1
|
Piel S, McManus MJ, Heye KN, Beaulieu F, Fazelinia H, Janowska JI, MacTurk B, Starr J, Gaudio H, Patel N, Hefti MM, Smalley ME, Hook JN, Kohli NV, Bruton J, Hallowell T, Delso N, Roberts A, Lin Y, Ehinger JK, Karlsson M, Berg RA, Morgan RW, Kilbaugh TJ. Effect of dimethyl fumarate on mitochondrial metabolism in a pediatric porcine model of asphyxia-induced in-hospital cardiac arrest. Sci Rep 2024; 14:13852. [PMID: 38879681 PMCID: PMC11180202 DOI: 10.1038/s41598-024-64317-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/07/2024] [Indexed: 06/19/2024] Open
Abstract
Neurological and cardiac injuries are significant contributors to morbidity and mortality following pediatric in-hospital cardiac arrest (IHCA). Preservation of mitochondrial function may be critical for reducing these injuries. Dimethyl fumarate (DMF) has shown potential to enhance mitochondrial content and reduce oxidative damage. To investigate the efficacy of DMF in mitigating mitochondrial injury in a pediatric porcine model of IHCA, toddler-aged piglets were subjected to asphyxia-induced CA, followed by ventricular fibrillation, high-quality cardiopulmonary resuscitation, and random assignment to receive either DMF (30 mg/kg) or placebo for four days. Sham animals underwent similar anesthesia protocols without CA. After four days, tissues were analyzed for mitochondrial markers. In the brain, untreated CA animals exhibited a reduced expression of proteins of the oxidative phosphorylation system (CI, CIV, CV) and decreased mitochondrial respiration (p < 0.001). Despite alterations in mitochondrial content and morphology in the myocardium, as assessed per transmission electron microscopy, mitochondrial function was unchanged. DMF treatment counteracted 25% of the proteomic changes induced by CA in the brain, and preserved mitochondrial structure in the myocardium. DMF demonstrates a potential therapeutic benefit in preserving mitochondrial integrity following asphyxia-induced IHCA. Further investigation is warranted to fully elucidate DMF's protective mechanisms and optimize its therapeutic application in post-arrest care.
Collapse
Affiliation(s)
- Sarah Piel
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA.
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty of the Heinrich-Heine-University, Düsseldorf, Germany.
| | - Meagan J McManus
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Kristina N Heye
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Forrest Beaulieu
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Hossein Fazelinia
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Joanna I Janowska
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Bryce MacTurk
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Jonathan Starr
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Hunter Gaudio
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Nisha Patel
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Marco M Hefti
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Martin E Smalley
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jordan N Hook
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Neha V Kohli
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - James Bruton
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Thomas Hallowell
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Nile Delso
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Anna Roberts
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Yuxi Lin
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Johannes K Ehinger
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Otorhinolaryngology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Otorhinolaryngology, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden
| | | | - Robert A Berg
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Ryan W Morgan
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Todd J Kilbaugh
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| |
Collapse
|
2
|
Rubio T, Campos-Rodríguez Á, Sanz P. Beneficial Effect of Fingolimod in a Lafora Disease Mouse Model by Preventing Reactive Astrogliosis-Derived Neuroinflammation and Brain Infiltration of T-lymphocytes. Mol Neurobiol 2024; 61:3105-3120. [PMID: 37971656 PMCID: PMC11087365 DOI: 10.1007/s12035-023-03766-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Lafora disease (LD; OMIM#254780) is a rare, devastating, and fatal form of progressive myoclonus epilepsy that affects young adolescents and has no treatment yet. One of the hallmarks of the disease is the accumulation of aberrant poorly branched forms of glycogen (polyglucosans, PGs) in the brain and peripheral tissues. The current hypothesis is that this accumulation is causative of the pathophysiology of the disease. Another hallmark of LD is the presence of neuroinflammation. We have recently reported the presence of reactive glia-derived neuroinflammation in LD mouse models and defined the main inflammatory pathways that operate in these mice, mainly TNF and IL-6 signaling pathways. In addition, we described the presence of infiltration of peripheral immune cells in the brain parenchyma, which could cooperate and aggravate the neuroinflammatory landscape of LD. In this work, we have checked the beneficial effect of two compounds with the capacity to ameliorate neuroinflammation and reduce leukocyte infiltration into the brain, namely fingolimod and dimethyl fumarate. Our results indicate a beneficial effect of fingolimod in reducing reactive astrogliosis-derived neuroinflammation and T-lymphocyte infiltration, which correlated with the improved behavioral performance of the treated Epm2b-/- mice. On the contrary, dimethyl fumarate, although it was able to reduce reactive astrogliosis, was less effective in preventing neuroinflammation and T-lymphocyte infiltration and in modifying behavioral tests.
Collapse
Affiliation(s)
- Teresa Rubio
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Jaime Roig 11, 46010, Valencia, Spain
| | - Ángela Campos-Rodríguez
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Jaime Roig 11, 46010, Valencia, Spain
| | - Pascual Sanz
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Jaime Roig 11, 46010, Valencia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010, Valencia, Spain.
| |
Collapse
|
3
|
Lima ADR, Ferrari BB, Pradella F, Carvalho RM, Rivero SLS, Quintiliano RPS, Souza MA, Brunetti NS, Marques AM, Santos IP, Farias AS, Oliveira EC, Santos LMB. Dimethyl fumarate modulates the regulatory T cell response in the mesenteric lymph nodes of mice with experimental autoimmune encephalomyelitis. Front Immunol 2024; 15:1391949. [PMID: 38765015 PMCID: PMC11099268 DOI: 10.3389/fimmu.2024.1391949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024] Open
Abstract
Dimethyl fumarate (DMF, Tecfidera) is an oral drug utilized to treat relapsing-remitting multiple sclerosis (MS). DMF treatment reduces disease activity in MS. Gastrointestinal discomfort is a common adverse effect of the treatment with DMF. This study aimed to investigate the effect of DMF administration in the gut draining lymph nodes cells of C57BL6/J female mice with experimental autoimmune encephalomyelitis (EAE), an animal model of MS. We have demonstrated that the treatment with DMF (7.5 mg/kg) significantly reduces the severity of EAE. This reduction of the severity is accompanied by the increase of both proinflammatory and anti-inflammatory mechanisms at the beginning of the treatment. As the treatment progressed, we observed an increasing number of regulatory Foxp3 negative CD4 T cells (Tr1), and anti-inflammatory cytokines such as IL-27, as well as the reduction of PGE2 level in the mesenteric lymph nodes of mice with EAE. We provide evidence that DMF induces a gradual anti-inflammatory response in the gut draining lymph nodes, which might contribute to the reduction of both intestinal discomfort and the inflammatory response of EAE. These findings indicate that the gut is the first microenvironment of action of DMF, which may contribute to its effects of reducing disease severity in MS patients.
