score. Furthermore, in the cuprizone model, ACT-1004-1239 treatment significantly increased the number of mature myelinating oligodendrocytes and enhanced myelination in vivo. In vitro, ACT-1004-1239 promoted the maturation of OPCs into myelinating oligodendrocytes. These results provide evidence that ACT-1004-1239 both reduces neuroinflammation and enhances myelin repair substantiating the rationale to explore its therapeutic potential in a clinical setting.
Collapse
Affiliation(s)
| | | | - Anna Sassi
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Mélanie Tunis
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Julia Marrie
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | - Hervé Farine
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | | |
Collapse
|
7
|
Manousi A, Göttle P, Reiche L, Cui QL, Healy LM, Akkermann R, Gruchot J, Schira-Heinen J, Antel JP, Hartung HP, Küry P. Identification of novel myelin repair drugs by modulation of oligodendroglial differentiation competence. EBioMedicine 2021; 65:103276. [PMID: 33714029 PMCID: PMC7970057 DOI: 10.1016/j.ebiom.2021.103276] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In multiple sclerosis loss of myelin and oligodendrocytes impairs saltatory signal transduction and leads to neuronal loss and functional deficits. Limited capacity of oligodendroglial precursor cells to differentiate into mature cells is the main reason for inefficient myelin repair in the central nervous system. Drug repurposing constitutes a powerful approach for identification of pharmacological compounds promoting this process. METHODS A phenotypic compound screening using the subcellular distribution of a potent inhibitor of oligodendroglial cell differentiation, namely p57kip2, as differentiation competence marker was conducted. Hit compounds were validated in terms of their impact on developmental cell differentiation and myelination using both rat and human primary cell cultures and organotypic cerebellar slice cultures, respectively. Their effect on spontaneous remyelination was then investigated following cuprizone-mediated demyelination of the corpus callosum. FINDINGS A number of novel small molecules able to promote oligodendroglial cell differentiation were identified and a subset was found to foster human oligodendrogenesis as well as myelination ex vivo. Among them the steroid danazol and the anthelminthic parbendazole were found to increase myelin repair. INTERPRETATION We provide evidence that early cellular processes involved in differentiation decisions are applicable for the identification of regeneration promoting drugs and we suggest danazol and parbendazole as potent therapeutic candidates for demyelinating diseases. FUNDING This work was supported by the Jürgen Manchot Foundation, Düsseldorf; Research Commission of the Medical Faculty of Heinrich-Heine-University Düsseldorf; Christiane and Claudia Hempel Foundation; Stifterverband/Novartisstiftung; James and Elisabeth Cloppenburg, Peek and Cloppenburg Düsseldorf Stiftung and International Progressive MS Alliance (BRAVEinMS).
Collapse
Affiliation(s)
- Anastasia Manousi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Qiao-Ling Cui
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H4A 3K9, Canada
| | - Luke M Healy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H4A 3K9, Canada
| | - Rainer Akkermann
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Joel Gruchot
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Jessica Schira-Heinen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany
| | - Jack P Antel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H4A 3K9, Canada
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany; Brain and Mind Centre, University of Sydney, Camperdown NSW 2050, Australia
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225, Germany.
| |
Collapse
|
8
|
Vaes JEG, Brandt MJV, Wanders N, Benders MJNL, de Theije CGM, Gressens P, Nijboer CH. The impact of trophic and immunomodulatory factors on oligodendrocyte maturation: Potential treatments for encephalopathy of prematurity. Glia 2020; 69:1311-1340. [PMID: 33595855 PMCID: PMC8246971 DOI: 10.1002/glia.23939] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Encephalopathy of prematurity (EoP) is a major cause of morbidity in preterm neonates, causing neurodevelopmental adversities that can lead to lifelong impairments. Preterm birth-related insults, such as cerebral oxygen fluctuations and perinatal inflammation, are believed to negatively impact brain development, leading to a range of brain abnormalities. Diffuse white matter injury is a major hallmark of EoP and characterized by widespread hypomyelination, the result of disturbances in oligodendrocyte lineage development. At present, there are no treatment options available, despite the enormous burden of EoP on patients, their families, and society. Over the years, research in the field of neonatal brain injury and other white matter pathologies has led to the identification of several promising trophic factors and cytokines that contribute to the survival and maturation of oligodendrocytes, and/or dampening neuroinflammation. In this review, we discuss the current literature on selected factors and their therapeutic potential to combat EoP, covering a wide range of in vitro, preclinical and clinical studies. Furthermore, we offer a future perspective on the translatability of these factors into clinical practice.
Collapse
Affiliation(s)
- Josine E G Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Myrna J V Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Nikki Wanders
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | | | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
9
|
Fumagalli A, Heuninck J, Pizzoccaro A, Moutin E, Koenen J, Séveno M, Durroux T, Junier MP, Schlecht-Louf G, Bachelerie F, Schütz D, Stumm R, Smit MJ, Guérineau NC, Chaumont-Dubel S, Marin P. The atypical chemokine receptor 3 interacts with Connexin 43 inhibiting astrocytic gap junctional intercellular communication. Nat Commun 2020; 11:4855. [PMID: 32978390 PMCID: PMC7519114 DOI: 10.1038/s41467-020-18634-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The atypical chemokine receptor 3 (ACKR3) plays a pivotal role in directing the migration of various cellular populations and its over-expression in tumors promotes cell proliferation and invasiveness. The intracellular signaling pathways transducing ACKR3-dependent effects remain poorly characterized, an issue we addressed by identifying the interactome of ACKR3. Here, we report that recombinant ACKR3 expressed in HEK293T cells recruits the gap junction protein Connexin 43 (Cx43). Cx43 and ACKR3 are co-expressed in mouse brain astrocytes and human glioblastoma cells and form a complex in embryonic mouse brain. Functional in vitro studies show enhanced ACKR3 interaction with Cx43 upon ACKR3 agonist stimulation. Furthermore, ACKR3 activation promotes β-arrestin2- and dynamin-dependent Cx43 internalization to inhibit gap junctional intercellular communication in primary astrocytes. These results demonstrate a functional link between ACKR3 and gap junctions that might be of pathophysiological relevance. The atypical chemokine receptor 3 (ACKR3) is known to regulate cell migration, but the underlying mechanisms are unclear. Here, the authors show, from an interactome analysis, ACKR3 association with the gap junction protein Connexin 43 in vivo and ACKR3-mediated inhibition of astrocyte gap junctional communication.
Collapse
Affiliation(s)
- Amos Fumagalli
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Heuninck
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Pizzoccaro
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Enora Moutin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Koenen
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France.,Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Martial Séveno
- Biocampus Montpellier, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Marie-Pierre Junier
- CNRS UMR8246, Inserm U1130, Neuroscience Paris Seine-IBPS, Sorbonne Universités, Paris, France
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Francoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Séverine Chaumont-Dubel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
10
|
Dietrich M, Koska V, Hecker C, Göttle P, Hilla AM, Heskamp A, Lepka K, Issberner A, Hallenberger A, Baksmeier C, Steckel J, Balk L, Knier B, Korn T, Havla J, Martínez-Lapiscina EH, Solà-Valls N, Manogaran P, Olbert ED, Schippling S, Cruz-Herranz A, Yiu H, Button J, Caldito NG, von Gall C, Mausberg AK, Stettner M, Zimmermann HG, Paul F, Brandt AU, Küry P, Goebels N, Aktas O, Berndt C, Saidha S, Green AJ, Calabresi PA, Fischer D, Hartung HP, Albrecht P. Protective effects of 4-aminopyridine in experimental optic neuritis and multiple sclerosis. Brain 2020; 143:1127-1142. [PMID: 32293668 DOI: 10.1093/brain/awaa062] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/08/2019] [Accepted: 01/20/2020] [Indexed: 12/30/2022] Open
Abstract
Chronic disability in multiple sclerosis is linked to neuroaxonal degeneration. 4-aminopyridine (4-AP) is used and licensed as a symptomatic treatment to ameliorate ambulatory disability in multiple sclerosis. The presumed mode of action is via blockade of axonal voltage gated potassium channels, thereby enhancing conduction in demyelinated axons. In this study, we provide evidence that in addition to those symptomatic effects, 4-AP can prevent neuroaxonal loss in the CNS. Using in vivo optical coherence tomography imaging, visual function testing and histologic assessment, we observed a reduction in retinal neurodegeneration with 4-AP in models of experimental optic neuritis and optic nerve crush. These effects were not related to an anti-inflammatory mode of action or a direct impact on retinal ganglion cells. Rather, histology and in vitro experiments indicated 4-AP stabilization of myelin and oligodendrocyte precursor cells associated with increased nuclear translocation of the nuclear factor of activated T cells. In experimental optic neuritis, 4-AP potentiated the effects of immunomodulatory treatment with fingolimod. As extended release 4-AP is already licensed for symptomatic multiple sclerosis treatment, we performed a retrospective, multicentre optical coherence tomography study to longitudinally compare retinal neurodegeneration between 52 patients on continuous 4-AP therapy and 51 matched controls. In line with the experimental data, during concurrent 4-AP therapy, degeneration of the macular retinal nerve fibre layer was reduced over 2 years. These results indicate disease-modifying effects of 4-AP beyond symptomatic therapy and provide support for the design of a prospective clinical study using visual function and retinal structure as outcome parameters.
