1
|
Gluck L, Gerstein B, Kaunzner UW. Repair mechanisms of the central nervous system: From axon sprouting to remyelination. Neurotherapeutics 2025:e00583. [PMID: 40348704 DOI: 10.1016/j.neurot.2025.e00583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
The central nervous system (CNS), comprising the brain, spinal cord, and optic nerve, has limited regenerative capacity, posing significant challenges in treating neurological disorders. Recent advances in neuroscience and neurotherapeutics have introduced promising strategies to stimulate CNS repair, particularly in the context of neurodegenerative diseases such as multiple sclerosis. This review explores the complex interplay between inflammation, demyelination, and remyelination possibilities. Glial cells, including oligodendrocyte precursors, oligodendrocytes, astrocytes and microglia play dual roles in injury response, with reactive gliosis promoting repair but also potentially inhibiting recovery through glial scar formation. There is also an emphasis on axonal regeneration, axonal sprouting and stem cell therapies. We highlight the role of neuroplasticity in recovery post-injury and the limited regenerative potential of axons in the CNS due to inhibitory factors such as myelin-associated inhibitors. Moreover, neurotrophic factors support neuronal survival and axonal growth, while stem cell-based approaches offer promise for replacing lost neurons and glial cells. However, challenges such as stem cell survival, integration, and risk of tumor formation remain. Furthermore, we examine the role of neurogenesis in CNS repair and the remodeling of the extracellular matrix, which can facilitate regeneration. Through these diverse mechanisms, ongoing research aims to overcome the intrinsic and extrinsic barriers to CNS repair and advance therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lauren Gluck
- Montefiore Medical Center, 1250 Waters Place Tower 2, Bronx, NY 10461, USA.
| | - Brittany Gerstein
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| | - Ulrike W Kaunzner
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| |
Collapse
|
2
|
Arab Firozjae A, Shiran MR, Ajami A, Farzin D, Rashidi M. Lutein improves remyelination by reducing of neuroinflammation in C57BL/6 mouse models of multiple sclerosis. Heliyon 2024; 10:e39253. [PMID: 39640747 PMCID: PMC11620235 DOI: 10.1016/j.heliyon.2024.e39253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 12/07/2024] Open
Abstract
Multiple sclerosis (MS) is an inflammatory neurodegenerative disorder characterized by demyelination. Lutein, a xanthophyll carotenoid, has well-known antioxidant and anti-inflammatory properties. In this experiment, we aimed to investigate the neuroprotective and remyelination potential of lutein in comparison with dimethyl fumarate (DMF) as a reference drug in post-cuprizone-intoxicated C57BL/6 mice. Lutein (50, 100, and 200 mg/kg/day; p.o.) and DMF (15 mg/kg/day, i.p.) were administered either alone or in combination for three weeks at the end of the six-week cuprizone (0.2 % w/w) feeding period. At the end of the study, behavioral tests, histopathological staining, immunohistochemistry (olig2), ELISA, and real-time PCR were performed to evaluate the target parameters. Lutein treatment significantly enhanced motor functions, reversed cuprizone-induced demyelination and increased serum TAC. In addition, treatment with lutein increased the number of Olig2+ cells in the corpus callosum, reduced the IL-1β and TNF-α and increased BDNF. Lutein administration significantly increased the expression levels of genes involved in myelin production (MBP, PLP, MOG, MAG, and OLIG-1) and notably reduced GFAP expression levels. In the present study, our results showed that lutein treatment could promote remyelination and neuroprotective effects by reducing neuroinflammation and upregulating the expression of the genes involved in myelin formation These findings suggest that lutein could serve as a potential adjuvant therapy for patients with multiple sclerosis. Further clinical trials are necessary to confirm its efficacy.