Collapse
Affiliation(s)
- Amanda D. R. Lima
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Breno B. Ferrari
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Fernando Pradella
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Rodrigo M. Carvalho
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Sandra L. S. Rivero
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Raphael P. S. Quintiliano
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Matheus A. Souza
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Natália S. Brunetti
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Ana M. Marques
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Irene P. Santos
- Departamento de Citometria do Centro de Hematologia e Hemoterapia da UNICAMP, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Alessandro S. Farias
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Elaine C. Oliveira
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Technology Faculty of Sorocaba- Paula Souza State Center of Technological Education, Sorocaba, Brazil
| | - Leonilda M. B. Santos
- Unidade de Neuroimunologia, Dept.Genética, Evolução, Microbiologia e Imunologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, (INCT-NIM), National Council for Scientific and Technological Development (CNPq), Brasilia, Brazil
| |
Collapse
|
4
|
Sanz P, Rubio T, Garcia-Gimeno MA. Neuroinflammation and Epilepsy: From Pathophysiology to Therapies Based on Repurposing Drugs. Int J Mol Sci 2024; 25:4161. [PMID: 38673747 PMCID: PMC11049926 DOI: 10.3390/ijms25084161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Neuroinflammation and epilepsy are different pathologies, but, in some cases, they are so closely related that the activation of one of the pathologies leads to the development of the other. In this work, we discuss the three main cell types involved in neuroinflammation, namely (i) reactive astrocytes, (ii) activated microglia, and infiltration of (iii) peripheral immune cells in the central nervous system. Then, we discuss how neuroinflammation and epilepsy are interconnected and describe the use of different repurposing drugs with anti-inflammatory properties that have been shown to have a beneficial effect in different epilepsy models. This review reinforces the idea that compounds designed to alleviate seizures need to target not only the neuroinflammation caused by reactive astrocytes and microglia but also the interaction of these cells with infiltrated peripheral immune cells.
Collapse
Affiliation(s)
- Pascual Sanz
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Jaime Roig 11, 46010 Valencia, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Teresa Rubio
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Jaime Roig 11, 46010 Valencia, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
- Faculty of Health Science, Universidad Europea de Valencia, 46010 Valencia, Spain
| | - Maria Adelaida Garcia-Gimeno
- Department of Biotechnology, Escuela Técnica Superior de Ingeniería Agronómica y del Medio Natural, Universitat Politécnica de València, 46022 Valencia, Spain;
| |
Collapse
|
5
|
Nicola MA, Attaai AH, Abdel-Raheem MH, Mohammed AF, Abu-Elhassan YF. Neuroprotective effects of rutin against cuprizone-induced multiple sclerosis in mice. Inflammopharmacology 2024; 32:1295-1315. [PMID: 38512652 PMCID: PMC11006763 DOI: 10.1007/s10787-024-01442-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 01/24/2024] [Indexed: 03/23/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease of the central nervous system that injures the myelin sheath, provoking progressive axonal degeneration and functional impairments. No efficient therapy is available at present to combat such insults, and hence, novel safe and effective alternatives for MS therapy are extremely required. Rutin (RUT) is a flavonoid that exhibits antioxidant, anti-inflammatory, and neuroprotective effects in several brain injuries. The present study evaluated the potential beneficial effects of two doses of RUT in a model of pattern-III lesion of MS, in comparison to the conventional standard drug; dimethyl fumarate (DMF). Demyelination was induced in in male adult C57BL/6 mice by dietary 0.2% (w/w) cuprizone (CPZ) feeding for 6 consecutive weeks. Treated groups received either oral RUT (50 or 100 mg/kg) or DMF (15 mg/kg), along with CPZ feeding, for 6 consecutive weeks. Mice were then tested for behavioral changes, followed by biochemical analyses and histological examinations of the corpus callosum (CC). Results revealed that CPZ caused motor dysfunction, demyelination, and glial activation in demyelinated lesions, as well as significant oxidative stress, and proinflammatory cytokine elevation. Six weeks of RUT treatment significantly improved locomotor activity and motor coordination. Moreover, RUT considerably improved remyelination in the CC of CPZ + RUT-treated mice, as revealed by luxol fast blue staining and transmission electron microscopy. Rutin also significantly attenuated CPZ-induced oxidative stress and inflammation in the CC of tested animals. The effect of RUT100 was obviously more marked than either that of DMF, regarding most of the tested parameters, or even its smaller tested dose. In silico docking revealed that RUT binds tightly within NF-κB at the binding site of the protein-DNA complex, with a good negative score of -6.79 kcal/mol. Also, RUT-Kelch-like ECH-associated protein 1 (Keap1) model clarifies the possible inhibition of Keap1-Nrf2 protein-protein interaction. Findings of the current study provide evidence for the protective effect of RUT in CPZ-induced demyelination and behavioral dysfunction in mice, possibly by modulating NF-κB and Nrf2 signaling pathways. The present study may be one of the first to indicate a pro-remyelinating effect for RUT, which might represent a potential additive benefit in treating MS.
