1
|
Ibrahim Fouad G, Ahmed KA. Remyelinating activities of Carvedilol or alpha lipoic acid in the Cuprizone-Induced rat model of demyelination. Int Immunopharmacol 2023; 118:110125. [PMID: 37028277 DOI: 10.1016/j.intimp.2023.110125] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
Multiple sclerosis (MS) is a complex and multifactorial neurodegenerative disease with unknown etiology, MS is featured by multifocal demyelinated lesions distributed throughout the brain. It is assumed to result from an interaction between genetic and environmental factors, including nutrition. Therefore, different therapeutic approaches are aiming to stimulate remyelination which could be defined as an endogenous regeneration and repair of myelin in the central nervous system. Carvedilol is an adrenergic receptor antagonist. Alpha lipoic acid (ALA) is a well-known antioxidant. Herein, we investigated the remyelination potential of Carvedilol or ALA post-Cuprizone (CPZ) intoxication. Carvedilol or ALA (20 mg/kg/d) was administrated orally for two weeks at the end of the five weeks of CPZ (0.6%) administration. CPZ provoked demyelination, enhanced oxidative stress, and stimulated neuroinflammation. Histological investigation of CPZ-induced brains showed obvious demyelination in the corpus callosum (CC). Both Carvedilol and ALA demonstrated remyelinating activities, with corresponding upregulation of the expression of MBP and PLP, the major myelin proteins, downregulation of the expression of TNF-α and MMP-9, and decrement of serum IFN-γ levels. Moreover, both Carvedilol and ALA alleviated oxidative stress, and ameliorated muscle fatigue. This study highlights the neurotherapeutic potential of Carvedilol or ALA in CPZ-induced demyelination, and offers a better model for the exploring of neuroregenerative strategies. The current study is the first to demonstrate a pro-remyelinating activity for Carvedilol, as compared to ALA, which might represent a potential additive benefit in halting demyelination and alleviating neurotoxicity. However, we could declare that Carvedilol showed a lower neuroprotective potential than ALA.
Collapse
|
2
|
Racheva K, Totev T, Natchev E, Bocheva N, Beirne R, Zlatkova M. Elimination of the color discrimination impairment along the blue-yellow axis in patients with hypothyroidism after treatment with levothyroxine as assessed by the Farnsworth-Munsell 100 hue test. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2023; 40:A26-A32. [PMID: 37132999 DOI: 10.1364/josaa.476139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Our previous study has shown that individuals with untreated hypothyroidism display significantly higher partial error scores (P E S) along the blue-yellow axis compared to the red-green axis than normal individuals using the Farnsworth-Munsell 100 hue test [J. Opt. Soc. Am. A37, A18 (2020)JOAOD60740-323210.1364/JOSAA.382390]. We wished to determine how color discrimination may change when hypothyroidism has been treated to the point of euthyroidism. Color discrimination was reassessed for 17 female individuals who had undergone treatment for hypothyroidism, and the results were compared with 22 female individuals without thyroid dysfunction. No statistically significant difference was found in the total error score (T E S) for the first and second measurements for both groups (p>0.45). The P E S for the hypothyroid group improved significantly in the previously impaired color regions after the treatment. Color discrimination defects found in untreated hypothyroidism can be negated with treatment of the condition over an appropriate time period.
Collapse
|
3
|
Drake SS, Zaman A, Simas T, Fournier AE. Comparing RNA-sequencing datasets from astrocytes, oligodendrocytes, and microglia in multiple sclerosis identifies novel dysregulated genes relevant to inflammation and myelination. WIREs Mech Dis 2023; 15:e1594. [PMID: 36600404 DOI: 10.1002/wsbm.1594] [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: 06/20/2022] [Revised: 09/25/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023]
Abstract
Central nervous system (CNS) inflammation is a key factor in multiple sclerosis (MS). Invasion of peripheral immune cells into the CNS resulting from an unknown signal or combination of signals results in activation of resident immune cells and the hallmark feature of the disease: demyelinating lesions. These lesion sites are an amalgam of reactive peripheral and central immune cells, astrocytes, damaged and dying oligodendrocytes, and injured neurons and axons. Sustained inflammation affects cells directly located within the lesion site and further abnormalities are apparent diffusely throughout normal-appearing white matter and grey matter. It is only relatively recently, using animal models, new tissue sampling techniques, and next-generation sequencing, that molecular changes occurring in CNS resident cells have been broadly captured. Advances in cell isolation through Fluorescence Activated Cell Sorting (FACS) and laser-capture microdissection together with the emergence of single-cell sequencing have enabled researchers to investigate changes in gene expression in astrocytes, microglia, and oligodendrocytes derived from animal models of MS as well as from primary patient tissue. The contribution of some dysregulated pathways has been followed up in individual studies; however, corroborating results often go unreported between sequencing studies. To this end, we have consolidated results from numerous RNA-sequencing studies to identify and review novel patterns of differentially regulated genes and pathways occurring within CNS glial cells in MS. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Sienna S Drake
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Aliyah Zaman
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Tristan Simas
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Alyson E Fournier