Collapse
Affiliation(s)
- Atefeh Arab Firozjae
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Mohammad Reza Shiran
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Abolghasem Ajami
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Davood Farzin
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Mohsen Rashidi
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
3
|
Vinnenberg L, Rychlik N, Oniani T, Williams B, White JA, Kovac S, Meuth SG, Budde T, Hundehege P. Assessing neuroprotective effects of diroximel fumarate and siponimod via modulation of pacemaker channels in an experimental model of remyelination. Exp Neurol 2024; 371:114572. [PMID: 37852467 DOI: 10.1016/j.expneurol.2023.114572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/04/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Cuprizone (CPZ)-induced alterations in axonal myelination are associated with a period of neuronal hyperexcitability and increased activity of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels in the thalamocortical (TC) system. Substances used for the treatment of multiple sclerosis (MS) have been shown to normalize neuronal excitability in CPZ-treated mice. Therefore, we aimed to examine the effects of diroximel fumarate (DRF) and the sphingosine 1-phospate receptor (S1PR) modulator siponimod on action potential firing and the inward current (Ih) carried by HCN ion channels in naive conditions and during different stages of de- and remyelination. Here, DRF application reduced Ih current density in ex vivo patch clamp recordings from TC neurons of the ventrobasal thalamic complex (VB), thereby counteracting the increase of Ih during early remyelination. Siponimod reduced Ih in VB neurons under control conditions but had no effect in neurons of the auditory cortex (AU). Furthermore, siponimod increased and decreased AP firing properties of neurons in VB and AU, respectively. Computational modeling revealed that both DRF and siponimod influenced thalamic bursting during early remyelination by delaying the onset and decreasing the interburst frequency. Thus, substances used in MS treatment normalize excitability in the TC system by influencing AP firing and Ih.
Collapse
Affiliation(s)
- Laura Vinnenberg
- Department of Neurology with Institute of Translational Neurology, Münster University, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Nicole Rychlik
- Institute of Physiology I, Münster University, Robert-Koch-Str. 27a, D-48149 Münster, Germany.
| | - Tengiz Oniani
- Institute of Physiology I, Münster University, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Brandon Williams
- Department of Biomedical Engineering, Center for Systems Neuroscience, Neurophotonics Center, Boston University, 610 Commonwealth Ave, Boston MA-02215, USA
| | - John A White
- Department of Biomedical Engineering, Center for Systems Neuroscience, Neurophotonics Center, Boston University, 610 Commonwealth Ave, Boston MA-02215, USA
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, Münster University, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Sven G Meuth
- Neurology Clinic, Medical Faculty, University Clinic Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Thomas Budde
- Institute of Physiology I, Münster University, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Münster University, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| |
Collapse
|
4
|
Hoeferlin GF, Bajwa T, Olivares H, Zhang J, Druschel LN, Sturgill BS, Sobota M, Boucher P, Duncan J, Hernandez-Reynoso AG, Cogan SF, Pancrazio JJ, Capadona JR. Antioxidant Dimethyl Fumarate Temporarily but Not Chronically Improves Intracortical Microelectrode Performance. MICROMACHINES 2023; 14:1902. [PMID: 37893339 PMCID: PMC10609067 DOI: 10.3390/mi14101902] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/24/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023]
Abstract
Intracortical microelectrode arrays (MEAs) can be used in a range of applications, from basic neuroscience research to providing an intimate interface with the brain as part of a brain-computer interface (BCI) system aimed at restoring function for people living with neurological disorders or injuries. Unfortunately, MEAs tend to fail prematurely, leading to a loss in functionality for many applications. An important contributing factor in MEA failure is oxidative stress resulting from chronically inflammatory-activated microglia and macrophages releasing reactive oxygen species (ROS) around the implant site. Antioxidants offer a means for mitigating oxidative stress and improving tissue health and MEA performance. Here, we investigate using the clinically available antioxidant dimethyl fumarate (DMF) to reduce the neuroinflammatory response and improve MEA performance in a rat MEA model. Daily treatment of DMF for 16 weeks resulted in a significant improvement in the recording capabilities of MEA devices during the sub-chronic (Weeks 5-11) phase (42% active electrode yield vs. 35% for control). However, these sub-chronic improvements were lost in the chronic implantation phase, as a more exacerbated neuroinflammatory response occurs in DMF-treated animals by 16 weeks post-implantation. Yet, neuroinflammation was indiscriminate between treatment and control groups during the sub-chronic phase. Although worse for chronic use, a temporary improvement (<12 weeks) in MEA performance is meaningful. Providing short-term improvement to MEA devices using DMF can allow for improved use for limited-duration studies. Further efforts should be taken to explore the mechanism behind a worsened neuroinflammatory response at the 16-week time point for DMF-treated animals and assess its usefulness for specific applications.