Collapse
Affiliation(s)
- Mariam A Nicola
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Asyût, 71526, Egypt.
| | - Abdelraheim H Attaai
- Department of Anatomy and Histology, School of Veterinary Medicine, Badr University in Assiut, New Nasser City, West of Assiut, Asyût, Egypt
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Asyût, 71526, Egypt
| | | | - Anber F Mohammed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Asyût, 71526, Egypt
| | - Yasmin F Abu-Elhassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Asyût, 71526, Egypt
| |
Collapse
|
6
|
Kreiter D, Postma AA, Hupperts R, Gerlach O. Hallmarks of spinal cord pathology in multiple sclerosis. J Neurol Sci 2024; 456:122846. [PMID: 38142540 DOI: 10.1016/j.jns.2023.122846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
A disparity exists between spinal cord and brain involvement in multiple sclerosis (MS), each independently contributing to disability. Underlying differences between brain and cord are not just anatomical in nature (volume, white/grey matter organization, vascularization), but also in barrier functions (differences in function and composition of the blood-spinal cord barrier compared to blood-brain barrier) and possibly in repair mechanisms. Also, immunological phenotypes seem to influence localization of inflammatory activity. Whereas the brain has gained a lot of attention in MS research, the spinal cord lags behind. Advanced imaging techniques and biomarkers are improving and providing us with tools to uncover the mechanisms of spinal cord pathology in MS. In the present review, we elaborate on the underlying anatomical and physiological factors driving differences between brain and cord involvement in MS and review current literature on pathophysiology of spinal cord involvement in MS and the observed differences to brain involvement.
Collapse
Affiliation(s)
- Daniel Kreiter
- Academic MS Center Zuyd, Department of Neurology, Zuyderland MC, Sittard-Geleen, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands.
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Raymond Hupperts
- Academic MS Center Zuyd, Department of Neurology, Zuyderland MC, Sittard-Geleen, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Oliver Gerlach
- Academic MS Center Zuyd, Department of Neurology, Zuyderland MC, Sittard-Geleen, the Netherlands; School for Mental Health and Neuroscience, Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
7
|
Bresciani G, Manai F, Davinelli S, Tucci P, Saso L, Amadio M. Novel potential pharmacological applications of dimethyl fumarate-an overview and update. Front Pharmacol 2023; 14:1264842. [PMID: 37745068 PMCID: PMC10512734 DOI: 10.3389/fphar.2023.1264842] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Dimethyl fumarate (DMF) is an FDA-approved drug for the treatment of psoriasis and multiple sclerosis. DMF is known to stabilize the transcription factor Nrf2, which in turn induces the expression of antioxidant response element genes. It has also been shown that DMF influences autophagy and participates in the transcriptional control of inflammatory factors by inhibiting NF-κB and its downstream targets. DMF is receiving increasing attention for its potential to be repurposed for several diseases. This versatile molecule is indeed able to exert beneficial effects on different medical conditions through a pleiotropic mechanism, in virtue of its antioxidant, immunomodulatory, neuroprotective, anti-inflammatory, and anti-proliferative effects. A growing number of preclinical and clinical studies show that DMF may have important therapeutic implications for chronic diseases, such as cardiovascular and respiratory pathologies, cancer, eye disorders, neurodegenerative conditions, and systemic or organ specific inflammatory and immune-mediated diseases. This comprehensive review summarizes and highlights the plethora of DMF's beneficial effects and underlines its repurposing opportunities in a variety of clinical conditions.
Collapse
Affiliation(s)
- Giorgia Bresciani
- Section of Pharmacology, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Federico Manai
- Department of Biology and Biotechnology L. Spallanzani, University of Pavia, Pavia, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology Vittorio Erspamer, Sapienza University, Rome, Italy
| | - Marialaura Amadio
- Section of Pharmacology, Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
8
|
Sharkus R, Thakkar R, Kolson DL, Constantinescu CS. Dimethyl Fumarate as Potential Treatment for Alzheimer's Disease: Rationale and Clinical Trial Design. Biomedicines 2023; 11:1387. [PMID: 37239057 PMCID: PMC10216730 DOI: 10.3390/biomedicines11051387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's Disease (AD) is a debilitating disease that leads to severe cognitive impairment and functional decline. The role of tau hyperphosphorylation and amyloid plaque deposition in the pathophysiology of AD has been well described; however, neuroinflammation and oxidative stress related to sustained microglial activation is thought to play a significant role in the disease process as well. NRF-2 has been identified in modulating the effects of inflammation and oxidative stress in AD. Activation of NRF-2 leads to an increased production of antioxidant enzymes, including heme oxygenase, which has been shown to have protective effects in neurodegenerative disorders such as AD. Dimethyl fumarate and diroximel fumarate (DMF) have been approved for the use in relapsing-remitting multiple sclerosis. Research indicates that they can modulate the effects of neuroinflammation and oxidative stress through the NRF-2 pathway, and as such, could serve as a potential therapeutic option in AD. We propose a clinical trial design that could be used to assess DMF as a treatment option for AD.
Collapse
Affiliation(s)
- Robert Sharkus
- Department of Neurology, Cooper Neurological Institute, Cherry Hill, NJ 08002, USA; (R.S.); (R.T.)
| | - Richa Thakkar
- Department of Neurology, Cooper Neurological Institute, Cherry Hill, NJ 08002, USA; (R.S.); (R.T.)
| | - Dennis L. Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Cris S. Constantinescu
- Department of Neurology, Cooper Neurological Institute, Cherry Hill, NJ 08002, USA; (R.S.); (R.T.)