Collapse
Affiliation(s)
- Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Valeria Koska
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexander M Hilla
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Annemarie Heskamp
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Klaudia Lepka
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Angelika Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Christine Baksmeier
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Steckel
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Lisanne Balk
- Department of Neurology, Amsterdam Neuroscience, MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Benjamin Knier
- Department of Experimental Neuroimmunology, Technische Universität München, Munich, Germany
| | - Thomas Korn
- Department of Experimental Neuroimmunology, Technische Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians University, Munich, Germany.,Data Integration for Future Medicine consortium (DIFUTURE), Ludwig-Maximilians University, Munich, Germany
| | - Elena H Martínez-Lapiscina
- Service of Neurology, Hospital Clinic, University of Barcelona, Spain Neuroimmunology Program, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Solà-Valls
- Service of Neurology, Hospital Clinic, University of Barcelona, Spain Neuroimmunology Program, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Praveena Manogaran
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland.,Department of Information Technology and Electrical Engineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Elisabeth D Olbert
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Sven Schippling
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University Hospital Zürich and University of Zürich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Andrés Cruz-Herranz
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Hao Yiu
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Julia Button
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | | | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Anne K Mausberg
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Mark Stettner
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Hannah G Zimmermann
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Alexander U Brandt
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrueck Center for Molecular Medicine, Berlin, Germany.,Department of Neurology, University of California, Irvine, USA
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Norbert Goebels
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Shiv Saidha
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | - Ari J Green
- Division of Neuroinflammation and Glial Biology, Department of Neurology, University of California San Francisco, San Francisco, USA.,Department of Ophthalmology, University of California San Francisco, San Francisco, USA
| | - Peter A Calabresi
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Baltimore, USA
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
11
|
Watson AES, Goodkey K, Footz T, Voronova A. Regulation of CNS precursor function by neuronal chemokines. Neurosci Lett 2020; 715:134533. [DOI: 10.1016/j.neulet.2019.134533] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
|
12
|
Gruchot J, Weyers V, Göttle P, Förster M, Hartung HP, Küry P, Kremer D. The Molecular Basis for Remyelination Failure in Multiple Sclerosis. Cells 2019; 8:cells8080825. [PMID: 31382620 PMCID: PMC6721708 DOI: 10.3390/cells8080825] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022] Open
Abstract
Myelin sheaths in the central nervous system (CNS) insulate axons and thereby allow saltatory nerve conduction, which is a prerequisite for complex brain function. Multiple sclerosis (MS), the most common inflammatory autoimmune disease of the CNS, leads to the destruction of myelin sheaths and the myelin-producing oligodendrocytes, thus leaving behind demyelinated axons prone to injury and degeneration. Clinically, this process manifests itself in significant neurological symptoms and disability. Resident oligodendroglial precursor cells (OPCs) and neural stem cells (NSCs) are present in the adult brain, and can differentiate into mature oligodendrocytes which then remyelinate the demyelinated axons. However, for multiple reasons, in MS the regenerative capacity of these cell populations diminishes significantly over time, ultimately leading to neurodegeneration, which currently remains untreatable. In addition, microglial cells, the resident innate immune cells of the CNS, can contribute further to inflammatory and degenerative axonal damage. Here, we review the molecular factors contributing to remyelination failure in MS by inhibiting OPC and NSC differentiation or modulating microglial behavior.
Collapse
Affiliation(s)
- Joel Gruchot
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Vivien Weyers
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Moritz Förster
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany.
| |
Collapse
|
13
|
pHERV-W envelope protein fuels microglial cell-dependent damage of myelinated axons in multiple sclerosis. Proc Natl Acad Sci U S A 2019; 116:15216-15225. [PMID: 31213545 DOI: 10.1073/pnas.1901283116] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Axonal degeneration is central to clinical disability and disease progression in multiple sclerosis (MS). Myeloid cells such as brain-resident microglia and blood-borne monocytes are thought to be critically involved in this degenerative process. However, the exact underlying mechanisms have still not been clarified. We have previously demonstrated that human endogenous retrovirus type W (HERV-W) negatively affects oligodendroglial precursor cell (OPC) differentiation and remyelination via its envelope protein pathogenic HERV-W (pHERV-W) ENV (formerly MS-associated retrovirus [MSRV]-ENV). In this current study, we investigated whether pHERV-W ENV also plays a role in axonal injury in MS. We found that in MS lesions, pHERV-W ENV is present in myeloid cells associated with axons. Focusing on progressive disease stages, we could then demonstrate that pHERV-W ENV induces a degenerative phenotype in microglial cells, driving them toward a close spatial association with myelinated axons. Moreover, in pHERV-W ENV-stimulated myelinated cocultures, microglia were found to structurally damage myelinated axons. Taken together, our data suggest that pHERV-W ENV-mediated microglial polarization contributes to neurodegeneration in MS. Thus, this analysis provides a neurobiological rationale for a recently completed clinical study in MS patients showing that antibody-mediated neutralization of pHERV-W ENV exerts neuroprotective effects.
Collapse
|
14
|
Yao F, Li Z, Cheng L, Zhang L, Zha X, Jing J. Low frequency pulsed electromagnetic field promotes differentiation of oligodendrocyte precursor cells through upregulation of miR-219-5p in vitro. Life Sci 2019; 223:185-193. [PMID: 30885522 DOI: 10.1016/j.lfs.2019.03.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 12/19/2022]
Abstract
AIM Spinal cord injury (SCI) is a common demyelinating disorder of the central nervous system. The differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes (OLs), which induce myelination, plays a critical role in the functional recovery following SCI. In this study, the effect of low frequency pulsed electromagnetic field (PEMF) on the differentiation of OPCs and the potential underlying mechanisms were investigated. MAIN METHODS OPCs were randomly divided into the PEMF and non-PEMF (NPEMF) groups. Immunofluorescence and western blot assays were performed to assess the expression levels of OLs stage-specific markers after 3, 7, 14, and 21 days of PEMF or NPEMF exposure. qRT-PCR was used to further assess the expression levels of miR-219-5p, miR-338, miR-138, and miR-9, which are associated with OPCs differentiation, and the expression levels of genes associated with miR-219-5p. Finally, following PEMF or NPEMF exposure, qRT-PCR and western blot assays were performed to explore the relationship between miR-219-5p and Lingo1 and between miR-219-5p and PEMF in promoting OPCs differentiation. KEY FINDINGS PEMF promoted the differentiation of OPCs. PEMF upregulated the expression level of miR-219-5p and downregulated the expression level of Lingo1 during the differentiation of OPCs. Under PEMF exposure, miR-219-5p targeted Lingo1 and reversed the inhibitory effect of miR-219-5p inhibitor on OPCs differentiation. In addition, PEMF synergized with miR-219-5p to promote OPCs differentiation. SIGNIFICANCE Our results, for the first time, indicated that PEMF promoted OPCs differentiation by regulating miR-219-5p activity in vitro.
Collapse
Affiliation(s)
- Fei Yao
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, 678 FuRong Road, Hefei, Anhui Province 230601, China
| | - Ziyu Li
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, 678 FuRong Road, Hefei, Anhui Province 230601, China
| | - Li Cheng
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, 678 FuRong Road, Hefei, Anhui Province 230601, China
| | - Liqian Zhang
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, 678 FuRong Road, Hefei, Anhui Province 230601, China
| | - Xiaowei Zha
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, 678 FuRong Road, Hefei, Anhui Province 230601, China
| | - Juehua Jing
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, 678 FuRong Road, Hefei, Anhui Province 230601, China.
| |
Collapse
|
15
|
Harnisch K, Teuber-Hanselmann S, Macha N, Mairinger F, Fritsche L, Soub D, Meinl E, Junker A. Myelination in Multiple Sclerosis Lesions Is Associated with Regulation of Bone Morphogenetic Protein 4 and Its Antagonist Noggin. Int J Mol Sci 2019; 20:ijms20010154. [PMID: 30609838 PMCID: PMC6337410 DOI: 10.3390/ijms20010154] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/22/2018] [Accepted: 12/27/2018] [Indexed: 11/25/2022] Open
Abstract
Remyelination is a central aspect of new multiple sclerosis (MS) therapies, in which one aims to alleviate disease symptoms by improving axonal protection. However, a central problem is mediators expressed in MS lesions that prevent effective remyelination. Bone morphogenetic protein4 (BMP4) inhibits the development of mature oligodendrocytes in cell culture and also blocks the expression of myelin proteins. Additionally, numerous studies have shown that Noggin (SYM1)—among other physiological antagonists of BMP4—plays a prominent role in myelin formation in the developing but also the adult central nervous system. Nonetheless, neither BMP4 nor Noggin have been systematically studied in human MS lesions. In this study, we demonstrated by transcript analysis and immunohistochemistry that BMP4 is expressed by astrocytes and microglia/macrophages in association with inflammatory infiltrates in MS lesions, and that astrocytes also express BMP4 in chronic inactive lesions that failed to remyelinate. Furthermore, the demonstration of an increased expression of Noggin in so-called shadow plaques (i.e., remyelinated lesions with thinner myelin sheaths) in comparison to chronically inactive demyelinated lesions implies that antagonizing BMP4 is associated with successful remyelination in MS plaques in humans. However, although BMP4 is strongly overexpressed in inflammatory lesion areas, its levels are also elevated in remyelinated lesion areas, which raises the possibility that BMP4 signaling itself may be required for remyelination. Therefore, remyelination might be influenced by a small number of key factors. Manipulating these molecules, i.e., BMP4 and Noggin, could be a promising therapeutic approach for effective remyelination.
Collapse
Affiliation(s)
- Kim Harnisch
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | | | - Nicole Macha
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Lena Fritsche
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Daniel Soub
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians-Universität München, D-82152 Martinsried, Germany.
| | - Andreas Junker
- Institute of Neuropathology, University Hospital Essen, D-45147 Essen, Germany.
| |
Collapse
|
16
|
Muñoz-Esquivel J, Göttle P, Aguirre-Cruz L, Flores-Rivera J, Corona T, Reyes-Terán G, Küry P, Torres KJ. Sildenafil Inhibits Myelin Expression and Myelination of Oligodendroglial Precursor Cells. ASN Neuro 2019; 11:1759091419832444. [PMID: 30849920 PMCID: PMC6410393 DOI: 10.1177/1759091419832444] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/21/2019] [Accepted: 01/25/2019] [Indexed: 12/15/2022] Open
Abstract
Phosphodiesterases (PDEs) have previously been implicated in oligodendrocyte maturation and myelination of central nervous system axons. Sildenafil citrate is a phosphodiesterase inhibitor known to block PDE5, which also reduces inflammation in the experimental autoimmune encephalomyelitis demyelinating model. To find out whether this inhibitor might exert beneficial effects on central nervous system myelin repair activities, we investigated to what degree sildenafil modulates differentiation and maturation of cultured primary rat oligodendroglial precursor cells (OPCs). To this end, gene and protein expression of 2',3'-cyclic-nucleotide 3'-phosphodiesterase, myelin basic protein, and myelin oligodendrocyte glycoprotein, as well as of negative regulators of myelin expression (Hes1, Hes5, Id2, Id4, Rock2, and p57Kip2) were measured in OPCs treated with sildenafil. Moreover, the subcellular distribution of the p57kip2 protein was determined after sildenafil treatment, as this revealed to be an early predictor of the oligodendroglial differentiation capacity. In vitro myelination assays were done to measure the myelination capacity of oligodendrocytes treated with sildenafil. We found that sildenafil significantly diminished myelin gene expression and protein expression. Moreover, sildenafil also increased the expression of Id2 and Id4 negative transcriptional regulators, and the degree of OPCs with cytoplasmic p57kip2 protein localization was reduced, providing evidence that the PDE blocker impaired the differentiation capacity. Finally, sildenafil also interfered with the establishment of internodes as revealed by in vitro myelination assays. We therefore conclude that blocking PDE5 activities exerts a negative impact on intrinsic oligodendroglial differentiation processes.