Collapse
Affiliation(s)
- George F. Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Tejas Bajwa
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Hannah Olivares
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Jichu Zhang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Lindsey N. Druschel
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Brandon S. Sturgill
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Michael Sobota
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Pierce Boucher
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Jonathan Duncan
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Ana G. Hernandez-Reynoso
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Stuart F. Cogan
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Joseph J. Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| |
Collapse
|
5
|
Gola L, Bierhansl L, Csatári J, Schroeter CB, Korn L, Narayanan V, Cerina M, Abdolahi S, Speicher A, Hermann AM, König S, Dinkova-Kostova AT, Shekh-Ahmad T, Meuth SG, Wiendl H, Gorji A, Pawlowski M, Kovac S. NOX4-derived ROS are neuroprotective by balancing intracellular calcium stores. Cell Mol Life Sci 2023; 80:127. [PMID: 37081190 PMCID: PMC10119225 DOI: 10.1007/s00018-023-04758-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/22/2023]
Abstract
Hyperexcitability is associated with neuronal dysfunction, cellular death, and consequently neurodegeneration. Redox disbalance can contribute to hyperexcitation and increased reactive oxygen species (ROS) levels are observed in various neurological diseases. NOX4 is an NADPH oxidase known to produce ROS and might have a regulating function during oxidative stress. We, therefore, aimed to determine the role of NOX4 on neuronal firing, hyperexcitability, and hyperexcitability-induced changes in neural network function. Using a multidimensional approach of an in vivo model of hyperexcitability, proteomic analysis, and cellular function analysis of ROS, mitochondrial integrity, and calcium levels, we demonstrate that NOX4 is neuroprotective by regulating ROS and calcium homeostasis and thereby preventing hyperexcitability and consequently neuronal death. These results implicate NOX4 as a potential redox regulator that is beneficial in hyperexcitability and thereby might have an important role in neurodegeneration.
Collapse
Affiliation(s)
- Lukas Gola
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Laura Bierhansl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Júlia Csatári
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Lisanne Korn
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Venu Narayanan
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Sara Abdolahi
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Anna Speicher
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Alexander M Hermann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Simone König
- Core Unit Proteomics, Interdisciplinary Center for Clinical Research, Medical Faculty, University of Münster, 48149, Münster, Germany
| | | | - Tawfeeq Shekh-Ahmad
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany
| | - Matthias Pawlowski
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany.
| |
Collapse
|
6
|
Leo H, Kipp M. Remyelination in Multiple Sclerosis: Findings in the Cuprizone Model. Int J Mol Sci 2022; 23:ijms232416093. [PMID: 36555733 PMCID: PMC9783537 DOI: 10.3390/ijms232416093] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Remyelination therapies, which are currently under development, have a great potential to delay, prevent or even reverse disability in multiple sclerosis patients. Several models are available to study the effectiveness of novel compounds in vivo, among which is the cuprizone model. This model is characterized by toxin-induced demyelination, followed by endogenous remyelination after cessation of the intoxication. Due to its high reproducibility and ease of use, this model enjoys high popularity among various research and industrial groups. In this review article, we will summarize recent findings using this model and discuss the potential of some of the identified compounds to promote remyelination in multiple sclerosis patients.
Collapse
Affiliation(s)
| | - Markus Kipp
- Correspondence: ; Tel.: +49-(0)-381-494-8400
| |
Collapse
|
7
|
Omeprazole Suppresses Oxaliplatin-Induced Peripheral Neuropathy in a Rodent Model and Clinical Database. Int J Mol Sci 2022; 23:ijms23168859. [PMID: 36012136 PMCID: PMC9408309 DOI: 10.3390/ijms23168859] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/11/2022] Open
Abstract
(1) Background: Oxaliplatin is used as first-line chemotherapy not only for colorectal cancer but also for gastric and pancreatic cancers. However, it induces peripheral neuropathy with high frequency as an adverse event, and there is no effective preventive or therapeutic method. (2) Methods: The effects of omeprazole, a proton pump inhibitor (PPI), on oxaliplatin-induced peripheral neuropathy (OIPN) was investigated using an in vivo model and a real-world database. (3) Results: In a rat model, oxaliplatin (4 mg/kg, i.p., twice a week for 4 weeks) caused mechanical hypersensitivity accompanied by sciatic nerve axonal degeneration and myelin sheath disorder. Repeated injection of omeprazole (5−20 mg/kg, i.p., five times per week for 4 weeks) ameliorated these behavioral and pathological abnormalities. Moreover, omeprazole did not affect the tumor growth inhibition of oxaliplatin in tumor bearing mice. Furthermore, clinical database analysis of the Food and Drug Administration Adverse Event Reporting System (FAERS) suggests that the group using omeprazole has a lower reporting rate of peripheral neuropathy of oxaliplatin-treated patients than the group not using (3.06% vs. 6.48%, p < 0.001, reporting odds ratio 0.44, 95% confidence interval 0.32−0.61). (4) Conclusions: These results show the preventing effect of omeprazole on OIPN.