- Department of Neurology, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
9
|
Abstract
Significance: Central nervous system (CNS) diseases are disorders of the brain and/or spinal cord and include neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor belonging to the cap-n-collar family that harbors a unique basic leucine zipper motif and plays as a master regulator of homeostatic responses. Recent Advances: Kelch-like ECH-associated protein 1 (KEAP1) is an adaptor of the Cullin3 (CUL3)-based ubiquitin E3 ligase that enhances the ubiquitylation of NRF2, which promotes the degradation of NRF2 to suppress its transcriptional activity in the absence of stress. Cysteine residues of KEAP1 are modified under stress conditions, and NRF2 degradation is attenuated, allowing it to accumulate and induce the expression of target genes. This regulatory system is referred to as the KEAP1-NRF2 system and plays a central role in protecting cells against various stresses. NRF2 also negatively regulates the expression of inflammatory cytokine and chemokine genes and suppresses pathological inflammation. As oxidative stress, inflammation, and proteostasis are known to contribute to neurodegenerative diseases, the KEAP1-NRF2 system is an attractive target for the treatment of these diseases. Critical Issues: In mouse models of neurodegenerative diseases, Nrf2 depletion exacerbates symptoms and enhances oxidative damage and inflammation in the CNS. In contrast, chemical or genetic NRF2 activation improves these symptoms. Indeed, the NRF2-activating chemical dimethyl fumarate is now widely used for the clinical treatment of MS. Future Directions: The KEAP1-NRF2 system is a promising therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Akira Uruno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
10
|
Jiang X, Song Y, Fang J, Yang X, Mu S, Zhang J. Neuroprotective effect of Vesatolimod in an experimental autoimmune encephalomyelitis mice model. Int Immunopharmacol 2023; 116:109717. [PMID: 36738672 DOI: 10.1016/j.intimp.2023.109717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/27/2022] [Accepted: 01/08/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Multiple sclerosis is a chronic demyelinating autoimmune disease accompanied by inflammation and loss of axons and neurons. Toll-like receptors play crucial roles in the innate immune system and inflammation. However, few studies have explored the specific effects of toll-like receptor 7 signaling pathway in multiple sclerosis. To explore underlying effects to develop a new therapeutic target, we use Vesatolimod, a safe and well-tolerated agonist of toll-like receptor 7, to assess the possible effects in Experimental autoimmune encephalomyelitis (EAE) animal model. METHODS EAE animal model was induced by injection of MOG35-55 and monitored daily for clinical symptoms, and the treatment group was given Vesatolimod at the onset of illness. The therapeutic effects of Vesatolimod on EAE inflammation, demyelination, CD107b cells and T cells infiltration, and microglia activation was evaluated. Autophagy within the spinal cords of EAE mice was also preliminarily assessed. RESULTS Treatment with Vesatolimod significantly alleviated clinical symptoms of EAE from day 18 post-immunization and decreased the expression levels of inflammatory cytokines, particularly Eotaxin and IL-12 (P40), in peripheral blood. It also inhibited demyelination in spinal cords. Moreover, VES treatment reduced activation of microglia, infiltration of CD3 + T cells and CD107b + cells, as well as inhibited the autophagy-related proteins expression in the spinal cords of EAE mice. CONCLUSION Our results indicate that Vesatolimod exhibits protective effects on EAE mice and is promising for treatment of MS.
Collapse
Affiliation(s)
- Xian Jiang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Yifan Song
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Jie Fang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Xiaosheng Yang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Shuhua Mu
- School of Psychology, Shenzhen University, Shenzhen 518060, Guangdong, China.
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
11
|
Uruno A, Yamamoto M. The KEAP1-NRF2 system and neurodegenerative diseases. Antioxid Redox Signal 2023. [PMID: 36734430 DOI: 10.1089/ars.2023.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Significance: Central nervous system (CNS) diseases are disorders of the brain and/or spinal cord and include neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). NF-E2-related factor 2 (NRF2) is a transcription factor belonging to the cap-n-collar (CNC) family that harbors a unique basic leucine zipper motif and plays as a master regulator of homeostatic responses. Recent Advances: Kelch-like ECH-associated protein 1 (KEAP1) is an adaptor of the Cullin3 (CUL3)-based ubiquitin E3 ligase that enhances the ubiquitylation of NRF2, which promotes the degradation of NRF2 to suppress its transcriptional activity in the absence of stress. Cysteine residues of KEAP1 are modified under stress conditions, and NRF2 degradation is attenuated, allowing it to accumulate and induce the expression of target genes. This regulatory system is referred to as the KEAP1-NRF2 system and plays a central role in protecting cells against various stresses. NRF2 also negatively regulates the expression of inflammatory cytokine and chemokine genes and suppresses pathological inflammation. As oxidative stress, inflammation, and proteostasis are known to contribute to neurodegenerative diseases, the KEAP1-NRF2 system is an attractive target for the treatment of these diseases. Critical Issues: In mouse models of neurodegenerative diseases, Nrf2 depletion exacerbates symptoms and enhances oxidative damage and inflammation in the CNS. In contrast, chemical or genetic NRF2 activation improves these symptoms. Indeed, the NRF2-activating chemical dimethyl fumarate (DMF) is now widely used for the clinical treatment of MS. Future Directions: The KEAP1-NRF2 system is a promising therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Akira Uruno
- Tohoku University, 13101, 2-1 Seiryo-cho, Aoba-ku, Sendai, Sendai, Miyagi, Japan, 980-8577;
| | - Masayuki Yamamoto
- Tohoku University Graduate School of Medicine, Department of Medical Biochemistry, 2-1 Seiryo-machi, Aoba-ku, Sendai, Sendai, Japan, 980-8575;
| |
Collapse
|
12
|
Chen BY, Salas JR, Trias AO, Rodriguez AP, Tsang JE, Guemes M, Le TM, Galic Z, Shepard HM, Steinman L, Nathanson DA, Czernin J, Witte ON, Radu CG, Schultz KA, Clark PM. Targeting deoxycytidine kinase improves symptoms in mouse models of multiple sclerosis. Immunology 2023; 168:152-169. [PMID: 35986643 PMCID: PMC9844239 DOI: 10.1111/imm.13569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/12/2022] [Indexed: 01/19/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease driven by lymphocyte activation against myelin autoantigens in the central nervous system leading to demyelination and neurodegeneration. The deoxyribonucleoside salvage pathway with the rate-limiting enzyme deoxycytidine kinase (dCK) captures extracellular deoxyribonucleosides for use in intracellular deoxyribonucleotide metabolism. Previous studies have shown that deoxyribonucleoside salvage activity is enriched in lymphocytes and required for early lymphocyte development. However, specific roles for the deoxyribonucleoside salvage pathway and dCK in autoimmune diseases such as MS are unknown. Here we demonstrate that dCK activity is necessary for the development of clinical symptoms in the MOG35-55 and MOG1-125 experimental autoimmune encephalomyelitis (EAE) mouse models of MS. During EAE disease, deoxyribonucleoside salvage activity is elevated in the spleen and lymph nodes. Targeting dCK with the small molecule dCK inhibitor TRE-515 limits disease severity when treatments are started at disease induction or when symptoms first appear. EAE mice treated with TRE-515 have significantly fewer infiltrating leukocytes in the spinal cord, and TRE-515 blocks activation-induced B and T cell proliferation and MOG35-55 -specific T cell expansion without affecting innate immune cells or naïve T and B cell populations. Our results demonstrate that targeting dCK limits symptoms in EAE mice and suggest that dCK activity is required for MOG35-55 -specific lymphocyte activation-induced proliferation.