Collapse
Affiliation(s)
- Jonathan Muñoz-Esquivel
- Laboratory of Neuroimmunoendocrinology, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico
- *These authors contributed equally to this work
| | - Peter Göttle
- Neuroregeneration, Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
- *These authors contributed equally to this work
| | - Lucinda Aguirre-Cruz
- Laboratory of Neuroimmunoendocrinology, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico
| | - José Flores-Rivera
- Clinical Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico
| | - Teresa Corona
- Clinical Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico
| | - Gustavo Reyes-Terán
- Infectious Diseases Research Department, National Institute of Respiratory Diseases Ismael Cossio Villegas, Mexico
| | - Patrick Küry
- Neuroregeneration, Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany
- These authors are joint senior authors
| | - Klintsy J. Torres
- Laboratory of Neuroimmunoendocrinology, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Mexico
- Infectious Diseases Research Department, National Institute of Respiratory Diseases Ismael Cossio Villegas, Mexico
- These authors are joint senior authors
| |
Collapse
|
17
|
Neuroinflammation in preterm babies and autism spectrum disorders. Pediatr Res 2019; 85:155-165. [PMID: 30446768 DOI: 10.1038/s41390-018-0208-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 12/23/2022]
Abstract
Genetic anomalies have a role in autism spectrum disorders (ASD). Each genetic factor is responsible for a small fraction of cases. Environment factors, like preterm delivery, have an important role in ASD. Preterm infants have a 10-fold higher risk of developing ASD. Preterm birth is often associated with maternal/fetal inflammation, leading to a fetal/neonatal inflammatory syndrome. There are demonstrated experimental links between fetal inflammation and the later development of behavioral symptoms consistent with ASD. Preterm infants have deficits in connectivity. Most ASD genes encode synaptic proteins, suggesting that ASD are connectivity pathologies. Microglia are essential for normal synaptogenesis. Microglia are diverted from homeostatic functions towards inflammatory phenotypes during perinatal inflammation, impairing synaptogenesis. Preterm infants with ASD have a different phenotype from term born peers. Our original hypothesis is that exposure to inflammation in preterm infants, combined with at risk genetic background, deregulates brain development leading to ASD.
Collapse
|
18
|
Göttle P, Förster M, Gruchot J, Kremer D, Hartung HP, Perron H, Küry P. Rescuing the negative impact of human endogenous retrovirus envelope protein on oligodendroglial differentiation and myelination. Glia 2018; 67:160-170. [PMID: 30430656 DOI: 10.1002/glia.23535] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022]
Abstract
Remyelination in the adult CNS depends on activation, differentiation, and functional integration of resident oligodendroglial precursor cells (OPCs) and constitutes the only spontaneous neuroregenerative process able to compensate for functional deficits upon loss of oligodendrocytes and myelin sheaths as it is observed in multiple sclerosis. The proteins encoded by p57kip2- and by human endogenous retrovirus type W (pHERV-W) envelope genes were previously identified as negative regulators of OPC maturation. We here focused on the activity of the ENV protein and investigated how it can be neutralized for an improved myelin repair. We could demonstrate that myelination in vitro is severely affected by this protein but that application of an anti-ENV neutralizing antibody, currently investigated in clinical trials, can rescue the generation of internodes. We then compared p57kip2 and ENV dependent inhibitory mechanisms and found that a dominant negative version of the p57kip2 protein can equally save OPCs from myelination failure in response to ENV-mediated TLR4 activation. Additional experiments addressing p57kip2's underlying mode of action revealed a direct interaction with ATP6v1d, a central component of a vascular ATPase. Its pharmacological blocking was then shown to exert an analogous myelination rescue effect in presence of the ENV protein. Therefore, our study provides mechanistic insights into oligodendroglial inhibition processes and presents three different means to counteract the anti-myelination effect of the ENV protein. These observations are therefore of interest in light of understanding the complexity of the numerous oligodendroglial inhibitors and might promote the establishment of novel regenerative therapies.
Collapse
Affiliation(s)
- Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Moritz Förster
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Joel Gruchot
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hervé Perron
- GeNeuro, Chemin du Pré-Fleuri 3, Geneva, Switzerland
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
19
|
Rangon CM, Schang AL, Van Steenwinckel J, Schwendimann L, Lebon S, Fu T, Chen L, Beneton V, Journiac N, Young-Ten P, Bourgeois T, Maze J, Matrot B, Baburamani AA, Supramaniam V, Mallard C, Trottet L, Edwards AD, Hagberg H, Fleiss B, Li J, Chuang TT, Gressens P. Myelination induction by a histamine H3 receptor antagonist in a mouse model of preterm white matter injury. Brain Behav Immun 2018; 74:265-276. [PMID: 30218783 DOI: 10.1016/j.bbi.2018.09.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/17/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Fifteen million babies are born preterm every year and a significant number suffer from permanent neurological injuries linked to white matter injury (WMI). A chief cause of preterm birth itself and predictor of the severity of WMI is exposure to maternal-fetal infection-inflammation such as chorioamnionitis. There are no neurotherapeutics for this WMI. To affect this healthcare need, the repurposing of drugs with efficacy in other white matter injury models is an attractive strategy. As such, we tested the efficacy of GSK247246, an H3R antagonist/inverse agonist, in a model of inflammation-mediated WMI of the preterm born infant recapitulating the main clinical hallmarks of human brain injury, which are oligodendrocyte maturation arrest, microglial reactivity, and hypomyelination. WMI is induced by mimicking the effects of maternal-fetal infection-inflammation and setting up neuroinflammation. We induce this process at the time in the mouse when brain development is equivalent to the human third trimester; postnatal day (P)1 through to P5 with i.p. interleukin-1β (IL-1β) injections. We initiated GSK247246 treatment (i.p at 7 mg/kg or 20 mg/kg) after neuroinflammation was well established (on P6) and it was administered twice daily through to P10. Outcomes were assessed at P10 and P30 with gene and protein analysis. A low dose of GSK247246 (7 mg/kg) lead to a recovery in protein expression of markers of myelin (density of Myelin Basic Protein, MBP & Proteolipid Proteins, PLP) and a reduction in macro- and microgliosis (density of ionising adaptor protein, IBA1 & glial fibrillary acid protein, GFAP). Our results confirm the neurotherapeutic efficacy of targeting the H3R for WMI seen in a cuprizone model of multiple sclerosis and a recently reported clinical trial in relapsing-remitting multiple sclerosis patients. Further work is needed to develop a slow release strategy for this agent and test its efficacy in large animal models of preterm infant WMI.
Collapse
Affiliation(s)
- Claire-Marie Rangon
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Anne-Laure Schang
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France; UMR CNRS 8638-Chimie Toxicologie Analytique et Cellulaire, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie de Paris, 4 Avenue de l'Observatoire, F-75006 Paris, France
| | - Juliette Van Steenwinckel
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Leslie Schwendimann
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Sophie Lebon
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Tingting Fu
- Platform Technologies and Science, GlaxoSmithKline R&D, Shanghai 201203, China; Platform Technologies and Science, GlaxoSmithKline R&D, Stevenage, SG1 2NY, UK
| | - Libo Chen
- Platform Technologies and Science, GlaxoSmithKline R&D, Shanghai 201203, China; Platform Technologies and Science, GlaxoSmithKline R&D, Stevenage, SG1 2NY, UK
| | | | - Nathalie Journiac
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Pierrette Young-Ten
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Thomas Bourgeois
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Johanna Maze
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Boris Matrot
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France
| | - Ana A Baburamani
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Veena Supramaniam
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | | | - A David Edwards
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Henrik Hagberg
- Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom; Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden; Department of Clinical Sciences, Sahlgrenska Academy/East Hospital, 416 85 Gothenburg, Sweden
| | - Bobbi Fleiss
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France; Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom; School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| | - Jingjun Li
- Regenerative Medicine DPU, GlaxoSmithKline, Shanghai 201023, China; Regenerative Medicine DPU, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Tsu Tshen Chuang
- Regenerative Medicine DPU, GlaxoSmithKline, Shanghai 201023, China; Regenerative Medicine DPU, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Pierre Gressens
- PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, F-75019 Paris, France; PremUP, F-75006 Paris, France; Centre for the Developing Brain, School of Biomedical Engineering & Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
20
|
Santos AK, Vieira MS, Vasconcellos R, Goulart VAM, Kihara AH, Resende RR. Decoding cell signalling and regulation of oligodendrocyte differentiation. Semin Cell Dev Biol 2018; 95:54-73. [PMID: 29782926 DOI: 10.1016/j.semcdb.2018.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes are fundamental for the functioning of the nervous system; they participate in several cellular processes, including axonal myelination and metabolic maintenance for astrocytes and neurons. In the mammalian nervous system, they are produced through waves of proliferation and differentiation, which occur during embryogenesis. However, oligodendrocytes and their precursors continue to be generated during adulthood from specific niches of stem cells that were not recruited during development. Deficiencies in the formation and maturation of these cells can generate pathologies mainly related to myelination. Understanding the mechanisms involved in oligodendrocyte development, from the precursor to mature cell level, will allow inferring therapies and treatments for associated pathologies and disorders. Such mechanisms include cell signalling pathways that involve many growth factors, small metabolic molecules, non-coding RNAs, and transcription factors, as well as specific elements of the extracellular matrix, which act in a coordinated temporal and spatial manner according to a given stimulus. Deciphering those aspects will allow researchers to replicate them in vitro in a controlled environment and thus mimic oligodendrocyte maturation to understand the role of oligodendrocytes in myelination in pathologies and normal conditions. In this study, we review these aspects, based on the most recent in vivo and in vitro data on oligodendrocyte generation and differentiation.