Collapse
|
8
|
Oniani T, Vinnenberg L, Chaudhary R, Schreiber JA, Riske K, Williams B, Pape HC, White JA, Junker A, Seebohm G, Meuth SG, Hundehege P, Budde T, Zobeiri M. Effects of Axonal Demyelination, Inflammatory Cytokines and Divalent Cation Chelators on Thalamic HCN Channels and Oscillatory Bursting. Int J Mol Sci 2022; 23:ijms23116285. [PMID: 35682964 PMCID: PMC9181513 DOI: 10.3390/ijms23116285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system that is characterized by the progressive loss of oligodendrocytes and myelin and is associated with thalamic dysfunction. Cuprizone (CPZ)-induced general demyelination in rodents is a valuable model for studying different aspects of MS pathology. CPZ feeding is associated with the altered distribution and expression of different ion channels along neuronal somata and axons. However, it is largely unknown whether the copper chelator CPZ directly influences ion channels. Therefore, we assessed the effects of different divalent cations (copper; zinc) and trace metal chelators (EDTA; Tricine; the water-soluble derivative of CPZ, BiMPi) on hyperpolarization-activated cyclic nucleotide-gated (HCN) channels that are major mediators of thalamic function and pathology. In addition, alterations of HCN channels induced by CPZ treatment and MS-related proinflammatory cytokines (IL-1β; IL-6; INF-α; INF-β) were characterized in C57Bl/6J mice. Thus, the hyperpolarization-activated inward current (Ih) was recorded in thalamocortical (TC) neurons and heterologous expression systems (mHCN2 expressing HEK cells; hHCN4 expressing oocytes). A number of electrophysiological characteristics of Ih (potential of half-maximal activation (V0.5); current density; activation kinetics) were unchanged following the extracellular application of trace metals and divalent cation chelators to native neurons, cell cultures or oocytes. Mice were fed a diet containing 0.2% CPZ for 35 days, resulting in general demyelination in the brain. Withdrawal of CPZ from the diet resulted in rapid remyelination, the effects of which were assessed at three time points after stopping CPZ feeding (Day1, Day7, Day25). In TC neurons, Ih was decreased on Day1 and Day25 and revealed a transient increased availability on Day7. In addition, we challenged naive TC neurons with INF-α and IL-1β. It was found that Ih parameters were differentially altered by the application of the two cytokines to thalamic cells, while IL-1β increased the availability of HCN channels (depolarized V0.5; increased current density) and the excitability of TC neurons (depolarized resting membrane potential (RMP); increased the number of action potentials (APs); produced a larger voltage sag; promoted higher input resistance; increased the number of burst spikes; hyperpolarized the AP threshold), INF-α mediated contrary effects. The effect of cytokine modulation on thalamic bursting was further assessed in horizontal slices and a computational model of slow thalamic oscillations. Here, IL-1β and INF-α increased and reduced oscillatory bursting, respectively. We conclude that HCN channels are not directly modulated by trace metals and divalent cation chelators but are subject to modulation by different MS-related cytokines.
Collapse
Affiliation(s)
- Tengiz Oniani
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - Laura Vinnenberg
- Department of Neurology with Institute of Translational Neurology, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany; (L.V.); (P.H.)
| | - Rahul Chaudhary
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - Julian A. Schreiber
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität, Corren-Str. 48, D-48149 Münster, Germany;
- Cellular Electrophysiology and Molecular Biology, Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany;
| | - Kathrin Riske
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität, Waldeyer-Str. 15, D-48149 Münster, Germany; (K.R.); (A.J.)
| | - Brandon Williams
- Center for Systems Neuroscience, Neurophotonics Center, Department of Biomedical Engineering, Boston University, 610 Commonwealth Ave., Boston, MA 02215, USA; (B.W.); (J.A.W.)
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - John A. White
- Center for Systems Neuroscience, Neurophotonics Center, Department of Biomedical Engineering, Boston University, 610 Commonwealth Ave., Boston, MA 02215, USA; (B.W.); (J.A.W.)
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität, Waldeyer-Str. 15, D-48149 Münster, Germany; (K.R.); (A.J.)
| | - Guiscard Seebohm
- Cellular Electrophysiology and Molecular Biology, Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany;
| | - Sven G. Meuth
- Neurology Clinic, University Clinic Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany;
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany; (L.V.); (P.H.)