Collapse
Affiliation(s)
- Bao Ying Chen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jessica R. Salas
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alyssa O. Trias
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Arely Perez Rodriguez
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan E. Tsang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Miriam Guemes
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Thuc M. Le
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Ahmanson Translational Imaging Division, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zoran Galic
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - David A. Nathanson
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Ahmanson Translational Imaging Division, University of California, Los Angeles, Los Angeles, CA, USA
| | - Johannes Czernin
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Ahmanson Translational Imaging Division, University of California, Los Angeles, Los Angeles, CA, USA
| | - Owen N. Witte
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Ahmanson Translational Imaging Division, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Peter M. Clark
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
13
|
Melamed E, Palmer JL, Fonken C. Advantages and limitations of experimental autoimmune encephalomyelitis in breaking down the role of the gut microbiome in multiple sclerosis. Front Mol Neurosci 2022; 15:1019877. [PMID: 36407764 PMCID: PMC9672668 DOI: 10.3389/fnmol.2022.1019877] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/11/2022] [Indexed: 08/22/2023] Open
Abstract
Since the first model of experimental autoimmune encephalomyelitis (EAE) was introduced almost a century ago, there has been an ongoing scientific debate about the risks and benefits of using EAE as a model of multiple sclerosis (MS). While there are notable limitations of translating EAE studies directly to human patients, EAE continues to be the most widely used model of MS, and EAE studies have contributed to multiple key breakthroughs in our understanding of MS pathogenesis and discovery of MS therapeutics. In addition, insights from EAE have led to a better understanding of modifiable environmental factors that can influence MS initiation and progression. In this review, we discuss how MS patient and EAE studies compare in our learning about the role of gut microbiome, diet, alcohol, probiotics, antibiotics, and fecal microbiome transplant in neuroinflammation. Ultimately, the combination of rigorous EAE animal studies, novel bioinformatic approaches, use of human cell lines, and implementation of well-powered, age- and sex-matched randomized controlled MS patient trials will be essential for improving MS patient outcomes and developing novel MS therapeutics to prevent and revert MS disease progression.
Collapse
Affiliation(s)
- Esther Melamed
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | | | | |
Collapse
|
14
|
Bevilacqua Rolfsen Ferreira da Silva G, Pereira das Neves S, Roque Oliveira SC, Marques F, Gomes de Oliveira A, de Lima Leite F, Cerqueira JJ. Comparative effectiveness of preventive treatment with dimethyl fumarate-loaded solid lipid nanoparticles and oral dimethyl fumarate in a mouse model of multiple sclerosis. J Autoimmun 2022; 132:102893. [PMID: 36029715 DOI: 10.1016/j.jaut.2022.102893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Orally administered dimethyl fumarate (DMF) presents gastrointestinal adverse effects, such as pain and diarrhea, in addition to flushing and lymphopenia. OBJECTIVE Solid lipid nanoparticles (SLNs) with DMF were developed for subcutaneous administration. METHODS DMF-incorporated SLNs and free DMF were tested in mice induced with experimental autoimmune encephalomyelitis (EAE). RESULTS Preventive treatment of free or incorporated DMF were able to reduce the EAE clinical scores, increase the weight of the animals, reduce the lesion area (demyelination and infiltration), reduce microglial fluorescence intensity and reduce the number of microglial cells and astrocytes, when compared to untreated EAE animals. Groups that received DMF had reduced numbers of T cells, B cells and natural killer (NK) cells in the blood, when compared to the non-induced group. CONCLUSIONS DMF incorporated in SLNs was as effective as free DMF in reducing the clinical scores of the animals, but with reduced administrations when given subcutaneously. In addition, SLN-DMF preventive treatment partially prevented a reduction in the percentages of T and B cells, in the blood, when compared to preventive treatment with free DMF (oral), which suggests reduction of lymphopenia.
Collapse
Affiliation(s)
- Gisela Bevilacqua Rolfsen Ferreira da Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; Nanoneurobiophysics Research Group, Department of Physics, Chemistry and Mathematics, Federal University of São Carlos (UFSCAR), Sorocaba, São Paulo, Brazil; São Paulo State University (UNESP), Department of Drugs and Medicines, Araraquara, São Paulo, Brazil.