Collapse
Affiliation(s)
- A K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - M S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - R Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - V A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - A H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - R R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
21
|
Yuan F, Chang S, Luo L, Li Y, Wang L, Song Y, Qu M, Zhang Z, Yang GY, Wang Y. cxcl12 gene engineered endothelial progenitor cells further improve the functions of oligodendrocyte precursor cells. Exp Cell Res 2018; 367:222-231. [PMID: 29614310 DOI: 10.1016/j.yexcr.2018.03.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 11/18/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) are needed for white matter repair after various brain injury. Means that promote OPC functions could benefit white matter recovery after injury. Chemokine CXCL12 and endothelial progenitor cells (EPCs) both have been shown to promote remyelination. We hypothesize that the beneficial effects of EPCs and CXCL12 can be harnessed by genetically modifying EPCs with cxcl12 to synergistically improve the functions of OPCs. In this work, CXCL12-EPC was generated using virus-mediated gene transfer. OPCs were cultured with CXCL12-EPC conditioned media (CM) to analyze its impact on the proliferation, migration, differentiation and survival properties of OPCs. We blocked or knocked-down the receptors of CXCL12, namely CXCR4 and CXCR7, respectively to investigate their functions in regulating OPCs properties. Results revealed that CXCL12-EPC CM further promoted OPCs behavioral properties and upregulated the expression of PDGFR-α, bFGF, CXCR4 and CXCR7 in OPCs, albeit following different time course. Blocking CXCR4 diminished the beneficial effects of CXCL12 on OPCs proliferation and migration, while knocking down CXCR7 inhibited OPCs differentiation. Our results supported that cxcl12 gene modification of EPCs further promoted EPCs' ability in augmenting the remyelination properties of OPCs, suggesting that CXCL12-EPC hold great potential in white matter repair.
Collapse
Affiliation(s)
- Fang Yuan
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shuang Chang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Longlong Luo
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaning Li
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liping Wang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yaying Song
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Meijie Qu
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
22
|
Göttle P, Manousi A, Kremer D, Reiche L, Hartung HP, Küry P. Teriflunomide promotes oligodendroglial differentiation and myelination. J Neuroinflammation 2018; 15:76. [PMID: 29534752 PMCID: PMC5851312 DOI: 10.1186/s12974-018-1110-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/28/2018] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a neuroinflammatory autoimmune disease of the central nervous system (CNS) which in most cases initially presents with episodes of transient functional deficits (relapsing-remitting MS; RRMS) and eventually develops into a secondary progressive form (SPMS). Aside from neuroimmunological activities, MS is also characterized by neurodegenerative and regenerative processes. The latter involve the restoration of myelin sheaths-electrically insulating structures which are the primary targets of autoimmune attacks. Spontaneous endogenous remyelination takes place even in the adult CNS and is primarily mediated by activation, recruitment, and differentiation of resident oligodendroglial precursor cells (OPCs). However, the overall efficiency of remyelination is limited and further declines with disease duration and progression. From a therapeutic standpoint, it is therefore key to understand how oligodendroglial maturation can be modulated pharmacologically. Teriflunomide has been approved as a first-line treatment for RRMS in the USA and the European Union. As the active metabolite of leflunomide, an established disease-modifying anti-rheumatic drug, it mainly acts via an inhibition of de novo pyrimidine synthesis exerting a cytostatic effect on proliferating B and T cells. METHODS We investigated teriflunomide-dependent effects on primary rat oligodendroglial homeostasis, proliferation, and differentiation related to cellular processes important for myelin repair hence CNS regeneration in vitro. To this end, several cellular parameters, including specific oligodendroglial maturation markers, in vitro myelination, and p53 family member signaling, were examined by means of gene/protein expression analyses. The rate of myelination was determined using neuron-oligodendrocyte co-cultures. RESULTS Low teriflunomide concentrations resulted in cell cycle exit while higher doses led to decreased cell survival. Short-term teriflunomide pulses can efficiently promote oligodendroglial cell differentiation suggesting that young, immature cells could benefit from such stimulation. In vitro myelination can be boosted by means of an early stimulation window with teriflunomide. p73 signaling is functionally involved in promoting OPC differentiation and myelination. CONCLUSION Our findings indicate a critical window of opportunity during which regenerative oligodendroglial activities including myelination of CNS axons can be stimulated by teriflunomide.
Collapse
Affiliation(s)
- Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - Anastasia Manousi
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany
| |
Collapse
|
23
|
Kucharska-Mazur J, Jabłoński M, Misiak B, Frydecka D, Rybakowski J, Ratajczak MZ, Samochowiec J. Adult stem cells in psychiatric disorders - New discoveries in peripheral blood. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:23-27. [PMID: 28392482 DOI: 10.1016/j.pnpbp.2017.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/28/2017] [Accepted: 04/05/2017] [Indexed: 12/15/2022]
Abstract
The new area of research in psychiatric disorders is concerned with abnormal regeneration processes. The role of brain neurogenesis has been studied for decades. New discoveries, concerned with the pluripotency of VSEL cells and the role of factors involved in stem cell trafficking in peripheral blood create hope that it will be possible to develop a better understanding of the processes of neuroregeneration/neurodegeneration. There is an ongoing research investigating concentrations of: sphingosine -1-phosphate, SDF-1, elements of complement cascade, and stem cells in peripheral blood, including their possible connection to psychiatric disorders. Collected data, suggesting an abnormal course of regeneration processes in psychiatric disorders, raises hope of finding new potential markers of psychosis and anxiety disorders.
Collapse
Affiliation(s)
- Jolanta Kucharska-Mazur
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460 Szczecin, Poland
| | - Marcin Jabłoński
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460 Szczecin, Poland
| | - Błażej Misiak
- Department of Genetics, Wroclaw Medical University, Marcinkowskiego 1, 50-368 Wrocław, Poland
| | - Dorota Frydecka
- Department of Psychiatry, Wroclaw Medical University, Pasteur 10, 50-367 Wroclaw, Poland
| | - Janusz Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572 Poznan, Poland
| | | | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian University of Medicine, Broniewskiego 26, 71-460 Szczecin, Poland.
| |
Collapse
|
24
|
Jadasz JJ, Tepe L, Beyer F, Samper Agrelo I, Akkermann R, Spitzhorn LS, Silva ME, Oreffo ROC, Hartung HP, Prigione A, Rivera FJ, Adjaye J, Küry P. Human mesenchymal factors induce rat hippocampal- and human neural stem cell dependent oligodendrogenesis. Glia 2017; 66:145-160. [PMID: 28940767 DOI: 10.1002/glia.23233] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/04/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022]
Abstract
The generation of new oligodendrocytes is essential for adult brain repair in diseases such as multiple sclerosis. We previously identified the multifunctional p57kip2 protein as a negative regulator of myelinating glial cell differentiation and as an intrinsic switch of glial fate decision in adult neural stem cells (aNSCs). In oligodendroglial precursor cells (OPCs), p57kip2 protein nuclear exclusion was recently found to be rate limiting for differentiation to proceed. Furthermore, stimulation with mesenchymal stem cell (MSC)-derived factors enhanced oligodendrogenesis by yet unknown mechanisms. To elucidate this instructive interaction, we investigated to what degree MSC secreted factors are species dependent, whether hippocampal aNSCs respond equally well to such stimuli, whether apart from oligodendroglial differentiation also tissue integration and axonal wrapping can be promoted and whether the oligodendrogenic effect involved subcellular translocation of p57kip2. We found that CC1 positive oligodendrocytes within the hilus express nuclear p57kip2 protein and that MSC dependent stimulation of cultured hippocampal aNSCs was not accompanied by nuclear p57kip2 exclusion as observed for parenchymal OPCs after spontaneous differentiation. Stimulation with human MSC factors was observed to equally promote rat stem cell oligodendrogenesis, axonal wrapping and tissue integration. As forced nuclear shuttling of p57kip2 led to decreased CNPase- but elevated GFAP expression levels, this indicates heterogenic oligodendroglial mechanisms occurring between OPCs and aNSCs. We also show for the first time that dominant pro-oligodendroglial factors derived from human fetal MSCs can instruct human induced pluripotent stem cell-derived NSCs to differentiate into O4 positive oligodendrocytes.
Collapse
Affiliation(s)
- Janusz J Jadasz
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Lena Tepe
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Felix Beyer
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Iria Samper Agrelo
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Rainer Akkermann
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Lucas-Sebastian Spitzhorn
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, D-40225, Germany
| | - Maria Elena Silva
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Institute of Pharmacy, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Hans-Peter Hartung
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | | | - Francisco J Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Institute of Molecular Regenerative Medicine & Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, D-40225, Germany
| | - Patrick Küry
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| |
Collapse
|
25
|
Chu T, Shields LBE, Zhang YP, Feng SQ, Shields CB, Cai J. CXCL12/CXCR4/CXCR7 Chemokine Axis in the Central Nervous System: Therapeutic Targets for Remyelination in Demyelinating Diseases. Neuroscientist 2017; 23:627-648. [PMID: 29283028 DOI: 10.1177/1073858416685690] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The chemokine CXCL12 plays a vital role in regulating the development of the central nervous system (CNS) by binding to its receptors CXCR4 and CXCR7. Recent studies reported that the CXCL12/CXCR4/CXCR7 axis regulates both embryonic and adult oligodendrocyte precursor cells (OPCs) in their proliferation, migration, and differentiation. The changes in the expression and distribution of CXCL12 and its receptors are tightly associated with the pathological process of demyelination in multiple sclerosis (MS), suggesting that modulating the CXCL12/CXCR4/CXCR7 axis may benefit myelin repair by enhancing OPC recruitment and differentiation. This review aims to integrate the current findings of the CXCL12/CXCR4/CXCR7 signaling pathway in the CNS and to highlight its role in oligodendrocyte development and demyelinating diseases. Furthermore, this review provides potential therapeutic strategies for myelin repair by analyzing the relevance between the pathological changes and the regulatory roles of CXCL12/CXCR4/CXCR7 during MS.