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
- Correspondence:
| | - Mehrnoush Zobeiri
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| |
Collapse
|
9
|
Chaudhary R, Albrecht S, Datunashvili M, Cerina M, Lüttjohann A, Han Y, Narayanan V, Chetkovich DM, Ruck T, Kuhlmann T, Pape HC, Meuth SG, Zobeiri M, Budde T. Modulation of Pacemaker Channel Function in a Model of Thalamocortical Hyperexcitability by Demyelination and Cytokines. Cereb Cortex 2022; 32:4397-4421. [PMID: 35076711 PMCID: PMC9574242 DOI: 10.1093/cercor/bhab491] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 12/02/2022] Open
Abstract
A consensus is yet to be reached regarding the exact prevalence of epileptic seizures or epilepsy in multiple sclerosis (MS). In addition, the underlying pathophysiological basis of the reciprocal interaction among neuroinflammation, demyelination, and epilepsy remains unclear. Therefore, a better understanding of cellular and network mechanisms linking these pathologies is needed. Cuprizone-induced general demyelination in rodents is a valuable model for studying MS pathologies. Here, we studied the relationship among epileptic activity, loss of myelin, and pro-inflammatory cytokines by inducing acute, generalized demyelination in a genetic mouse model of human absence epilepsy, C3H/HeJ mice. Both cellular and network mechanisms were studied using in vivo and in vitro electrophysiological techniques. We found that acute, generalized demyelination in C3H/HeJ mice resulted in a lower number of spike–wave discharges, increased cortical theta oscillations, and reduction of slow rhythmic intrathalamic burst activity. In addition, generalized demyelination resulted in a significant reduction in the amplitude of the hyperpolarization-activated inward current (Ih) in thalamic relay cells, which was accompanied by lower surface expression of hyperpolarization-activated, cyclic nucleotide-gated channels, and the phosphorylated form of TRIP8b (pS237-TRIP8b). We suggest that demyelination-related changes in thalamic Ih may be one of the factors defining the prevalence of seizures in MS.
Collapse
Affiliation(s)
- Rahul Chaudhary
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149 Münster, Germany
| | - Stefanie Albrecht
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
| | - Maia Datunashvili
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149 Münster, Germany
| | - Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, 48149 Münster, Germany
| | - Annika Lüttjohann
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149 Münster, Germany
| | - Ye Han
- Vanderbilt University Medical Center, Department of Neurology, Nashville, TN 37232, USA
| | - Venu Narayanan
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, 48149 Münster, Germany
| | - Dane M Chetkovich
- Vanderbilt University Medical Center, Department of Neurology, Nashville, TN 37232, USA
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, 48149 Münster, Germany
| | - Hans-Christian Pape
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149 Münster, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, Westfälische Wilhelms-Universität, 48149 Münster, Germany
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Mehrnoush Zobeiri
- Address correspondence to Dr Thomas Budde, Wilhelms-Universität, Institut für Physiologie I, Robert-Koch-Str. 27a, D-48149 Münster, Germany. ; Dr Mehrnoush Zobeiri, Wilhelms-Universität, Institut für Physiologie I, Robert-Koch-Str. 27a, D-48149 Münster, Germany.
| | - Thomas Budde
- Address correspondence to Dr Thomas Budde, Wilhelms-Universität, Institut für Physiologie I, Robert-Koch-Str. 27a, D-48149 Münster, Germany. ; Dr Mehrnoush Zobeiri, Wilhelms-Universität, Institut für Physiologie I, Robert-Koch-Str. 27a, D-48149 Münster, Germany.
| |
Collapse
|
10
|
Allanach JR, Farrell JW, Mésidor M, Karimi-Abdolrezaee S. Current status of neuroprotective and neuroregenerative strategies in multiple sclerosis: A systematic review. Mult Scler 2022; 28:29-48. [PMID: 33870797 PMCID: PMC8688986 DOI: 10.1177/13524585211008760] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/13/2021] [Accepted: 03/21/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Immune-mediated demyelination and consequent degeneration of oligodendrocytes and axons are hallmark features of multiple sclerosis (MS). Remyelination declines in progressive MS, causing permanent axonal loss and irreversible disabilities. Strategies aimed at enhancing remyelination are critical to attenuate disease progression. OBJECTIVE We systematically reviewed recent advances in neuroprotective and regenerative therapies for MS, covering preclinical and clinical studies. METHODS We searched three biomedical databases using defined keywords. Two authors independently reviewed articles for inclusion based on pre-specified criteria. The data were extracted from each study and assessed for risk of bias. RESULTS Our search identified 7351 studies from 2014 to 2020, of which 221 met the defined criteria. These studies reported 262 interventions, wherein 92% were evaluated in animal models. These interventions comprised protein, RNA, lipid and cellular biologics, small molecules, inorganic compounds, and dietary and physiological interventions. Small molecules were the most highly represented strategy, followed by antibody therapies and stem cell transplantation. CONCLUSION While significant strides have been made to develop regenerative treatments for MS, the current evidence illustrates a skewed representation of the types of strategies that advance to clinical trials. Further examination is thus required to address current barriers to implementing experimental treatments in clinical settings.