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Ibrahim Fouad G, Mabrouk M, El-Sayed SAM, Rizk MZ, Beherei HH. Neurotherapeutic efficacy of loaded sulforaphane on iron oxide nanoparticles against cuprizone-induced neurotoxicity: role of MMP-9 and S100β. Toxicol Mech Methods 2023:1-17. [PMID: 36775846 DOI: 10.1080/15376516.2023.2177219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Cuprizone (CUP) induces neurotoxicity and demyelination in animal models by provoking the activation of glial cells and the generation of reactive oxygen species (ROS). Sulforaphane (SF) is a phytochemical that exhibits a neuroprotective potential. In this study, we investigated the neurotherapeutic and pro-remyelinating activities of SF and SF-loaded within iron oxide nanoparticles (IONP-SF) in CUP-exposed rats. Magnetite iron oxide nanoparticles (IONPs) were prepared using the hydrothermal method that was further loaded with SF (IONP-SF). The loading of SF within the magnetite nanoparticles was assessed using FTIR, TEM, DLS, Zetasizer, and XPS. For the in vivo investigations, adult male Wistar rats (n = 40) were administrated either on a regular diet or a diet with CUP (0.2%) for 5 weeks. The rats were divided into four groups: negative control, CUP-induced, CUP + SF, and CUP + IONP-SF. CUP-exposed brains exhibited a marked elevation in lipid peroxidation, along with a significant decrease in the activities of glutathione peroxidase (GPx), and catalase (CAT). In addition, CUP intoxication downregulated the expression of myelin basic protein (MBP) and myelin proteolipid protein (PLP), upregulated the expression of Matrix metallopeptidase-9 (MMP-9) and S100β, and increased caspase-3 immunoexpression, these results were supported histopathologically in the cerebral cortexes. Treatment of CUP-rats with either SF or IONP-SF demonstrated remyelinating and neurotherapeutic activities. We could conclude that IONP-SF was more effective than free SF in mitigating the CUP-induced downregulation of MBP, upregulation of S100β, and caspase-3 immunoexpression.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Cairo, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt
| | - Sara A M El-Sayed
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt
| | - Maha Z Rizk
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Cairo, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
5
|
Al-Otaibi KM, Alghamdi BS, Al-Ghamdi MA, Mansouri RA, Ashraf GM, Omar UM. Therapeutic effect of combination vitamin D3 and siponimod on remyelination and modulate microglia activation in cuprizone mouse model of multiple sclerosis. Front Behav Neurosci 2023; 16:1068736. [PMID: 36688131 PMCID: PMC9849768 DOI: 10.3389/fnbeh.2022.1068736] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/29/2022] [Indexed: 01/07/2023] Open
Abstract
Stimulation of remyelination is critical for the treatment of multiple sclerosis (MS) to alleviate symptoms and protect the myelin sheath from further damage. The current study aimed to investigate the possible therapeutic effects of combining vitamin D3 (Vit D3) and siponimod (Sipo) on enhancing remyelination and modulating microglia phenotypes in the cuprizone (CPZ) demyelination mouse model. The study was divided into two stages; demyelination (first 5 weeks) and remyelination (last 4 weeks). In the first 5 weeks, 85 mice were randomly divided into two groups, control (n = 20, standard rodent chow) and CPZ (n = 65, 0.3% CPZ mixed with chow for 6 weeks, followed by 3 weeks of standard rodent chow). At week 5, the CPZ group was re-divided into four groups (n = 14) for remyelination stages; untreated CPZ (0.2 ml of CMC orally), CPZ+Vit D3 (800 IU/kg Vit D3 orally), CPZ+Sipo (1.5 mg/kg Sipo orally), and CPZ+Vit D3 (800 IU/kg Vit D3) + Sipo (1.5 mg/kg Sipo orally). Various behavioral tasks were performed to evaluate motor performance. Luxol Fast Blue (LFB) staining, the expression level of myelin basic protein (MBP), and M1/M2 microglia phenotype genes were assessed in the corpus callosum (CC). The results showed that the combination of Vit D3 and Sipo improved behavioral deficits, significantly promoted remyelination, and modulated expression levels of microglia phenotype genes in the CC at early and late remyelination stages. These results demonstrate for the first time that a combination of Vit D3 and Sipo can improve the remyelination process in the cuprizone (CPZ) mouse model by attenuating the M1 microglia phenotype. This may help to improve the treatment of MS patients.
Collapse
Affiliation(s)
- Kholoud M. Al-Otaibi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Chemistry, Faculty of Science, Albaha University, Albaha, Saudi Arabia,*Correspondence: Badrah S. Alghamdi Kholoud M. Al-Otaibi
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia,Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Badrah S. Alghamdi Kholoud M. Al-Otaibi
| | - Maryam A. Al-Ghamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia,Vitamin D Pharmacogenomics Research Group, King Abdulaziz University, Jeddah, Saudi Arabia,Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rasha A. Mansouri
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ulfat M. Omar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia,Princess Dr. Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
6
|
Soleimani A, Ezabadi SG, Möhn N, Esfandabadi ZM, Khosravizadeh Z, Skripuletz T, Azimzadeh M. Influence of hormones in multiple sclerosis: focus on the most important hormones. Metab Brain Dis 2023; 38:739-747. [PMID: 36595158 DOI: 10.1007/s11011-022-01138-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/30/2022] [Indexed: 01/04/2023]
Abstract
Hormonal imbalance may be an important factor in the severity of multiple sclerosis (MS) disease. In this context, hormone therapy has been shown to have immunoregulatory potential in various experimental approaches. There is increasing evidence of potentially beneficial effects of thyroid, melatonin, and sex hormones in MS models. These hormones may ameliorate the neurological impairment through immunoregulatory and neuroprotective effects, as well as by reducing oxidative stress. Expanding our knowledge of hormone therapy may be an effective step toward identifying additional molecular/cellular pathways in MS disease. In this review, we discuss the role of several important hormones in MS pathogenesis in terms of their effects on immunoregulatory aspects and neuroprotection.