Collapse
Affiliation(s)
- Tianci Chu
- 1 Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Lisa B E Shields
- 2 Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, USA
| | - Yi Ping Zhang
- 2 Norton Neuroscience Institute, Norton Healthcare, Louisville, KY, USA
| | - Shi-Qing Feng
- 3 Department of Orthopedics, General Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | | | - Jun Cai
- 1 Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.,4 Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
26
|
Lindsay SL, Johnstone SA, McGrath MA, Mallinson D, Barnett SC. Comparative miRNA-Based Fingerprinting Reveals Biological Differences in Human Olfactory Mucosa- and Bone-Marrow-Derived Mesenchymal Stromal Cells. Stem Cell Reports 2016; 6:729-742. [PMID: 27117785 PMCID: PMC4940454 DOI: 10.1016/j.stemcr.2016.03.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/22/2022] Open
Abstract
Previously we reported that nestin-positive human mesenchymal stromal cells (MSCs) derived from the olfactory mucosa (OM) enhanced CNS myelination in vitro to a greater extent than bone-marrow-derived MSCs (BM-MSCs). miRNA-based fingerprinting revealed the two MSCs were 64% homologous, with 26 miRNAs differentially expressed. We focused on miR-146a-5p and miR-140-5p due to their reported role in the regulation of chemokine production and myelination. The lower expression of miR-140-5p in OM-MSCs correlated with higher secretion of CXCL12 compared with BM-MSCs. Addition of CXCL12 and its pharmacological inhibitors to neural co-cultures supported these data. Studies on related miR-146a-5p targets demonstrated that OM-MSCs had lower levels of Toll-like receptors and secreted less pro-inflammatory cytokines, IL-6, IL-8, and CCL2. OM-MSCs polarized microglia to an anti-inflammatory phenotype, illustrating potential differences in their inflammatory response. Nestin-positive OM-MSCs could therefore offer a cell transplantation alternative for CNS repair, should these biological behaviors be translated in vivo. OM-MSCs share 64% miRNA homology to BM-MSCs and differentially express 26 miRNAs CXCL12 promotes CNS myelination and is negatively regulated by miR-140-5p in BM-MSCs miR-146a-5p negatively regulates IL-6, IL-8, TLR2, and TLR4 on OM-MSCs These properties make OM-MSCs a suitable candidate for transplant-mediated CNS repair
Collapse
Affiliation(s)
- Susan Louise Lindsay
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Steven Andrew Johnstone
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Michael Anthony McGrath
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - David Mallinson
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK; Sistemic UK, Kelvin Campus, Maryhill Road, Glasgow G20 0SP, UK
| | - Susan Carol Barnett
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
27
|
Bianco J, De Berdt P, Deumens R, des Rieux A. Taking a bite out of spinal cord injury: do dental stem cells have the teeth for it? Cell Mol Life Sci 2016; 73:1413-37. [PMID: 26768693 PMCID: PMC11108394 DOI: 10.1007/s00018-015-2126-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/16/2015] [Accepted: 12/22/2015] [Indexed: 12/15/2022]
Abstract
Dental stem cells are an emerging star on a stage that is already quite populated. Recently, there has been a lot of hype concerning these cells in dental therapies, especially in regenerative endodontics. It is fitting that most research is concentrated on dental regeneration, although other uses for these cells need to be explored in more detail. Being a true mesenchymal stem cell, their capacities could also prove beneficial in areas outside their natural environment. One such field is the central nervous system, and in particular, repairing the injured spinal cord. One of the most formidable challenges in regenerative medicine is to restore function to the injured spinal cord, and as yet, a cure for paralysis remains to be discovered. A variety of approaches have already been tested, with graft-based strategies utilising cells harbouring appropriate properties for neural regeneration showing encouraging results. Here we present a review focusing on properties of dental stem cells that endorse their use in regenerative medicine, with particular emphasis on repairing the damaged spinal cord.
Collapse
Affiliation(s)
- John Bianco
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier, 73, B1 73.12, 1200, Brussels, Belgium.
- Integrated Center for Cell Therapy and Regenerative Medicine, International Clinical Research Center (FNUSA-ICRC), St. Anne's University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic.
| | - Pauline De Berdt
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier, 73, B1 73.12, 1200, Brussels, Belgium
| | - Ronald Deumens
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
| | - Anne des Rieux
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier, 73, B1 73.12, 1200, Brussels, Belgium
- Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, 1348, Louvain-La-Neuve, Belgium
| |
Collapse
|
28
|
Zilkha-Falb R, Kaushansky N, Kawakami N, Ben-Nun A. Post-CNS-inflammation expression of CXCL12 promotes the endogenous myelin/neuronal repair capacity following spontaneous recovery from multiple sclerosis-like disease. J Neuroinflammation 2016; 13:7. [PMID: 26747276 PMCID: PMC4706716 DOI: 10.1186/s12974-015-0468-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/26/2015] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Demyelination and axonal degeneration, hallmarks of multiple sclerosis (MS), are associated with the central nervous system (CNS) inflammation facilitated by C-X-C motif chemokine 12 (CXCL12) chemokine. Both in MS and in experimental autoimmune encephalomyelitis (EAE), the deleterious CNS inflammation has been associated with upregulation of CXCL12 expression in the CNS. We investigated the expression dynamics of CXCL12 in the CNS with progression of clinical EAE and following spontaneous recovery, with a focus on CXCL12 expression in the hippocampal neurogenic dentate gyrus (DG) and in the corpus callosum (CC) of spontaneously recovered mice, and its potential role in promoting the endogenous myelin/neuronal repair capacity. METHODS CNS tissue sections from mice with different clinical EAE phases or following spontaneous recovery and in vitro differentiated adult neural stem cell cultures were analyzed by immunofluorescent staining and confocal imaging for detecting and enumerating neuronal progenitor cells (NPCs) and oligodendrocyte precursor cells (OPCs) and for expression of CXCL12. RESULTS Our expression dynamics analysis of CXCL12 in the CNS with EAE progression revealed elevated CXCL12 expression in the DG and CC, which persistently increases following spontaneous recovery even though CNS inflammation has subsided. Correspondingly, the numbers of NPCs and OPCs in the DG and CC, respectively, of EAE-recovered mice increased compared to that of naïve mice (NPCs, p < 0.0001; OPCs, p < 0.00001) or mice with active disease (OPCs, p < 0.0005). Notably, about 30 % of the NPCs and unexpectedly also OPCs (~50 %) express CXCL12, and their numbers in DG and CC, respectively, are higher in EAE-recovered mice compared with naïve mice and also compared with mice with ongoing clinical EAE (CXCL12(+) NPCs, p < 0.005; CXCL12(+) OPCs, p < 0.0005). Moreover, a significant proportion (>20 %) of the CXCL12(+) NPCs and OPCs co-express the CXCL12 receptor, CXCR4, and their numbers significantly increase with recovery from EAE not only relative to naïve mice (p < 0.0002) but also to mice with ongoing EAE (p < 0.004). CONCLUSIONS These data link CXCL12 expression in the DG and CC of EAE-recovering mice to the promotion of neuro/oligodendrogenesis generating CXCR4(+) CXCL12(+) neuronal and oligodendrocyte progenitor cells endowed with intrinsic neuro/oligondendroglial differentiation potential. These findings highlight the post-CNS-inflammation role of CXCL12 in augmenting the endogenous myelin/neuronal repair capacity in MS-like disease, likely via CXCL12/CXCR4 autocrine signaling.
Collapse
Affiliation(s)
- Rina Zilkha-Falb
- Present address: Multiple Sclerosis Center, Neurogenomics Laboratory, Sheba Medical Center, Tel-Hashomer, Israel.
| | - Nathali Kaushansky
- Department of Immunology, The Weizmann Institute of Science, 234 Herzl Street, Rehovot, 7610001, Israel
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians-University, 81377, Munich, Germany.
| | - Avraham Ben-Nun
- Department of Immunology, The Weizmann Institute of Science, 234 Herzl Street, Rehovot, 7610001, Israel.
| |
Collapse
|
29
|
Kremer D, Cui QL, Göttle P, Kuhlmann T, Hartung HP, Antel J, Küry P. CXCR7 Is Involved in Human Oligodendroglial Precursor Cell Maturation. PLoS One 2016; 11:e0146503. [PMID: 26741980 PMCID: PMC4704822 DOI: 10.1371/journal.pone.0146503] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/17/2015] [Indexed: 11/18/2022] Open
Abstract
Differentiation of oligodendroglial precursor cells (OPCs), a crucial prerequisite for central nervous system (CNS) remyelination in diseases such as Multiple Sclerosis (MS), is modulated by a multitude of extrinsic and intrinsic factors. In a previous study we revealed that the chemokine CXCL12 stimulates rodent OPC differentiation via activation of its receptor CXCR7. We could now demonstrate that CXCR7 is also expressed on NogoA- and Nkx2.2-positive oligodendroglial cells in human MS brains and that stimulation of cultured primary fetal human OPCs with CXCL12 promotes their differentiation as measured by surface marker expression and morphologic complexity. Pharmacological inhibition of CXCR7 effectively blocks these CXCL12-dependent effects. Our findings therefore suggest that a specific activation of CXCR7 could provide a means to promote oligodendroglial differentiation facilitating endogenous remyelination activities.
Collapse
Affiliation(s)
- David Kremer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- * E-mail:
| | - Qiao-Ling Cui
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jack Antel
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
30
|
Itoh K, Maki T, Lok J, Arai K. Mechanisms of cell-cell interaction in oligodendrogenesis and remyelination after stroke. Brain Res 2015; 1623:135-49. [PMID: 25960351 PMCID: PMC4569526 DOI: 10.1016/j.brainres.2015.04.039] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/19/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022]
Abstract
White matter damage is a clinically important aspect of several central nervous system diseases, including stroke. Cerebral white matter primarily consists of axonal bundles ensheathed with myelin secreted by mature oligodendrocytes, which play an important role in neurotransmission between different areas of gray matter. During the acute phase of stroke, damage to oligodendrocytes leads to white matter dysfunction through the loss of myelin. On the contrary, during the chronic phase, white matter components promote an environment, which is favorable for neural repair, vascular remodeling, and remyelination. For effective remyelination to take place, oligodendrocyte precursor cells (OPCs) play critical roles by proliferating and differentiating into mature oligodendrocytes, which help to decrease the burden of axonal injury. Notably, other types of cells contribute to these OPC responses under the ischemic conditions. This mini-review summarizes the non-cell autonomous mechanisms in oligodendrogenesis and remyelination after white matter damage, focusing on how OPCs receive support from their neighboring cells. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Kanako Itoh
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
31
|
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: 117] [Impact Index Per Article: 11.7] [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
|
32
|
Abstract
Oligodendrocyte precursor cells (OPCs) originate in the ventricular zones (VZs) of the brain and spinal cord and migrate throughout the developing central nervous system (CNS) before differentiating into myelinating oligodendrocytes (OLs). It is not known whether OPCs or OLs from different parts of the VZ are functionally distinct. OPCs persist in the postnatal CNS, where they continue to divide and generate myelinating OLs at a decreasing rate throughout adult life in rodents. Adult OPCs respond to injury or disease by accelerating their cell cycle and increasing production of OLs to replace lost myelin. They also form synapses with unmyelinated axons and respond to electrical activity in those axons by generating more OLs and myelin locally. This experience-dependent "adaptive" myelination is important in some forms of plasticity and learning, for example, motor learning. We review the control of OL lineage development, including OL population dynamics and adaptive myelination in the adult CNS.