Collapse
Affiliation(s)
- Jessica R Allanach
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - John W. Farrell
- Department of Health and Human Performance, Texas State University, San Marcos, TX, USA
| | - Miceline Mésidor
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada/Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada/Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
11
|
Groppa S, Gonzalez-Escamilla G, Eshaghi A, Meuth SG, Ciccarelli O. Linking immune-mediated damage to neurodegeneration in multiple sclerosis: could network-based MRI help? Brain Commun 2021; 3:fcab237. [PMID: 34729480 PMCID: PMC8557667 DOI: 10.1093/braincomms/fcab237] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2021] [Indexed: 01/04/2023] Open
Abstract
Inflammatory demyelination characterizes the initial stages of multiple sclerosis, while progressive axonal and neuronal loss are coexisting and significantly contribute to the long-term physical and cognitive impairment. There is an unmet need for a conceptual shift from a dualistic view of multiple sclerosis pathology, involving either inflammatory demyelination or neurodegeneration, to integrative dynamic models of brain reorganization, where, glia-neuron interactions, synaptic alterations and grey matter pathology are longitudinally envisaged at the whole-brain level. Functional and structural MRI can delineate network hallmarks for relapses, remissions or disease progression, which can be linked to the pathophysiology behind inflammatory attacks, repair and neurodegeneration. Here, we aim to unify recent findings of grey matter circuits dynamics in multiple sclerosis within the framework of molecular and pathophysiological hallmarks combined with disease-related network reorganization, while highlighting advances from animal models (in vivo and ex vivo) and human clinical data (imaging and histological). We propose that MRI-based brain networks characterization is essential for better delineating ongoing pathology and elaboration of particular mechanisms that may serve for accurate modelling and prediction of disease courses throughout disease stages.
Collapse
Affiliation(s)
- Sergiu Groppa
- Imaging and Neurostimulation, Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Gabriel Gonzalez-Escamilla
- Imaging and Neurostimulation, Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55131, Germany
| | - Arman Eshaghi
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London WC1E 6BT, UK.,Department of Computer Science, Centre for Medical Image Computing (CMIC), University College London, London WC1E 6BT, UK
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf 40225, Germany
| | - Olga Ciccarelli
- Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London WC1E 6BT, UK
| |
Collapse
|
12
|
Ye N, Cruz J, Peng X, Ma J, Zhang A, Cheng X. Remyelination is enhanced by Astragalus polysaccharides through inducing the differentiation of oligodendrocytes from neural stem cells in cuprizone model of demyelination. Brain Res 2021; 1763:147459. [PMID: 33794147 DOI: 10.1016/j.brainres.2021.147459] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 01/03/2023]
Abstract
Demyelination is the hallmark of multiple sclerosis (MS). Promoting remyelination is an important strategy to treat MS. Our previous study showed that Astragalus polysaccharides (APS), the main bioactive component of Astragalus membranaceus, could prevent demyelination in experimental autoimmune encephalomyelitis mice. To investigate the effects of APS on remyelination and the underlying mechanisms, in this study we set up a cuprizone-induced demyelination model in mice and treated them with APS. It was found that APS relieved the neurobehavioral dysfunctions caused by demyelination, and efficaciously facilitated remyelination in vivo. In order to determine whether the mechanism of enhancing remyelination was associated with the differentiation of neural stem cells (NSCs), biomarkers of NSCs, astrocytes, oligodendrocytes and neurons were measured in the corpus callosum tissues of mice through Real-time PCR, Western blot and immunohistochemistry assays. Data revealed that APS suppressed the stemness of NSCs, reduced the differentiation of NSCs into astrocytes, and promoted the differentiation into oligodendrocytes and neurons. This phenomenon was confirmed in the differentiation model of C17.2 NSCs cultured in vitro. Since Sonic hedgehog signaling pathway has been proven to be crucial to the differentiation of NSCs into oligodendrocytes, we examined expression levels of the key molecules in this pathway in vivo and in vitro, and eventually found APS activated this signaling pathway. Together, our results demonstrated that APS probably activated Sonic hedgehog signaling pathway first, then induced NSCs to differentiate into oligodendrocytes and promoted remyelination, which suggested that APS might be a potential candidate in treating MS.