| | - Sofia Pereira das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Susana Cristina Roque Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Anselmo Gomes de Oliveira
- São Paulo State University (UNESP), Department of Drugs and Medicines, Araraquara, São Paulo, Brazil
| | - Fábio de Lima Leite
- Nanoneurobiophysics Research Group, Department of Physics, Chemistry and Mathematics, Federal University of São Carlos (UFSCAR), Sorocaba, São Paulo, Brazil
| | - João José Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
15
|
Guerriero C, Puliatti G, Di Marino T, Tata AM. Effects Mediated by Dimethyl Fumarate on In Vitro Oligodendrocytes: Implications in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23073615. [PMID: 35408975 PMCID: PMC8998768 DOI: 10.3390/ijms23073615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Dimethyl fumarate (DMF) is a drug currently in use in oral therapy for the treatment of relapsing-remitting multiple sclerosis (RRMS) due to its immunomodulatory and neuroprotective effects. The mechanisms by which DMF exerts its therapeutic effects in MS and in particular its influence on the oligodendrocytes (OLs) survival or differentiation have not yet been fully understood. Methods: Characterization of Oli neu cells was performed by immunocytochemistry and RT-PCR. The effect of DMF on cell proliferation and morphology was assessed by MTT assay, trypan blue staining, RT-PCR and Western blot analysis. The antioxidant and anti-inflammatory properties of DMF were analysed by ROS detection through DCFDA staining and lipid content analysis by Oil Red O staining and TLC. Results: DMF has been observed to induce a slowdown of cell proliferation, favoring the oligodendrocyte lineage cells (OLCs) differentiation. DMF has an antioxidant effect and is able to modify the lipid content even after the LPS-mediated inflammatory stimulus in Oli neu cells. Conclusions: The results obtained confirm that DMF has anti-inflammatory and antioxidant effects also on Oli neu cells. Interestingly, it appears to promote the OLCs differentiation towards mature and potentially myelinating cells.
Collapse
Affiliation(s)
- Claudia Guerriero
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (G.P.); (T.D.M.)
| | - Giulia Puliatti
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (G.P.); (T.D.M.)
| | - Tamara Di Marino
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (G.P.); (T.D.M.)
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (C.G.); (G.P.); (T.D.M.)
- Research Centre of Neurobiology Daniel Bovet, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
16
|
Vainio SK, Dickens AM, Matilainen M, López-Picón FR, Aarnio R, Eskola O, Solin O, Anthony DC, Rinne JO, Airas L, Haaparanta-Solin M. Dimethyl fumarate decreases short-term but not long-term inflammation in a focal EAE model of neuroinflammation. EJNMMI Res 2022; 12:6. [PMID: 35107664 PMCID: PMC8811048 DOI: 10.1186/s13550-022-00878-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
Background Dimethyl fumarate (DMF) is an oral immunomodulatory drug used in the treatment of autoimmune diseases. Here, we sought to study whether the effect of DMF can be detected using positron emission tomography (PET) targeting the 18-kDa translocator protein (TSPO) in the focal delayed-type hypersensitivity rat model of multiple sclerosis (fDTH-EAE). The rats were treated orally twice daily from lesion activation (day 0) with either vehicle (tap water with 0.08% Methocel, 200 µL; control group n = 4 (3 after week four)) or 15 mg/kg DMF (n = 4) in 0.08% aqueous Methocel (200 µL) for 8 weeks. The animals were imaged by PET using the TSPO tracer [18F]GE-180 in weeks 0, 1, 2, 4, 8, and 18 following lesion activation, and the non-displaceable binding potential (BPND) was calculated. Immunohistochemical staining for Iba1, CD4, and CD8 was performed in week 18, and in separate cohorts of animals, following 2 or 4 weeks of treatment. Results Using the fDTH-EAE model, DMF reduced the [18F]GE-180 BPND in the DMF-treated animals compared to control animals after 1 week of treatment (two-tailed unpaired t test, p = 0.031), but not in weeks 2, 4, 8, or 18 when imaged in vivo by PET. Immunostaining for Iba1 showed that DMF had no effect on the perilesional volume or the core lesion volume after 2 or 4 weeks of treatment, or at 18 weeks. However, the optical density (OD) measurements of CD4+ staining showed reduced OD in the lesions of the treated rats. Conclusions DMF reduced the microglial activation in the fDTH-EAE model after 1 week of treatment, as detected by PET imaging of the TSPO ligand [18F]GE-180. However, over an extended time course, reduced microglial activation was not observed using [18F]GE-180 or by immunohistochemistry for Iba1+ microglia/macrophages. Additionally, DMF did affect the infiltration of CD4+ and CD8+ T-lymphocytes at the fDTH-EAE lesion. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00878-y. In a focal rat DTH-EAE model of neuroinflammation, dimethyl fumarate decreases the uptake of TSPO PET tracer [18F]GE-180 in the short term. Long-term [18F]GE-180 follow-up did not indicate a treatment effect. Decreased neuroinflammation, CD4+ T cell infiltration, and CD8+ T cell infiltration were detected using immunohistochemistry.
Collapse
Affiliation(s)
- S K Vainio
- Turku PET Centre, Preclinical PET Imaging, Preclinical Imaging Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland. .,MediCity Research Laboratory, University of Turku, Turku, Finland.
| | - A M Dickens
- Department of Chemistry, University of Turku, Turku, Finland.,Turku Bioscience, Turku, Finland
| | - M Matilainen
- Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - F R López-Picón
- Turku PET Centre, Preclinical PET Imaging, Preclinical Imaging Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - R Aarnio
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - O Eskola
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - O Solin
- Accelerator Laboratory, Åbo Akademi University, Turku, Finland.,Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - D C Anthony
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - J O Rinne
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - L Airas
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland.,Department of Clinical Medicine, University of Turku, Turku, Finland
| | - M Haaparanta-Solin
- Turku PET Centre, Preclinical PET Imaging, Preclinical Imaging Laboratory, University of Turku, Tykistökatu 6 A, 20520, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| |
Collapse
|
17
|
Kuntzel T, Bagnard D. Manipulating Macrophage/Microglia Polarization to Treat Glioblastoma or Multiple Sclerosis. Pharmaceutics 2022; 14:344. [PMID: 35214076 PMCID: PMC8877500 DOI: 10.3390/pharmaceutics14020344] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Macrophages and microglia are implicated in several diseases with divergent roles in physiopathology. This discrepancy can be explained by their capacity to endorse different polarization states. Theoretical extremes of these states are called M1 and M2. M1 are pro-inflammatory, microbicidal, and cytotoxic whereas M2 are anti-inflammatory, immunoregulatory cells in favor of tumor progression. In pathological states, these polarizations are dysregulated, thus restoring phenotypes could be an interesting treatment approach against diseases. In this review, we will focus on compounds targeting macrophages and microglia polarization in two very distinctive pathologies: multiple sclerosis and glioblastoma. Multiple sclerosis is an inflammatory disease characterized by demyelination and axon degradation. In this case, macrophages and microglia endorse a M1-like phenotype inducing inflammation. Promoting the opposite M2-like polarization could be an interesting treatment strategy. Glioblastoma is a brain tumor in which macrophages and microglia facilitate tumor progression, spreading, and angiogenesis. They are part of the tumor associated macrophages displaying an anti-inflammatory phenotype, thereby inhibiting anti-tumoral immunity. Re-activating them could be a method to limit and reduce tumor progression. These two pathologies will be used to exemplify that targeting the polarization of macrophages and microglia is a promising approach with a broad spectrum of applications deserving more attention.