Collapse
Affiliation(s)
- Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, WBSB 1001, Baltimore, Maryland 21205
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
33
|
Schira J, Falkenberg H, Hendricks M, Waldera-Lupa DM, Kögler G, Meyer HE, Müller HW, Stühler K. Characterization of Regenerative Phenotype of Unrestricted Somatic Stem Cells (USSC) from Human Umbilical Cord Blood (hUCB) by Functional Secretome Analysis. Mol Cell Proteomics 2015; 14:2630-43. [PMID: 26183719 DOI: 10.1074/mcp.m115.049312] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 12/13/2022] Open
Abstract
Stem cell transplantation is a promising therapeutic strategy to enhance axonal regeneration after spinal cord injury. Unrestricted somatic stem cells (USSC) isolated from human umbilical cord blood is an attractive stem cell population available at GMP grade without any ethical concerns. It has been shown that USSC transplantation into acute injured rat spinal cords leads to axonal regrowth and significant locomotor recovery, yet lacking cell replacement. Instead, USSC secrete trophic factors enhancing neurite growth of primary cortical neurons in vitro. Here, we applied a functional secretome approach characterizing proteins secreted by USSC for the first time and validated candidate neurite growth promoting factors using primary cortical neurons in vitro. By mass spectrometric analysis and exhaustive bioinformatic interrogation we identified 1156 proteins representing the secretome of USSC. Using Gene Ontology we revealed that USSC secretome contains proteins involved in a number of relevant biological processes of nerve regeneration such as cell adhesion, cell motion, blood vessel formation, cytoskeleton organization and extracellular matrix organization. We found for instance that 31 well-known neurite growth promoting factors like, e.g. neuronal growth regulator 1, NDNF, SPARC, and PEDF span the whole abundance range of USSC secretome. By the means of primary cortical neurons in vitro assays we verified SPARC and PEDF as significantly involved in USSC mediated neurite growth and therewith underline their role in improved locomotor recovery after transplantation. From our data we are convinced that USSC are a valuable tool in regenerative medicine as USSC's secretome contains a comprehensive network of trophic factors supporting nerve regeneration not only by a single process but also maintained its regenerative phenotype by a multitude of relevant biological processes.
Collapse
Affiliation(s)
- Jessica Schira
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany; §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Heiner Falkenberg
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Marion Hendricks
- §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Daniel M Waldera-Lupa
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Gesine Kögler
- ¶Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Medical Center, Düsseldorf, Germany
| | - Helmut E Meyer
- ‖Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund, Germany
| | - Hans Werner Müller
- §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; **Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Kai Stühler
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany; **Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| |
Collapse
|
34
|
Gorris R, Fischer J, Erwes KL, Kesavan J, Peterson DA, Alexander M, Nöthen MM, Peitz M, Quandel T, Karus M, Brüstle O. Pluripotent stem cell-derived radial glia-like cells as stable intermediate for efficient generation of human oligodendrocytes. Glia 2015; 63:2152-67. [PMID: 26123132 DOI: 10.1002/glia.22882] [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] [Received: 12/19/2014] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 11/10/2022]
Abstract
Neural precursor cells (NPCs) derived from human pluripotent stem cells (hPSCs) represent an attractive tool for the in vitro generation of various neural cell types. However, the developmentally early NPCs emerging during hPSC differentiation typically show a strong propensity for neuronal differentiation, with more limited potential for generating astrocytes and, in particular, for generating oligodendrocytes. This phenomenon corresponds well to the consecutive and protracted generation of neurons and GLIA during normal human development. To obtain a more gliogenic NPC type, we combined growth factor-mediated expansion with pre-exposure to the differentiation-inducing agent retinoic acid and subsequent immunoisolation of CD133-positive cells. This protocol yields an adherent and self-renewing population of hindbrain/spinal cord radial glia (RG)-like neural precursor cells (RGL-NPCs) expressing typical neural stem cell markers such as nestin, ASCL1, SOX2, and PAX6 as well as RG markers BLBP, GLAST, vimentin, and GFAP. While RGL-NPCs maintain the ability for tripotential differentiation into neurons, astrocytes, and oligodendrocytes, they exhibit greatly enhanced propensity for oligodendrocyte generation. Under defined differentiation conditions promoting the expression of the major oligodendrocyte fate-determinants OLIG1/2, NKX6.2, NKX2.2, and SOX10, RGL-NPCs efficiently convert into NG2-positive oligodendroglial progenitor cells (OPCs) and are subsequently capable of in vivo myelination. Representing a stable intermediate between PSCs and OPCs, RGL-NPCs expedite the generation of PSC-derived oligodendrocytes with O4-, 4860-, and myelin basic protein (MBP)-positive cells that already appear within 7 weeks following growth factor withdrawal-induced differentiation. Thus, RGL-NPCs may serve as robust tool for time-efficient generation of human oligodendrocytes from embryonic and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Raphaela Gorris
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Julia Fischer
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Kim Lina Erwes
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Jaideep Kesavan
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Daniel A Peterson
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany.,Center for Stem Cell and Regenerative Medicine, Department of Neuroscience, Rosalind Franklin University of Medicine and Science, Chicago, Illinois
| | - Michael Alexander
- Institute of Human Genetics, LIFE & BRAIN Center, University of Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, LIFE & BRAIN Center, University of Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany.,German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Tamara Quandel
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Michael Karus
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn, Germany
| |
Collapse
|
35
|
Banisadr G, Podojil JR, Miller SD, Miller RJ. Pattern of CXCR7 Gene Expression in Mouse Brain Under Normal and Inflammatory Conditions. J Neuroimmune Pharmacol 2015; 11:26-35. [PMID: 25997895 DOI: 10.1007/s11481-015-9616-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 05/06/2015] [Indexed: 12/20/2022]
Abstract
The chemokine stromal cell-derived factor-1 (SDF-1)/CXCL12 acting via its G-protein coupled receptor (GPCR) CXCR4 has been implicated in neurogenesis, neuromodulation, brain inflammation, HIV-1 encephalopathy and tumor growth. CXCR7 was identified as an alternate receptor for SDF-1/CXCL12. Characterization of CXCR7-deficient mice demonstrated a role for CXCR7 in fetal endothelial biology, cardiac development, and B-cell localization. Despite its ligand binding properties, CXCR7 does not seem to signal like a conventional GPCR. It has been suggested that CXCR7 may not function alone but in combination with CXCR4. Here, we investigated the regional localization of CXCR7 receptors in adult mouse brain using CXCR7-EGFP transgenic mice. We found that the receptors were expressed in various brain regions including olfactory bulb, cerebral cortex, hippocampus, subventricular zone (SVZ), hypothalamus and cerebellum. Extensive CXCR7 expression was associated with cerebral blood vessels. Using cell type specific markers, CXCR7 expression was found in neurons, astrocytes and oligodendrocyte progenitors. GAD-expressing neurons exhibited CXCR7 expression in the hippocampus. Expression of CXCR7 in the dentate gyrus included cells that expressed nestin, GFAP and cells that appeared to be immature granule cells. In mice with Experimental Autoimmune Encephalomyelitis (EAE), CXCR7 was expressed by migrating oligodendrocyte progenitors in the SVZ. We then compared the distribution of SDF-1/CXCL12 and CXCR7 using bitransgenic mice expressing both CXCR7-EGFP and SDF-1-mRFP. Enhanced expression of SDF-1/CXCL12 and CXCR7 was observed in the corpus callosum, SVZ and cerebellum. Overall, the expression of CXCR7 in normal and pathological nervous system suggests CXCR4-independent functions of SDF-1/CXCL12 mediated through its interaction with CXCR7.
Collapse
Affiliation(s)
- Ghazal Banisadr
- Department of Pharmacology, Northwestern University Medical School, 303 E Superior Ave, Chicago, IL, 60611, USA.
| | - Joseph R Podojil
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E Chicago Ave, Chicago, IL, 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E Chicago Ave, Chicago, IL, 60611, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University Medical School, 303 E Superior Ave, Chicago, IL, 60611, USA
| |
Collapse
|
36
|
Abstract
Multiple sclerosis is an autoimmune disease of the CNS resulting in degeneration of myelin sheaths and loss of oligodendrocytes, which means that protection and electrical insulation of axons and rapid signal propagation are impaired, leading to axonal damage and permanent disabilities. Partial replacement of lost oligodendrocytes and remyelination can occur as a result of activation and recruitment of resident oligodendroglial precursor cells. However, the overall remyelination capacity remains inefficient because precursor cells often fail to generate new oligodendrocytes. Increasing evidence points to the existence of several molecular inhibitors that act on these cells and interfere with their cellular maturation. The p57kip2 gene encodes one such potent inhibitor of oligodendroglial differentiation and this study sheds light on the underlying mode of action. We found that subcellular distribution of the p57kip2 protein changed during differentiation of rat, mouse, and human oligodendroglial cells both in vivo and in vitro. Nuclear export of p57kip2 was correlated with promoted myelin expression, higher morphological phenotypes, and enhanced myelination in vitro. In contrast, nuclear accumulation of p57kip2 resulted in blocked oligodendroglial differentiation. Experimental evidence suggests that the inhibitory role of p57kip2 depends on specific interactions with binding proteins such as LIMK-1, CDK2, Mash1, and Hes5 either by controlling their site of action or their activity. Because functional restoration in demyelinating diseases critically depends on the successful generation of oligodendroglial cells, a therapeutic need that is currently unmet, the regulatory mechanism described here might be of particular interest for identifying suitable drug targets and devising novel therapeutic approaches.
Collapse
|
37
|
Bianco AM, Uno M, Oba-Shinjo SM, Clara CA, de Almeida Galatro TF, Rosemberg S, Teixeira MJ, Nagahashi Marie SK. CXCR7 and CXCR4 Expressions in Infiltrative Astrocytomas and Their Interactions with HIF1α Expression and IDH1 Mutation. Pathol Oncol Res 2014; 21:229-40. [PMID: 24970694 DOI: 10.1007/s12253-014-9813-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 06/04/2014] [Indexed: 11/27/2022]
Abstract
The CXCR7, a new receptor for CXCL12 with higher affinity than CXCR4 has raised key issues on glioma cell migration. The aim of this study is to investigate the CXCR7 mRNA expression in diffuse astrocytomas tissues and to evaluate its interactions with CXCR4 and HIF1α expression and IDH1 mutation. CXCR7, CXCR4 and HIF1α mRNA expression were evaluated in 129 frozen samples of astrocytomas. IDH1 mutation status was analyzed with gene expressions, matched with clinicopathological parameters and overall survival time. Protein expression was analyzed by immunohistochemistry in different grades of astrocytoma and in glioma cell line (U87MG) by confocal microscopy. There was significant difference in the expression levels of the genes studied between astrocytomas and non-neoplasic (NN) controls (p < 0.001). AGII showed no significant correlation between CXCR7/HIF1α (p = 0.548); there was significant correlation between CXCR7/CXCR4 (p = 0.042) and CXCR7/IDH1 (p = 0.008). GBM showed significant correlations between CXCR7/CXCR4 (p = 0.002), CXCR7/IDH1 (p < 0.001) and CXCR7/HIF1α (p = 0.008). HIF1α overexpression was associated with higher expressions of CXCR7 (p = 0.01) and CXCR4 (p < 0.0001), while IDH1 mutation was associated with lower CXCR7 (p = 0.009) and CXCR4 (p = 0.0005) mRNA expressions. Protein expression increased with malignancy and in U87MG cell line was mainly localized in the cellular membrane. CXCR7 was overexpressed in astrocytoma and correlates with CXCR4 and IDH1 in AGII and CXCR4, IDH1 and HIF1α in GBM. Overexpression HIF1α was related with higher expressions of CXCR7 and CXCR4, otherwise IDH1 mutation related with lower expression of both genes. No association between CXCR7 and CXCR4 expression and survival data was related.