Collapse
Affiliation(s)
- Ni Ye
- Institute of Clinical Immunology, Yue-Yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, PR China
| | - Jennifer Cruz
- Institute of Clinical Immunology, Yue-Yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, PR China; Doctoral Program of Acupuncture & Oriental Medicine, The Atlantic Institute of Oriental Medicine, FL 33301, USA
| | - Xiaoyan Peng
- Institute of Clinical Immunology, Yue-Yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, PR China
| | - Jinyun Ma
- Institute of Clinical Immunology, Yue-Yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, PR China
| | - Aiming Zhang
- Department of Neurology, Min-Hang Hospital of Integrative Medicine, Shanghai 200241, PR China
| | - Xiaodong Cheng
- Institute of Clinical Immunology, Yue-Yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, PR China.
| |
Collapse
|
13
|
Kourakis S, Timpani CA, de Haan JB, Gueven N, Fischer D, Rybalka E. Dimethyl Fumarate and Its Esters: A Drug with Broad Clinical Utility? Pharmaceuticals (Basel) 2020; 13:ph13100306. [PMID: 33066228 PMCID: PMC7602023 DOI: 10.3390/ph13100306] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Fumaric acid esters (FAEs) are small molecules with anti-oxidative, anti-inflammatory and immune-modulating effects. Dimethyl fumarate (DMF) is the best characterised FAE and is approved and registered for the treatment of psoriasis and Relapsing-Remitting Multiple Sclerosis (RRMS). Psoriasis and RRMS share an immune-mediated aetiology, driven by severe inflammation and oxidative stress. DMF, as well as monomethyl fumarate and diroximel fumarate, are commonly prescribed first-line agents with favourable safety and efficacy profiles. The potential benefits of FAEs against other diseases that appear pathogenically different but share the pathologies of oxidative stress and inflammation are currently investigated.
Collapse
Affiliation(s)
- Stephanie Kourakis
- College of Health and Biomedicine, Victoria University, Melbourne, VIC 8001, Australia;
| | - Cara A. Timpani
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, Victoria University, St Albans, VIC 3021, Australia
| | - Judy B. de Haan
- Oxidative Stress Laboratory, Baker Heart and Diabetes Institute, Basic Science Domain, Melbourne, VIC 3004, Australia;
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3083, Australia
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7005, Australia;
| | - Dirk Fischer
- Division of Developmental- and Neuropediatrics, University Children’s Hospital Basel, University of Basel, 4056 Basel, Switzerland;
| | - Emma Rybalka
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, Victoria University, St Albans, VIC 3021, Australia
- Correspondence: ; Tel.: +61-383-958-226
| |
Collapse
|
14
|
Carvalho NZM, Chiarotto GB, Bernardes D, Kempe PRG, Oliveira ALR. Neuroprotection by dimethyl fumarate following ventral root crush in C57BL/6J mice. Brain Res Bull 2020; 164:184-197. [PMID: 32866558 DOI: 10.1016/j.brainresbull.2020.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/22/2020] [Accepted: 08/13/2020] [Indexed: 01/22/2023]
Abstract
CNS lesions usually result in permanent loss of function and are an important problem in the medical field. In order to investigate neuroprotection/degeneration mechanisms and the synaptic plasticity of motoneurons, in addition to the potential for a variety of treatments, different experimental models of axonal injury have been proposed. Recent studies have tested the immunomodulatory drug dimethyl fumarate (DMF) for the treatment of neurodegenerative diseases and have shown promising outcomes. Therefore, in this work, we investigated the effects of DMF with regard to neuroprotection and its influence on the glial response in C57BL/6J animals subjected to crushing of the motor roots in the lumbar intumescence of the spinal cord. The animals were divided into a vehicle-treated injury group (0.08 % methylcellulose solution control group, n = 7) and injured groups treated with DMF at different doses (15, 30, 45, 90 and 180 mg/kg; n = 6-7 per dose). The 90 mg/kg dose showed the best neuroprotective results, so it was used for treatment over a period of eight weeks. Neuronal survival was assessed through Nissl staining, and functional recovery was evaluated with the CatWalk system (walking track test) and the von Frey test (mechanoreception). Immunohistochemistry was used to assess synaptic coverage and astroglial and microglial reactivity using the primary antibodies anti-synaptophysin (pre-synaptic terminal pan marker), GAD65 (GABAergic pre-synaptic terminations - inhibitory), and VGLUT1 (glutamatergic pre-synaptic terminations - excitatory). Glial reactions were evaluated with anti-IBA1 (microglia) and GFAP (astrocytes). Gene transcript levels of IL-3, IL-4, TNF-α, IL-6, TGF-β, iNOS-M1, and arginase-M2 were quantified by RT-qPCR. The results indicated that treatment with DMF, at a dose of 90 mg/kg, promoted neuroprotection and immunomodulation towards an anti-inflammatory response. It also resulted in greater preservation of inhibitory synapses and reduced astroglial reactivity, providing a more favorable environment for sensorimotor recovery.