Collapse
Affiliation(s)
- Alireza Soleimani
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sajjad Ghane Ezabadi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Zahra Khosravizadeh
- Clinical Research Development Unit, Amiralmomenin Hospital, Arak University of Medical Sciences, Arak, Iran
| | | | - Maryam Azimzadeh
- Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran.
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran.
| |
Collapse
|
7
|
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: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
8
|
Maglione AV, do Nascimento BPP, Ribeiro MO, de Souza TJL, da Silva REC, Sato MA, Penatti CAA, Britto LRG, de Souza JS, Maciel RMB, da Conceição RR, Laureano-Melo R, Giannocco G. Triiodothyronine Treatment reverses Depression-Like Behavior in a triple-transgenic animal model of Alzheimer's Disease. Metab Brain Dis 2022; 37:2735-2750. [PMID: 35951206 DOI: 10.1007/s11011-022-01055-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022]
Abstract
Alzheimer disease's (AD) is a neurodegenerative disorder characterized by cognitive and behavioral impairment. The central nervous system is an important target of thyroid hormones (TH). An inverse association between serum triiodothyronine (T3) levels and the risk of AD symptoms and progression has been reported. We investigated the effects of T3 treatment on the depression-like behavior in male transgenic 3xTg-AD mice. Animals were divided into 2 groups treated with daily intraperitoneal injections of 20 ng/g of body weight (b.w.) L-T3 (T3 group) or saline (vehicle, control group). The experimental protocol lasted 21 days, and behavioral tests were conducted on days 18-20. At the end of the experiment, the TH profile and hippocampal gene expression were evaluated. The T3-treated group significantly increased serum T3 and decreased thyroxine (T4) levels. When compared to control hippocampal samples, the T3 group exhibited attenuated glycogen synthase kinase-3 (GSK3), metalloproteinase 10 (ADAM10), amyloid-beta precursor-protein (APP), serotonin transporter (SERT), 5HT1A receptor, monocarboxylate transporter 8 (MCT8) and bone morphogenetic protein 7 (BMP-7) gene expression, whereas augmented superoxide dismutase 2 (SOD2) and Hairless gene expression. T3-treated animals also displayed reduced immobility time in both the tail suspension and forced swim tests, and in the latter presented a higher latency time compared to the control group. Therefore, our findings suggest that in an AD mouse model, T3 supplementation promotes improvements in depression-like behavior, through the modulation of the serotonergic related genes involved in the transmission mediated by 5HT1A receptors and serotonin reuptake, and attenuated disease progression.
Collapse
Affiliation(s)
- Andréa V Maglione
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Bruna P P do Nascimento
- Laboratory of Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, Brazil
| | - Miriam O Ribeiro
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, Brazil
| | - Talytha J L de Souza
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Renata E C da Silva
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Monica A Sato
- Dept. Morphology and Physiology, Faculdade de Medicina do ABC, Centro Universitário FMABC, Santo André- Brazil, São Paulo, Santo André, Brazil
| | - Carlos A A Penatti
- Laboratory of Human Physiology, Universidade Nove de Julho, São Paulo, Brazil
| | - Luiz R G Britto
- Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Janaina S de Souza
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Rui M B Maciel
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil
| | - Rodrigo Rodrigues da Conceição
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil.
| | - Roberto Laureano-Melo
- Laboratory of Physiopharmacoly and Behavior, Universidade de Barra Mansa, Rio de Janeiro, Brazil
| | - Gisele Giannocco
- Dept. Medicine, Laboratory of Endocrinology and Translational Medicine, Universidade Federal de São Paulo, UNIFESP/EPM, São Paulo, Brazil.
| |
Collapse
|
9
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
10
|
Moayedi K, Orandi S, Ebrahimi R, Tanhapour M, Moradi M, Abbastabar M, Golestani A. A novel approach to type 3 diabetes mechanism: The interplay between noncoding RNAs and insulin signaling pathway in Alzheimer's disease. J Cell Physiol 2022; 237:2838-2861. [PMID: 35580144 DOI: 10.1002/jcp.30779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 12/06/2022]
Abstract
Today, growing evidence indicates that patients with type 2 diabetes (T2D) are at a higher risk of developing Alzheimer's disease (AD). Indeed, AD as one of the main causes of dementia in people aged more than 65 years can be aggravated by insulin resistance (IR) and other metabolic risk factors related to T2D which are also linked to the function of the brain. Remarkably, a new term called "type 3 diabetes" has been suggested for those people who are diagnosed with AD while also showing the symptoms of IR and T2D. In this regard, the role of genetic and epigenetic changes associated with AD has been confirmed by many studies. On the other hand, it should be noted that the insulin signaling pathway is highly regulated by various mechanisms, including epigenetic factors. Among these, the role of noncoding RNAs (ncRNAs), including microRNAs and long noncoding RNAs has been comprehensively studied with respect to the pathology of AD and the most well-known underlying mechanisms. Nevertheless, the number of studies exploring the association between ncRNAs and the downstream targets of the insulin signaling pathway in the development of AD has notably increased in recent years. With this in view, the present study aimed to review the interplay between different ncRNAs and the insulin signaling pathway targets in the pathogenesis of AD to find a new approach in the field of combining biomarkers or therapeutic targets for this disease.