Collapse
Affiliation(s)
- Thomas Kuntzel
- UMR7242 Biotechnology and Cell Signaling, Centre National de la Recherche Scientifique, Strasbourg Drug Discovery and Development Institute (IMS), University of Strasbourg, 67400 Illkirch-Graffenstaden, France;
| | - Dominique Bagnard
- UMR7242 Biotechnology and Cell Signaling, Centre National de la Recherche Scientifique, Strasbourg Drug Discovery and Development Institute (IMS), University of Strasbourg, 67400 Illkirch-Graffenstaden, France;
- Ecole Supérieure de Biotechnologie de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
18
|
Pinto BF, Ribeiro LNB, da Silva GBRF, Freitas CS, Kraemer L, Oliveira FMS, Clímaco MC, Mourão FAG, Santos GSPD, Béla SR, Gurgel ILDS, Leite FDL, de Oliveira AG, Vilela MRSDP, Oliveira-Lima OC, Soriani FM, Fujiwara RT, Birbrair A, Russo RC, Carvalho-Tavares J. Inhalation of dimethyl fumarate-encapsulated solid lipid nanoparticles attenuate clinical signs of experimental autoimmune encephalomyelitis and pulmonary inflammatory dysfunction in mice. Clin Sci (Lond) 2022; 136:81-101. [PMID: 34904644 DOI: 10.1042/cs20210792] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022]
Abstract
RATIONALE The FDA-approved Dimethyl Fumarate (DMF) as an oral drug for Multiple Sclerosis (MS) treatment based on its immunomodulatory activities. However, it also caused severe adverse effects mainly related to the gastrointestinal system. OBJECTIVE Investigated the potential effects of solid lipid nanoparticles (SLNs) containing DMF, administered by inhalation on the clinical signs, central nervous system (CNS) inflammatory response, and lung function changes in mice with experimental autoimmune encephalomyelitis (EAE). MATERIALS AND METHODS EAE was induced using MOG35-55 peptide in female C57BL/6J mice and the mice were treated via inhalation with DMF-encapsulated SLN (CTRL/SLN/DMF and EAE/SLN/DMF), empty SLN (CTRL/SLN and EAE/SLN), or saline solution (CTRL/saline and EAE/saline), every 72 h during 21 days. RESULTS After 21 days post-induction, EAE mice treated with DMF-loaded SLN, when compared with EAE/saline and EAE/SLN, showed decreased clinical score and weight loss, reduction in brain and spinal cord injury and inflammation, also related to the increased influx of Foxp3+ cells into the spinal cord and lung tissues. Moreover, our data revealed that EAE mice showed signs of respiratory disease, marked by increased vascular permeability, leukocyte influx, production of TNF-α and IL-17, perivascular and peribronchial inflammation, with pulmonary mechanical dysfunction associated with loss of respiratory volumes and elasticity, which DMF-encapsulated reverted in SLN nebulization. CONCLUSION Our study suggests that inhalation of DMF-encapsulated SLN is an effective therapeutic protocol that reduces not only the CNS inflammatory process and disability progression, characteristic of EAE disease, but also protects mice from lung inflammation and pulmonary dysfunction.
Collapse
Affiliation(s)
- Bárbara Fernandes Pinto
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lorena Natasha Brito Ribeiro
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Gisela Bevilacqua Rolfsen Ferreira da Silva
- Nanoneurobiophysics Research Group, Department of Physics, Chemistry and Mathematics, Federal University of São Carlos (UFSCAR), Sorocaba, São Paulo, Brazil
- State of São Paulo University (UNESP), Drugs and Medicines Department, School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil
| | - Camila Simões Freitas
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lucas Kraemer
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Marianna Carvalho Clímaco
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Flávio Afonso Gonçalves Mourão
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | | | - Samantha Ribeiro Béla
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Isabella Luísa da Silva Gurgel
- Laboratory of Functional Genetics, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Fábio de Lima Leite
- Nanoneurobiophysics Research Group, Department of Physics, Chemistry and Mathematics, Federal University of São Carlos (UFSCAR), Sorocaba, São Paulo, Brazil
| | - Anselmo Gomes de Oliveira
- State of São Paulo University (UNESP), Drugs and Medicines Department, School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil
| | - Maura Regina Silva da Páscoa Vilela
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Onésia Cristina Oliveira-Lima
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás (UFG), Goiânia, GO, Brazil
| | - Frederico Marianetti Soriani
- Laboratory of Functional Genetics, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Juliana Carvalho-Tavares
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
19
|
Melnikov M, Sviridova A, Rogovskii V, Boyko A, Pashenkov M. The role of macrophages in the development of neuroinflammation in multiple sclerosis. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:51-56. [DOI: 10.17116/jnevro202212205151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
20
|
Correale J, Halfon MJ, Jack D, Rubstein A, Villa A. Acting centrally or peripherally: A renewed interest in the central nervous system penetration of disease-modifying drugs in multiple sclerosis. Mult Scler Relat Disord 2021; 56:103264. [PMID: 34547609 DOI: 10.1016/j.msard.2021.103264] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 09/03/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
With the recent approval of cladribine tablets, siponimod and ozanimod, there has been a renewed interest into the extent to which these current generation disease-modifying therapies (DMTs) are able to cross into the central nervous system (CNS), and how this penetration of the blood-brain barrier (BBB) may influence their ability to treat multiple sclerosis (MS). The integrity of the CNS is maintained by the BBB, blood-cerebrospinal fluid barrier, and the arachnoid barrier, which all play an important role in preserving the immunological environment and homeostasis within the CNS. The integrity of the BBB decreases during the course of MS, with a putative temporal relationship to disease worsening. Furthermore, it is currently considered that progression of the disease is mediated mainly by resident cells of the CNS. The existing literature provides evidence to show that some of the current generation DMTs for MS are able to penetrate the CNS and potentially exert direct effects on CNS-resident cells, in particular the CNS-penetrating prodrugs cladribine and fingolimod, and other sphingosine-1 phosphate receptor modulators; siponimod and ozanimod. Other current generation DMTs appear to be restricted to the periphery due to their high molecular weight or physicochemical properties. As more effective brain penetrant therapies are developed for the treatment of MS, there is a need to understand whether the potential for direct effects within the CNS are of significance, and whether this brings additional benefits over and above treatment effects mediated in the periphery. In turn, this will require an improved understanding of the structure and function of the BBB, the role it plays in MS and subsequent treatments. This narrative review summarizes the data supporting the biological plausibility of a potential benefit from therapeutic molecules entering the CNS, and discusses the potential significance in the current and future treatment of MS.