Collapse
Affiliation(s)
| | - Gabriela Bortolança Chiarotto
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil; University Center of Herminio Ometto Foundation, Post Graduate Program in Biomedical Science, Brazil.
| | - Danielle Bernardes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil; University Center of Herminio Ometto Foundation, Post Graduate Program in Biomedical Science, Brazil.
| | - Paula Regina Gelinski Kempe
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil.
| | - Alexandre Leite Rodrigues Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil.
| |
Collapse
|
15
|
Cerina M, Muthuraman M, Gallus M, Koirala N, Dik A, Wachsmuth L, Hundehege P, Schiffler P, Tenberge JG, Fleischer V, Gonzalez-Escamilla G, Narayanan V, Krämer J, Faber C, Budde T, Groppa S, Meuth SG. Myelination- and immune-mediated MR-based brain network correlates. J Neuroinflammation 2020; 17:186. [PMID: 32532336 PMCID: PMC7293122 DOI: 10.1186/s12974-020-01827-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/24/2020] [Indexed: 11/23/2022] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS), characterized by inflammatory and neurodegenerative processes. Despite demyelination being a hallmark of the disease, how it relates to neurodegeneration has still not been completely unraveled, and research is still ongoing into how these processes can be tracked non-invasively. Magnetic resonance imaging (MRI) derived brain network characteristics, which closely mirror disease processes and relate to functional impairment, recently became important variables for characterizing immune-mediated neurodegeneration; however, their histopathological basis remains unclear. Methods In order to determine the MRI-derived correlates of myelin dynamics and to test if brain network characteristics derived from diffusion tensor imaging reflect microstructural tissue reorganization, we took advantage of the cuprizone model of general demyelination in mice and performed longitudinal histological and imaging analyses with behavioral tests. By introducing cuprizone into the diet, we induced targeted and consistent demyelination of oligodendrocytes, over a period of 5 weeks. Subsequent myelin synthesis was enabled by reintroduction of normal food. Results Using specific immune-histological markers, we demonstrated that 2 weeks of cuprizone diet induced a 52% reduction of myelin content in the corpus callosum (CC) and a 35% reduction in the neocortex. An extended cuprizone diet increased myelin loss in the CC, while remyelination commenced in the neocortex. These histologically determined dynamics were reflected by MRI measurements from diffusion tensor imaging. Demyelination was associated with decreased fractional anisotropy (FA) values and increased modularity and clustering at the network level. MRI-derived modularization of the brain network and FA reduction in key anatomical regions, including the hippocampus, thalamus, and analyzed cortical areas, were closely related to impaired memory function and anxiety-like behavior. Conclusion Network-specific remyelination, shown by histology and MRI metrics, determined amelioration of functional performance and neuropsychiatric symptoms. Taken together, we illustrate the histological basis for the MRI-driven network responses to demyelination, where increased modularity leads to evolving damage and abnormal behavior in MS. Quantitative information about in vivo myelination processes is mirrored by diffusion-based imaging of microstructural integrity and network characteristics.
Collapse
Affiliation(s)
- Manuela Cerina
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Muthuraman Muthuraman
- Movement Disorders, Imaging and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Marco Gallus
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Nabin Koirala
- Movement Disorders, Imaging and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andre Dik
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Lydia Wachsmuth
- Departement of Radiology, University of Münster, Münster, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Patrick Schiffler
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Jan-Gerd Tenberge
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Vinzenz Fleischer
- Movement Disorders, Imaging and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Gabriel Gonzalez-Escamilla
- Movement Disorders, Imaging and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Venu Narayanan
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Julia Krämer
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| | - Cornelius Faber
- Departement of Radiology, University of Münster, Münster, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Münster, Germany
| | - Sergiu Groppa
- Movement Disorders, Imaging and Neurostimulation, Biomedical Statistics and Multimodal Signal Processing Unit, Department of Neurology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, Münster University Hospital, Münster, Germany
| |
Collapse
|
16
|
Evoked potentials as a translatable biomarker to track functional remyelination. Mol Cell Neurosci 2019; 99:103393. [DOI: 10.1016/j.mcn.2019.103393] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/18/2019] [Accepted: 07/25/2019] [Indexed: 11/21/2022] Open
|