Collapse
Affiliation(s)
- Kiana Moayedi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Orandi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Tanhapour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Moradi
- Pediatric Infectious Diseases Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Abbastabar
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
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: 6.5] [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
|
12
|
Shiri E, Pasbakhsh P, Borhani-Haghighi M, Alizadeh Z, Nekoonam S, Mojaverrostami S, Pirhajati Mahabadi V, Mehdi A, Zibara K, Kashani IR. Mesenchymal Stem Cells Ameliorate Cuprizone-Induced Demyelination by Targeting Oxidative Stress and Mitochondrial Dysfunction. Cell Mol Neurobiol 2021; 41:1467-1481. [PMID: 32594382 DOI: 10.1007/s10571-020-00910-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. The main causes of MS disease progression, demyelination, and tissue damage are oxidative stress and mitochondrial dysfunction. Hence, the latter are considered as important therapeutic targets. Recent studies have demonstrated that mesenchymal stem cells (MSCs) possess antioxidative properties and are able to target mitochondrial dysfunction. Therefore, we investigated the effect of transplanting Wharton's jelly-derived MSCs in a demyelination mouse model of MS in which mice were fed cuprizone (CPZ) for 12 weeks. CPZ is a copper chelator that impairs the activity of cytochrome oxidase, decreases oxidative phosphorylation, and produces degenerative changes in oligodendrocytes, leading to toxic demyelination similar to those found in MS patients. Results showed that MSCs caused a significant increase in the percentage of myelinated areas and in the number of myelinated fibers in the corpus callosum of the CPZ + MSC group, compared to the CPZ group, as assessed by Luxol fast blue staining and transmission electron microscopy. In addition, transplantation of MSCs significantly increased the number of oligodendrocytes while decreasing astrogliosis and microgliosis in the corpus callosum of the CPZ + MSC group, evaluated by immunofluorescence. Moreover, the mechanism by which MSCs exert these physiological effects was found to be through abolishing the effect of CPZ on oxidative stress markers and mitochondrial dysfunction. Indeed, malondialdehyde significantly decreased while glutathione and superoxide dismutase significantly increased in CPZ + MSC mice group, in comparison witth the CPZ group alone. Furthermore, cell therapy with MSC transplantation increased the expression levels of mitochondrial biogenesis transcripts PGC1α, NRF1, MFN2, and TFAM. In summary, these results demonstrate that MSCs may attenuate MS by promoting an antioxidant response, reducing oxidative stress, and improving mitochondrial homeostasis.
Collapse
Affiliation(s)
- Elham Shiri
- Department of Anatomy, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Zohreh Alizadeh
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saied Nekoonam
- Department of Anatomy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Vahid Pirhajati Mahabadi
- Neuroscience Research Center, Vice-Chancellor for Research and Technology, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Mehdi
- PRASE and Faculty of Agriculture, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- ER045, PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| | | |
Collapse
|
13
|
Chaudhary P, Marracci GH, Calkins E, Pocius E, Bensen AL, Scanlan TS, Emery B, Bourdette DN. Thyroid hormone and thyromimetics inhibit myelin and axonal degeneration and oligodendrocyte loss in EAE. J Neuroimmunol 2021; 352:577468. [PMID: 33422763 PMCID: PMC8748188 DOI: 10.1016/j.jneuroim.2020.577468] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated that thyromimetics stimulate oligodendrocyte precursor cell differentiation and promote remyelination in murine demyelination models. We investigated whether a thyroid receptor-beta selective thyromimetic, sobetirome (Sob), and its CNS-targeted prodrug, Sob-AM2, could prevent myelin and axonal degeneration in experimental autoimmune encephalomyelitis (EAE). Compared to controls, EAE mice receiving triiodothyronine (T3, 0.4 mg/kg), Sob (5 mg/kg) or Sob-AM2 (5 mg/kg) had reduced clinical disease and, within the spinal cord, less tissue damage, more normally myelinated axons, fewer degenerating axons and more oligodendrocytes. T3 and Sob also protected cultured oligodendrocytes against cell death. Thyromimetics thus might protect against oligodendrocyte death, demyelination and axonal degeneration as well as stimulate remyelination in multiple sclerosis.