Collapse
Affiliation(s)
- Andre Macedo Bianco
- Department of Neurology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil Laboratory of Molecular and Cellular Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, Sao Paulo, SP, Brazil, 01246-903,
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang Y, Fu W, Zhang S, He X, Liu Z, Gao D, Xu T. CXCR-7 receptor promotes SDF-1α-induced migration of bone marrow mesenchymal stem cells in the transient cerebral ischemia/reperfusion rat hippocampus. Brain Res 2014; 1575:78-86. [PMID: 24924806 DOI: 10.1016/j.brainres.2014.05.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 05/19/2014] [Accepted: 05/22/2014] [Indexed: 12/17/2022]
Abstract
The stromal cell-derived factor 1/C-X-C chemokine receptor type 4 (SDF-1/CXCR-4) axis plays an important role during stem cell recruitment. SDF-1 can also bind the more recently described CXCR-7 receptor, but effects of SDF-1/CXCR-7 signaling on stem cell migrating to ischemic brain injury area are little known. In the present study, we investigated the effect of CXCR-7 on bone marrow mesenchymal stem cell (BMSC) migration toward SDF-1α in the cerebral ischemia/reperfusion (I/R) rat hippocampus. We cultured BMSCs from rats and characterized them using flow cytometry, immunocytochemistry, western blotting, and immunofluorescence to detect SDF-1α, CXCR-4, and CXCR-7 expression in third passage BMSCs (P3-BMSCs). We also prepared the model of transient cerebral I/R by four-vessel occlusion (4-VO), and BMSCs were transplanted into I/R rat brain via lateral ventricle (LV) injection (20μl, 1×10(6)/ml). After that, we examined the effect of BMSCs migration in the cerebral I/R rat hippocampus through Transwell chamber assay. Our results show that SDF-1α, CXCR-4, and CXCR-7 were expressed in P3-BMSCs. Moreover, SDF-1α expression was increased in I/R hippocampus. At 48h after transplant, green fluorescent BrdU-BMSCs were observed in transplant groups, but no green fluorescent BrdU-BMSCs were seen in medium group. Among BMSCs transplant groups, the number of BrdU-BMSCs positive cell was the highest in BMSC group. Treatment with AMD3100 and/or CXCR-7 neutralizing antibody decreased the number of BMSC migration. Collectively, these findings indicate that CXCR-4 and -7 receptors were co-expressed in BMSCs and synergistically promoted BMSC migration. The effect of CXCR-7 was stronger than that of CXCR-4. Moreover, BMSCs that migrated to hippocampus promoted the autocrine and paracrine signaling of SDF-1α.
Collapse
Affiliation(s)
- Yulan Wang
- Department of Human Anatomy and Neurobiology, Xuzhou Medical College, No. 209 Tong-shan Road, Jiangsu, Xuzhou 221004, China.
| | - Wei Fu
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221004, China
| | - Shichun Zhang
- Department of Human Anatomy and Neurobiology, Xuzhou Medical College, No. 209 Tong-shan Road, Jiangsu, Xuzhou 221004, China
| | - Xiaomei He
- Department of Human Anatomy and Neurobiology, Xuzhou Medical College, No. 209 Tong-shan Road, Jiangsu, Xuzhou 221004, China
| | - Zhi'an Liu
- Department of Human Anatomy and Neurobiology, Xuzhou Medical College, No. 209 Tong-shan Road, Jiangsu, Xuzhou 221004, China
| | - Diansuai Gao
- Department of Human Anatomy and Neurobiology, Xuzhou Medical College, No. 209 Tong-shan Road, Jiangsu, Xuzhou 221004, China
| | - Tiejun Xu
- Department of Human Anatomy and Neurobiology, Xuzhou Medical College, No. 209 Tong-shan Road, Jiangsu, Xuzhou 221004, China
| |
Collapse
|
39
|
Tzekova N, Heinen A, Küry P. Molecules involved in the crosstalk between immune- and peripheral nerve Schwann cells. J Clin Immunol 2014; 34 Suppl 1:S86-104. [PMID: 24740512 DOI: 10.1007/s10875-014-0015-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022]
Abstract
Schwann cells are the myelinating glial cells of the peripheral nervous system and establish myelin sheaths on large caliber axons in order to accelerate their electrical signal propagation. Apart from this well described function, these cells revealed to exhibit a high degree of differentiation plasticity as they were shown to re- and dedifferentiate upon injury and disease as well as to actively participate in regenerative- and inflammatory processes. This review focuses on the crosstalk between glial- and immune cells observed in many peripheral nerve pathologies and summarizes functional evidences of molecules, regulators and factors involved in this process. We summarize data on Schwann cell's role presenting antigens, on interactions with the complement system, on Schwann cell surface molecules/receptors and on secreted factors involved in immune cell interactions or para-/autocrine signaling events, thus strengthening the view for a broader (patho) physiological role of this cell lineage.
Collapse
Affiliation(s)
- Nevena Tzekova
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, D-40225, Düsseldorf, Germany
| | | | | |
Collapse
|
40
|
Williams JL, Patel JR, Daniels BP, Klein RS. Targeting CXCR7/ACKR3 as a therapeutic strategy to promote remyelination in the adult central nervous system. ACTA ACUST UNITED AC 2014; 211:791-9. [PMID: 24733828 PMCID: PMC4010893 DOI: 10.1084/jem.20131224] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Current treatment modalities for the neurodegenerative disease multiple sclerosis (MS) use disease-modifying immunosuppressive compounds but do not promote repair. Although several potential targets that may induce myelin production have been identified, there has yet to be an approved therapy that promotes remyelination in the damaged central nervous system (CNS). Remyelination of damaged axons requires the generation of new oligodendrocytes from oligodendrocyte progenitor cells (OPCs). Although OPCs are detected in MS lesions, repair of myelin is limited, contributing to progressive clinical deterioration. In the CNS, the chemokine CXCL12 promotes remyelination via CXCR4 activation on OPCs, resulting in their differentiation into myelinating oligodendrocytes. Although the CXCL12 scavenging receptor CXCR7/ACKR3 (CXCR7) is also expressed by OPCs, its role in myelin repair in the adult CNS is unknown. We show that during cuprizone-induced demyelination, in vivo CXCR7 antagonism augmented OPC proliferation, leading to increased numbers of mature oligodendrocytes within demyelinated lesions. CXCR7-mediated effects on remyelination required CXCR4 activation, as assessed via both phospho-S339-CXCR4-specific antibodies and administration of CXCR4 antagonists. These findings identify a role for CXCR7 in OPC maturation during remyelination and are the first to use a small molecule to therapeutically enhance myelin repair in the demyelinated adult CNS.
Collapse
Affiliation(s)
- Jessica L Williams
- Department of Internal Medicine, 2 Department of Pathology and Immunology, and 3 Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO 63110
| | | | | | | |
Collapse
|
41
|
Boulanger JJ, Messier C. From precursors to myelinating oligodendrocytes: contribution of intrinsic and extrinsic factors to white matter plasticity in the adult brain. Neuroscience 2014; 269:343-66. [PMID: 24721734 DOI: 10.1016/j.neuroscience.2014.03.063] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/28/2014] [Accepted: 03/28/2014] [Indexed: 12/21/2022]
Abstract
Oligodendrocyte precursor cells (OPC) are glial cells that metamorphose into myelinating oligodendrocytes during embryogenesis and early stages of post-natal life. OPCs continue to divide throughout adulthood and some eventually differentiate into oligodendrocytes in response to demyelinating lesions. There is growing evidence that OPCs are also involved in activity-driven de novo myelination of previously unmyelinated axons and myelin remodeling in adulthood. In this review, we summarize the interwoven factors and cascades that promote the activation, recruitment and differentiation of OPCs into myelinating oligodendrocytes in the adult brain based mostly on results found in the study of demyelinating diseases. The goal of the review was to draw a complete picture of the transformation of OPCs into mature oligodendrocytes to facilitate the study of this transformation in both the normal and diseased adult brain.
Collapse
Affiliation(s)
| | - C Messier
- School of Psychology, University of Ottawa, Canada.
| |
Collapse
|
42
|
Abstract
Chemokines have fundamental roles in regulating immune and inflammatory responses, primarily through their control of leukocyte migration and localization. The biological functions of chemokines are typically mediated by signalling through G protein-coupled chemokine receptors, but chemokines are also bound by a small family of atypical chemokine receptors (ACKRs), the members of which are unified by their inability to initiate classical signalling pathways after ligand binding. These ACKRs are emerging as crucial regulatory components of chemokine networks in a wide range of developmental, physiological and pathological contexts. In this Review, we discuss the biochemical and immunological properties of ACKRs and the potential unifying themes in this family, and we highlight recent studies that identify novel roles for these molecules in development , homeostasis, inflammatory disease, infection and cancer.