Collapse
Affiliation(s)
- P Chaudhary
- Department of Neurology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR 97239, United States of America.
| | - G H Marracci
- Department of Neurology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR 97239, United States of America
| | - E Calkins
- Department of Neurology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR 97239, United States of America
| | - E Pocius
- Department of Neurology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR 97239, United States of America
| | - A L Bensen
- Department of Neurology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America; Jungers Center for Neurosciences Research, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America
| | - T S Scanlan
- Department of Chemical Physiology & Biochemistry and Program in Chemical Biology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America
| | - B Emery
- Department of Neurology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America; Jungers Center for Neurosciences Research, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America
| | - D N Bourdette
- Department of Neurology, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States of America; VA Portland Health Care System, 3710 SW US Veterans Hospital Rd, Portland, OR 97239, United States of America
| |
Collapse
|
14
|
Su M, Soomro SH, Jie J, Fu H. Effects of the extracellular matrix on myelin development and regeneration in the central nervous system. Tissue Cell 2021; 69:101444. [PMID: 33450651 DOI: 10.1016/j.tice.2020.101444] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 12/25/2022]
Abstract
Extracellular matrix (ECM) is a collection of extracellular molecules secreted by cells, providing structural and biochemical support for surrounding tissues. The ECM exerts biological effects by interacting with growth factors, signal receptors or adhesion molecules. In the case of myelin formation and regeneration, the combination of ECM and its receptors (for example, integrins) modulates signaling pathways such as PI3K, MAPK, etc., which in turn induces complex biological effects throughout various stages of myelination and regeneration. Studies have also found that myelin injury would cause changes in ECM composition and thus affecting the myelin regeneration process. Research on the ECM will provide a better understanding of how myelin is formed and regenerated, which will help to develop new therapies for demyelinating diseases. Future progress in this field will provide important information on how to modify the ECM to promote proliferation and differentiation of oligodendrocyte precursor cells (OPC), thereby stimulating myelin formation and regeneration and restoring normal neural function.
Collapse
Affiliation(s)
- Min Su
- Wuhan University, School of Basic Medical Sciences, Wuhan, China.
| | | | - Jifu Jie
- Health School of Bayinguoleng Mongolian Autonomous Prefecture, Xinjiang, China.
| | - Hui Fu
- Wuhan University, School of Basic Medical Sciences, Wuhan, China.
| |
Collapse
|
15
|
Mojaverrostami S, Pasbakhsh P, Madadi S, Nekoonam S, Zarini D, Noori L, Shiri E, Salama M, Zibara K, Kashani IR. Calorie restriction promotes remyelination in a Cuprizone-Induced demyelination mouse model of multiple sclerosis. Metab Brain Dis 2020; 35:1211-1224. [PMID: 32638202 DOI: 10.1007/s11011-020-00597-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022]
Abstract
Over the past few decades several attempts have been made to introduce a potential and promising therapy for Multiple sclerosis (MS). Calorie restriction (CR) is a dietary manipulation to reduce calorie intake which has been shown to improve neuroprotection and attenuate neurodegenerative disorders. Here, we evaluated the effect of 33% CR regimen for 4 weeks on the remyelination capacity of Cuprizone (CPZ) induced demyelination in a mouse model of MS. Results showed that CR induced a significant increase in motor coordination and balance performance in CPZ mice. Also, luxol fast blue (LFB) staining showed that CR regimen significantly improved the remyelination in the corpus callosum of CPZ + CR mice compared to the CPZ group. In addition, CR regimen significantly increased the transcript expression levels of BDNF, Sox2, and Sirt1 in the corpus callosum of CPZ mice, while decreasing the p53 levels. Moreover, CR regimen significantly decreased the apoptosis rate. Furthermore, astrogliosis (GFAP + astrocytes) and microgliosis (Iba-1 + microglia) were significantly decreased by CR regimen while oligodendrogenesis (Olig2+) and Sirt1 + cell expression were significantly increased in the corpus callosum of CPZ + CR mice compared to the CPZ group. In conclusion, CR regimen can promote remyelination potential in a CPZ-demyelinating mouse model of MS by increasing oligodendrocyte generation while decreasing their apoptosis.
Collapse
Affiliation(s)
- Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Madadi
- Department of Anatomy, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Saeid Nekoonam
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Zarini
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Noori
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Shiri
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Salama
- Neuroscience Unit, Menoufia Medical School, Shebin El Kom, Egypt
| | - Kazem Zibara
- ER045, PRASE, DSST and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Gingele S, Stangel M. Emerging myelin repair agents in preclinical and early clinical development for the treatment of multiple sclerosis. Expert Opin Investig Drugs 2020; 29:583-594. [PMID: 32348161 DOI: 10.1080/13543784.2020.1762567] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Remyelination is a highly effective regenerative process that can restore axon function, prevent axonal loss, and reverse clinical deficits after demyelination. Hence, the promotion of remyelination is a logical goal in patients with multiple sclerosis (MS) in which remyelination is often insufficient. However, despite great progress regarding the development of immunomodulatory therapies for MS and an abundance of promising evidence from preclinical experiments so far, no therapy has convincingly demonstrated clinically significant remyelination properties. Therefore, enhancing myelin repair is an urgent and unmet need in MS. AREAS COVERED We searched clinicaltrials.gov and pubmed.ncbi.nlm.nih.gov and focused on therapeutic agents in development from the preclinical stage to clinical phase II. We selected agents for which data are available from in vitro experiments and at least one toxic demyelination animal model that reached at least phase I in clinical development in MS patients. EXPERT OPINION The evidence to promote remyelination is very promising for several agents, some of which possess anti-muscarinergic properties. Since remyelination is a complex process that involves various coordinated steps, a combination of different therapeutic approaches addressing different aspects of this regenerative mechanism may be reasonable. Furthermore, suitable surrogate markers of remyelination are necessary for proof-of-concept clinical trials.
Collapse
Affiliation(s)
- Stefan Gingele
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School , Hannover, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School , Hannover, Germany
| |
Collapse
|
17
|
Nellessen A, Nyamoya S, Zendedel A, Slowik A, Wruck C, Beyer C, Fragoulis A, Clarner T. Nrf2 deficiency increases oligodendrocyte loss, demyelination, neuroinflammation and axonal damage in an MS animal model. Metab Brain Dis 2020; 35:353-362. [PMID: 31529356 DOI: 10.1007/s11011-019-00488-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023]
Abstract
Oxidative stress is a pathophysiological hallmark of many CNS diseases, among multiple sclerosis (MS). Accordingly, boosting the astrocytic transcription factor nuclear factor E2-related factor 2 (Nrf2) system in an MS mouse model efficiently ameliorates oligodendrocyte loss, neuroinflammation and axonal damage. Moreover, Dimethylfumarate, an efficient activator of Nrf2, has recently been approved as therapeutic option in MS treatment. Here, we use the cuprizone mouse model of MS to induce oxidative stress, selective oligodendrocyte loss, microglia and astrocyte activation as well as axonal damage in both wild type and Nrf2-deficient mice. We found increased oligodendrocyte apoptosis and loss, pronounced neuroinflammation and higher levels of axonal damage in cuprizone-fed Nrf2-deficient animals when compared to wild type controls. In addition, Nrf2-deficient animals showed a higher susceptibility towards cuprizone within the commissura anterior white matter tract, a structure that is relatively insensitive to cuprizone in wild type animals. Our data highlight the cuprizone model as a suitable tool to study the complex interplay of oxidative stress, neuroinflammation and axonal damage. Further studies will have to show whether distinct expression patterns of Nrf2 are involved in the variable susceptibility towards cuprizone in the mouse.
Collapse
Affiliation(s)
- Anna Nellessen
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, Wendlingweg 2, 52074, Aachen, Germany
| | - Stella Nyamoya
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, Wendlingweg 2, 52074, Aachen, Germany
- Faculty of Medicine, LMU Munich, Department of Anatomy, Neuroanatomy, Pettenkoferstr. 11, 80336, Munich, Germany
- Rostock University Medical Center, Rostock, Institut für Anatomie, Gertrudenstr. 9, 18057, Rostock, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, Wendlingweg 2, 52074, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, Wendlingweg 2, 52074, Aachen, Germany
| | - Christoph Wruck
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, Wendlingweg 2, 52074, Aachen, Germany
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Tim Clarner
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
18
|
de Jong JM, Wang P, Oomkens M, Baron W. Remodeling of the interstitial extracellular matrix in white matter multiple sclerosis lesions: Implications for remyelination (failure). J Neurosci Res 2020; 98:1370-1397. [PMID: 31965607 DOI: 10.1002/jnr.24582] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/29/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) provides protection, rigidity, and structure toward cells. It consists, among others, of a wide variety of glycoproteins and proteoglycans, which act together to produce a complex and dynamic environment, most relevant in transmembrane events. In the brain, the ECM occupies a notable proportion of its volume and maintains the homeostasis of central nervous system (CNS). In addition, remodeling of the ECM, that is transient changes in ECM proteins regulated by matrix metalloproteinases (MMPs), is an important process that modulates cell behavior upon injury, thereby facilitating recovery. Failure of ECM remodeling plays an important role in the pathogenesis of multiple sclerosis (MS), a neurodegenerative demyelinating disease of the CNS with an inflammatory response against protective myelin sheaths that surround axons. Remyelination of denuded axons improves the neuropathological conditions of MS, but this regeneration process fails over time, leading to chronic disease progression. In this review, we uncover abnormal ECM remodeling in MS lesions by discussing ECM remodeling in experimental demyelination models, that is when remyelination is successful, and compare alterations in ECM components to the ECM composition and MMP expression in the parenchyma of demyelinated MS lesions, that is when remyelination fails. Inter- and intralesional differences in ECM remodeling in the distinct white matter MS lesions are discussed in terms of consequences for oligodendrocyte behavior and remyelination (failure). Hence, the review will aid to understand how abnormal ECM remodeling contributes to remyelination failure in MS lesions and assists in developing therapeutic strategies to promote remyelination.