Collapse
|
43
|
The peculiarities of the SDF-1/CXCL12 system: in some cells, CXCR4 and CXCR7 sing solos, in others, they sing duets. Cell Tissue Res 2013; 355:239-53. [DOI: 10.1007/s00441-013-1747-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 10/17/2013] [Indexed: 12/26/2022]
|
44
|
Mesenchymal stem cell conditioning promotes rat oligodendroglial cell maturation. PLoS One 2013; 8:e71814. [PMID: 23951248 PMCID: PMC3741203 DOI: 10.1371/journal.pone.0071814] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 07/02/2013] [Indexed: 12/15/2022] Open
Abstract
Oligodendroglial progenitor/precursor cells (OPCs) represent the main cellular source for the generation of new myelinating oligodendrocytes in the adult central nervous system (CNS). In demyelinating diseases such as multiple sclerosis (MS) myelin repair activities based on recruitment, activation and differentiation of resident OPCs can be observed. However, the overall degree of successful remyelination is limited and the existence of an MS-derived anti-oligodendrogenic milieu prevents OPCs from contributing to myelin repair. It is therefore of considerable interest to understand oligodendroglial homeostasis and maturation processes in order to enable the development of remyelination therapies. Mesenchymal stem cells (MSC) have been shown to exert positive immunomodulatory effects, reduce demyelination, increase neuroprotection and to promote adult neural stem cell differentiation towards the oligodendroglial lineage. We here addressed whether MSC secreted factors can boost the OPC’s oligodendrogenic capacity in a myelin non-permissive environment. To this end, we analyzed cellular morphologies, expression and regulation of key factors involved in oligodendroglial fate and maturation of primary rat cells upon incubation with MSC-conditioned medium. This demonstrated that MSC-derived soluble factors promote and accelerate oligodendroglial differentiation, even under astrocytic endorsing conditions. Accelerated maturation resulted in elevated levels of myelin expression, reduced glial fibrillary acidic protein expression and was accompanied by downregulation of prominent inhibitory differentiation factors such as Id2 and Id4. We thus conclude that apart from their suggested application as potential anti-inflammatory and immunomodulatory MS treatment, these cells might also be exploited to support endogenous myelin repair activities.
Collapse
|
45
|
Bracchi-Ricard V, Lambertsen KL, Ricard J, Nathanson L, Karmally S, Johnstone J, Ellman DG, Frydel B, McTigue DM, Bethea JR. Inhibition of astroglial NF-κB enhances oligodendrogenesis following spinal cord injury. J Neuroinflammation 2013; 10:92. [PMID: 23880092 PMCID: PMC3751509 DOI: 10.1186/1742-2094-10-92] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 07/12/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Astrocytes are taking the center stage in neurotrauma and neurological diseases as they appear to play a dominant role in the inflammatory processes associated with these conditions. Previously, we reported that inhibiting NF-κB activation in astrocytes, using a transgenic mouse model (GFAP-IκBα-dn mice), results in improved functional recovery, increased white matter preservation and axonal sparing following spinal cord injury (SCI). In the present study, we sought to determine whether this improvement, due to inhibiting NF-κB activation in astrocytes, could be the result of enhanced oligodendrogenesis in our transgenic mice. METHODS To assess oligodendrogenesis in GFAP-IκBα-dn compared to wild-type (WT) littermate mice following SCI, we used bromodeoxyuridine labeling along with cell-specific immuno-histochemistry, confocal microscopy and quantitative cell counts. To further gain insight into the underlying molecular mechanisms leading to increased white matter, we performed a microarray analysis in naïve and 3 days, 3 and 6 weeks following SCI in GFAP-IκBα-dn and WT littermate mice. RESULTS Inhibition of astroglial NF-κB in GFAP-IκBα-dn mice resulted in enhanced oligodendrogenesis 6 weeks following SCI and was associated with increased levels of myelin proteolipid protein compared to spinal cord injured WT mice. The microarray data showed a large number of differentially expressed genes involved in inflammatory and immune response between WT and transgenic mice. We did not find any difference in the number of microglia/leukocytes infiltrating the spinal cord but did find differences in their level of expression of toll-like receptor 4. We also found increased expression of the chemokine receptor CXCR4 on oligodendrocyte progenitor cells and mature oligodendrocytes in the transgenic mice. Finally TNF receptor 2 levels were significantly higher in the transgenic mice compared to WT following injury. CONCLUSIONS These studies suggest that one of the beneficial roles of blocking NF-κB in astrocytes is to promote oligodendrogenesis through alteration of the inflammatory environment.
Collapse
|
46
|
Mithal DS, Ren D, Miller RJ. CXCR4 signaling regulates radial glial morphology and cell fate during embryonic spinal cord development. Glia 2013; 61:1288-305. [PMID: 23828719 DOI: 10.1002/glia.22515] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 03/04/2013] [Accepted: 04/01/2013] [Indexed: 12/20/2022]
Abstract
Embryonic meninges secrete the chemokine SDF-1/CXCL12 as a chemotactic guide for migrating neural stem cells, but SDF-1 is not known to directly regulate the functions of radial glia. Recently, the developing meninges have been shown to regulate radial glial function, yet the mechanisms and signals responsible for this phenomenon remain unclear. Moreover, as a nonmigratory cell type, radial glia do not conform to traditional models associated with chemokine signaling in the central nervous system. Using fluorescent transgenes, in vivo genetic manipulations and pharmacological techniques, we demonstrate that SDF-1 derived from the meninges exerts a CXCR4-dependent effect on radial glia. Deletion of CXCR4 expression by radial glia influences their morphology, mitosis, and progression through both oligodendroglial and astroglial lineages. Additionally, disruption of CXCR4 signaling in radial glia has a transient effect on the migration of oligodendrocyte progenitors. These data indicate that a specific chemokine signal derived from the meninges has multiple regulatory effects on radial glia.
Collapse
Affiliation(s)
- Divakar S Mithal
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | | | | |
Collapse
|
47
|
Astrocyte TNFR2 is required for CXCL12-mediated regulation of oligodendrocyte progenitor proliferation and differentiation within the adult CNS. Acta Neuropathol 2012; 124:847-60. [PMID: 22933014 PMCID: PMC3508279 DOI: 10.1007/s00401-012-1034-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 06/28/2012] [Accepted: 08/08/2012] [Indexed: 12/28/2022]
Abstract
Multiple sclerosis (MS) is characterized by episodes of inflammatory demyelination with progressive failure of remyelination. Prior studies using murine models of MS indicate that remyelination within the adult central nervous system (CNS) requires the expression and activity of TNFR2 and CXCR4 by oligodendrocyte progenitor cells (OPCs), promoting their proliferation and differentiation into mature oligodendrocytes. Here, we extend these studies by examining the role of TNFR2 in the expression of the CXCR4 ligand, CXCL12, within the corpus callosum (CC) during cuprizone (CPZ) intoxication and by demonstrating that lentiviral-mediated gene delivery of CXCL12 to the demyelinated CC improves OPC proliferation and myelin expression during remyelination. Activated astrocytes and microglia express both TNFR1 and TNFR2 within the demyelinated CC. However, CPZ intoxicated TNFR2−/− mice exhibit loss of up-regulation of CXCL12 in astrocytes with concomitant decreases in numbers of CXCR4+ NG2+ OPCs within the CC. While CXCR4 antagonism does not affect OPC migration from subventricular zones into the CC, it decreases their proliferation and differentiation within the CC. Stereotactic delivery of lentivirus expressing CXCL12 protein into the CC of acutely demyelinated TNFR2−/− mice increases OPC proliferation and expression of myelin. In contrast, chronically demyelinated wild-type mice, which exhibit significant loss of astrocytes and OPCs, are unable to be rescued via CXCL12 lentivirus alone but instead required engraftment of CXCL12-expressing astrocytes for increased myelin expression. Our results show that TNFR2 activation induces CXCL12 expression in the demyelinated CC via autocrine signaling specifically within astrocytes, which promotes OPC proliferation and differentiation. In addition, gene delivery of critical pro-myelinating proteins might be a feasible approach for the treatment of remyelination failure in MS.
Collapse
|
48
|
Jaerve A, Schira J, Müller HW. Concise review: the potential of stromal cell-derived factor 1 and its receptors to promote stem cell functions in spinal cord repair. Stem Cells Transl Med 2012. [PMID: 23197665 DOI: 10.5966/sctm.2012-0068] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Transplanted stem cells provide beneficial effects on regeneration/recovery after spinal cord injury (SCI) by the release of growth-promoting factors, increased tissue preservation, and provision of a permissive environment for axon regeneration. A rise in chemokine stromal cell-derived factor 1 (SDF-1/CXCL12) expression levels in central nervous system (CNS) injury sites has been shown to play a central role in recruiting transplanted stem cells. Although technically more challenging, it has been shown that after SCI few endogenous stem cells are recruited via SDF-1/CXCR4 signaling. Evidence is accumulating that increasing SDF-1 levels at the injury site (e.g., by exogenous application or transfection methods) further enhances stem cell recruitment. Moreover, SDF-1 might, in addition to migration, also influence survival, proliferation, differentiation, and cytokine secretion of stem cells. Here, we discuss the experimental data available on the role of SDF-1 in stem and progenitor cell biology following CNS injury and suggest strategies for how manipulation of the SDF-1 system could facilitate stem cell-based therapeutic approaches in SCI. In addition, we discuss challenges such as how to circumvent off-target effects in order to facilitate the transfer of SDF-1 to the clinic.
Collapse
Affiliation(s)
- Anne Jaerve
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|
49
|
Expression and function of the SDF-1 chemokine receptors CXCR4 and CXCR7 during mouse limb muscle development and regeneration. Exp Cell Res 2012; 318:2178-90. [DOI: 10.1016/j.yexcr.2012.06.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 06/22/2012] [Accepted: 06/23/2012] [Indexed: 12/17/2022]
|
50
|
Torres KJ, Göttle P, Kremer D, Rivera JF, Aguirre-Cruz L, Corona T, Hartung HP, Küry P. Vinpocetine inhibits oligodendroglial precursor cell differentiation. Cell Physiol Biochem 2012; 30:711-22. [PMID: 22854710 DOI: 10.1159/000341451] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In multiple sclerosis during periods of remission a limited degree of myelin repair can be observed mediated by oligodendroglial precursor cells. Phosphodiesterase inhibitors act as anti-inflammatory agents and might hold promise for future multiple sclerosis treatment. AIMS To investigate whether phosphodiesterase inhibitors could also influence myelin repair. METHODS We stimulated primary oligodendroglial precursor cells with cilostazol, rolipram and vinpocetine and assessed their effects on repair related cellular processes. RESULTS We found that vinpocetine exerted a strong negative effect on myelin expression while cilostazol and rolipram did not show such effects. In addition, vinpocetine decreased morphological complexities suggesting an overall negative impact on oligodendroglial cell maturation. We provide evidence that this is not mediated via a blockade of phosphodiesterase-1 but rather by inhibition of IĸB kinase. CONCLUSION These findings suggest that vinpocetine via IĸB inhibition exerts a strong negative impact on oligodendroglial cell maturation and may therefore provide the rationale to restrict its application during periods of remission in multiple sclerosis patients. This is of particular interest since vinpocetine is widely used as a health supplement thought to act as a cognitive and memory enhancer for healthy people and patients with neurological or muscle diseases.
Collapse
Affiliation(s)
- Klintsy Julieta Torres
- Heinrich-Heine-University, Medical Faculty, Department of Neurology, Düsseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|