Collapse
Affiliation(s)
- Jody M de Jong
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peng Wang
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michelle Oomkens
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
19
|
Hartley MD, Banerji T, Tagge IJ, Kirkemo LL, Chaudhary P, Calkins E, Galipeau D, Shokat MD, DeBell MJ, Van Leuven S, Miller H, Marracci G, Pocius E, Banerji T, Ferrara SJ, Meinig JM, Emery B, Bourdette D, Scanlan TS. Myelin repair stimulated by CNS-selective thyroid hormone action. JCI Insight 2019; 4:126329. [PMID: 30996143 PMCID: PMC6538346 DOI: 10.1172/jci.insight.126329] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/12/2019] [Indexed: 12/21/2022] Open
Abstract
Oligodendrocyte processes wrap axons to form neuroprotective myelin sheaths, and damage to myelin in disorders, such as multiple sclerosis (MS), leads to neurodegeneration and disability. There are currently no approved treatments for MS that stimulate myelin repair. During development, thyroid hormone (TH) promotes myelination through enhancing oligodendrocyte differentiation; however, TH itself is unsuitable as a remyelination therapy due to adverse systemic effects. This problem is overcome with selective TH agonists, sobetirome and a CNS-selective prodrug of sobetirome called Sob-AM2. We show here that TH and sobetirome stimulated remyelination in standard gliotoxin models of demyelination. We then utilized a genetic mouse model of demyelination and remyelination, in which we employed motor function tests, histology, and MRI to demonstrate that chronic treatment with sobetirome or Sob-AM2 leads to significant improvement in both clinical signs and remyelination. In contrast, chronic treatment with TH in this model inhibited the endogenous myelin repair and exacerbated disease. These results support the clinical investigation of selective CNS-penetrating TH agonists, but not TH, for myelin repair.
Collapse
Affiliation(s)
- Meredith D. Hartley
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| | - Tania Banerji
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | | | - Lisa L. Kirkemo
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| | - Priya Chaudhary
- VA Portland Health Care System, Portland, Oregon, USA
- Department of Neurology, and
| | - Evan Calkins
- VA Portland Health Care System, Portland, Oregon, USA
- Department of Neurology, and
| | - Danielle Galipeau
- VA Portland Health Care System, Portland, Oregon, USA
- Department of Neurology, and
| | - Mitra D. Shokat
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Margaret J. DeBell
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Shelby Van Leuven
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Hannah Miller
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Gail Marracci
- VA Portland Health Care System, Portland, Oregon, USA
- Department of Neurology, and
| | - Edvinas Pocius
- VA Portland Health Care System, Portland, Oregon, USA
- Department of Neurology, and
| | - Tapasree Banerji
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Skylar J. Ferrara
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - J. Matthew Meinig
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Ben Emery
- Department of Neurology, and
- Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, Oregon, USA
| | - Dennis Bourdette
- VA Portland Health Care System, Portland, Oregon, USA
- Department of Neurology, and
| | - Thomas S. Scanlan
- Department of Physiology & Pharmacology and Program in Chemical Biology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
20
|
Noorzehi G, Pasbakhsh P, Borhani-Haghighi M, Kashani IR, Madadi S, Tahmasebi F, Nekoonam S, Azizi M. Microglia polarization by methylprednizolone acetate accelerates cuprizone induced demyelination. J Mol Histol 2018; 49:471-479. [PMID: 30143908 DOI: 10.1007/s10735-018-9786-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/18/2018] [Indexed: 01/09/2023]
Abstract
Glucocorticoids (GC) are known as inflammatory drugs, which are used in neuroinflammatory diseases. Unlike the classic picture, recent studies have revealed that some GC drugs exacerbate inflammatory responses in their acute and prolonged administration. Multiple sclerosis (MS) is a demyelinating inflammatory disorder, in which reactive M1 microglia phenotype play a central role. Since methylprednisolone (MP), as a synthetic GC, are commonly used by MS patients, in this study, we evaluated the effect of long-term administration of MP on microglia polarization in cuprizone (CPZ)-induced MS model. The immunostaining results showed that chronic exposure to MP in the CPZ treated mice increased the number of Iba-1 positive microglia, which significantly expressed IP10 as M1 marker than arginase as M2 marker. MP treatment induced significant amplification in the transcript levels of iNOS and TNF-α (M1-related markers) in the corpus callosum of the MS mice, whereas no change detected in the expression of IL-10 (M2-related marker) between the groups. In addition, evaluation of myelin by luxol fast blue staining and transmission electron microscopy revealed that prolonged MP administration increased demyelination in comparison to the CPZ group. In conclusion, our results show that chronic MP therapy in the CPZ-induced demyelination model of MS polarized microglia to M1 pro-inflammatory phenotype.
Collapse
Affiliation(s)
- Golaleh Noorzehi
- School of Medicine, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina Street, 1417613151, Tehran, Iran
| | - Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina Street, 1417613151, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina Street, 1417613151, Tehran, Iran.
| | - Soheila Madadi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina Street, 1417613151, Tehran, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina Street, 1417613151, Tehran, Iran
| | - Saied Nekoonam
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina Street, 1417613151, Tehran, Iran
| | - Maryam Azizi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina Street, 1417613151, Tehran, Iran
| |
Collapse
|
21
|
Neuroprotective effect of linagliptin against cuprizone-induced demyelination and behavioural dysfunction in mice: A pivotal role of AMPK/SIRT1 and JAK2/STAT3/NF-κB signalling pathway modulation. Toxicol Appl Pharmacol 2018; 352:153-161. [DOI: 10.1016/j.taap.2018.05.035] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/11/2018] [Accepted: 05/29/2018] [Indexed: 12/14/2022]
|
22
|
De Angelis F, Plantone D, Chataway J. Pharmacotherapy in Secondary Progressive Multiple Sclerosis: An Overview. CNS Drugs 2018; 32:499-526. [PMID: 29968175 DOI: 10.1007/s40263-018-0538-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis is an immune-mediated inflammatory disease of the central nervous system characterised by demyelination, neuroaxonal loss and a heterogeneous clinical course. Multiple sclerosis presents with different phenotypes, most commonly a relapsing-remitting course and, less frequently, a progressive accumulation of disability from disease onset (primary progressive multiple sclerosis). The majority of people with relapsing-remitting multiple sclerosis, after a variable time, switch to a stage characterised by gradual neurological worsening known as secondary progressive multiple sclerosis. We have a limited understanding of the mechanisms underlying multiple sclerosis, and it is believed that multiple genetic, environmental and endogenous factors are elements driving inflammation and ultimately neurodegeneration. Axonal loss and grey matter damage have been regarded as amongst the leading causes of irreversible neurological disability in the progressive stages. There are over a dozen disease-modifying therapies currently licenced for relapsing-remitting multiple sclerosis, but none of these has provided evidence of effectiveness in secondary progressive multiple sclerosis. Recently, there has been some early modest success with siponimod in secondary progressive multiple sclerosis and ocrelizumab in primary progressive multiple sclerosis. Finding treatments to delay or prevent the courses of secondary progressive multiple sclerosis is an unmet and essential goal of the research in multiple sclerosis. In this review, we discuss new findings regarding drugs with immunomodulatory, neuroprotective or regenerative properties and possible treatment strategies for secondary progressive multiple sclerosis. We examine the field broadly to include trials where participants have progressive or relapsing phenotypes. We summarise the most relevant results from newer investigations from phase II and III randomised controlled trials over the past decade, with particular attention to the last 5 years.
Collapse
Affiliation(s)
- Floriana De Angelis
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK.
| | - Domenico Plantone
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| | - Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| |
Collapse
|
23
|
Narayan RN, Forsthuber T, Stüve O. Emerging drugs for primary progressive multiple sclerosis. Expert Opin Emerg Drugs 2018; 23:97-110. [DOI: 10.1080/14728214.2018.1463370] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ram Narendra Narayan
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Neurology Section, VA North Texas Health Care System, Dallas VA Medical Center, Dallas, TX, USA
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
24
|
Chami M, Halmer R, Schnoeder L, Anne Becker K, Meier C, Fassbender K, Gulbins E, Walter S. Acid sphingomyelinase deficiency enhances myelin repair after acute and chronic demyelination. PLoS One 2017; 12:e0178622. [PMID: 28582448 PMCID: PMC5459450 DOI: 10.1371/journal.pone.0178622] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/16/2017] [Indexed: 11/19/2022] Open
Abstract
The cuprizone animal model, also known as the toxic demyelination model, is a well-reproducible model of demyelination- and remyelination in mice, and has been useful in studying important aspect of human demyelinating diseases, including multiple sclerosis. In this study, we investigated the role of acid sphingomyelinase in demyelination and myelin repair by inducing acute and chronic demyelination with 5- or 12-week cuprizone treatment, followed by a 2-week cuprizone withdrawal phase to allow myelin repair. Sphingolipids, in particular ceramide and the enzyme acid sphingomyelinase, which generates ceramide from sphingomyelin, seem to be involved in astrocyte activation and neuronal damage in multiple sclerosis. We used immunohistochemistry to study glial reaction and oligodendrocyte distribution in acid sphingomyelinase deficient mice and wild-type C57BL/6J littermates at various time intervals after demyelination and remyelination. Axonal injury was quantified using amyloid precursor protein and synaptophysin, and gene expression and protein levels were measured using gene analysis and Western blotting, respectively. Our results show that mice lacking acid sphingomyelinase had a significant increase in myelin recovery and a significantly higher oligodendrocyte cell count after 2 weeks remyelination compared to wild-type littermates. Detrimental astroglial distribution was also significantly reduced in acid sphingomyelinase deficient animals. We obtained similar results in experiments using amitriptyline to inhibit acid sphingomyelinase. These findings suggest that acid sphingomyelinase plays a significant role in myelin repair, and its inhibition by amitriptyline may constitute a novel therapeutic approach for multiple sclerosis patients.
Collapse
Affiliation(s)
- Marwan Chami
- Department of Neurology, Saarland University Hospital, Homburg, Germany
- * E-mail:
| | - Ramona Halmer
- Department of Neurology, Saarland University Hospital, Homburg, Germany
| | - Laura Schnoeder
- Department of Neurology, Saarland University Hospital, Homburg, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carola Meier
- Institute of Anatomy and Cell Biology, Saarland University, Homburg, Germany
| | - Klaus Fassbender
- Department of Neurology, Saarland University Hospital, Homburg, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Silke Walter
- Department of Neurology, Saarland University Hospital, Homburg, Germany
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
25
|
Role of Matricellular Proteins in Disorders of the Central Nervous System. Neurochem Res 2016; 42:858-875. [DOI: 10.1007/s11064-016-2088-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022]
|