Collapse
Affiliation(s)
- Jorge Correale
- Department of Neurology, Fleni, Buenos Aires, Argentina.
| | | | - Dominic Jack
- Merck Serono Ltd, Feltham, United Kingdom (an affiliate of Merck KGaA)
| | - Adrián Rubstein
- Merck S.A., Buenos Aires, Argentina (an affiliate of Merck KGaA)
| | - Andrés Villa
- Hospital Ramos Mejía, Universidad de Buenos Aires, Argentina
| |
Collapse
|
21
|
Radandish M, Khalilian P, Esmaeil N. The Role of Distinct Subsets of Macrophages in the Pathogenesis of MS and the Impact of Different Therapeutic Agents on These Populations. Front Immunol 2021; 12:667705. [PMID: 34489926 PMCID: PMC8417824 DOI: 10.3389/fimmu.2021.667705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/31/2021] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating inflammatory disorder of the central nervous system (CNS). Besides the vital role of T cells, other immune cells, including B cells, innate immune cells, and macrophages (MФs), also play a critical role in MS pathogenesis. Tissue-resident MФs in the brain’s parenchyma, known as microglia and monocyte-derived MФs, enter into the CNS following alterations in CNS homeostasis that induce inflammatory responses in MS. Although the neuroprotective and anti-inflammatory actions of monocyte-derived MФs and resident MФs are required to maintain CNS tolerance, they can release inflammatory cytokines and reactivate primed T cells during neuroinflammation. In the CNS of MS patients, elevated myeloid cells and activated MФs have been found and associated with demyelination and axonal loss. Thus, according to the role of MФs in neuroinflammation, they have attracted attention as a therapeutic target. Also, due to their different origin, location, and turnover, other strategies may require to target the various myeloid cell populations. Here we review the role of distinct subsets of MФs in the pathogenesis of MS and different therapeutic agents that target these cells.
Collapse
Affiliation(s)
- Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Khalilian
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
22
|
Farina M, Vieira LE, Buttari B, Profumo E, Saso L. The Nrf2 Pathway in Ischemic Stroke: A Review. Molecules 2021; 26:5001. [PMID: 34443584 PMCID: PMC8399750 DOI: 10.3390/molecules26165001] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke, characterized by the sudden loss of blood flow in specific area(s) of the brain, is the leading cause of permanent disability and is among the leading causes of death worldwide. The only approved pharmacological treatment for acute ischemic stroke (intravenous thrombolysis with recombinant tissue plasminogen activator) has significant clinical limitations and does not consider the complex set of events taking place after the onset of ischemic stroke (ischemic cascade), which is characterized by significant pro-oxidative events. The transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates the expression of a great number of antioxidant and/or defense proteins, has been pointed as a potential pharmacological target involved in the mitigation of deleterious oxidative events taking place at the ischemic cascade. This review summarizes studies concerning the protective role of Nrf2 in experimental models of ischemic stroke, emphasizing molecular events resulting from ischemic stroke that are, in parallel, modulated by Nrf2. Considering the acute nature of ischemic stroke, we discuss the challenges in using a putative pharmacological strategy (Nrf2 activator) that relies upon transcription, translation and metabolically active cells in treating ischemic stroke patients.
Collapse
Affiliation(s)
- Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, Brazil;
| | - Leonardo Eugênio Vieira
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, Brazil;
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Elisabetta Profumo
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
23
|
Möhle L, Brackhan M, Bascuñana P, Pahnke J. Dimethyl fumarate does not mitigate cognitive decline and β-amyloidosis in female APPPS1 mice. Brain Res 2021; 1768:147579. [PMID: 34233173 DOI: 10.1016/j.brainres.2021.147579] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/14/2021] [Accepted: 06/30/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the leading cause of dementia and a major global health issue. Currently, only limited treatment options are available to patients. One possibility to expand the treatment repertoire is repurposing of existing drugs such as dimethyl fumarate (DMF). DMF is approved for treatment of multiple sclerosis and previous animal studies have suggested that DMF may also have a beneficial effect for the treatment of AD. METHODS We used an APPPS1 transgenic model of senile β-amyloidosis and treated female mice orally with DMF in two treatment paradigms (pre and post onset). We quantified learning and memory parameters, β-amyloidosis, and neuroinflammation to determine the potential of DMF as AD therapeutics. RESULTS Treatment with DMF had no influence on water maze performance, β-amyloid accumulation, plaque formation, microglia activation, and recruitment of immune cells to the brain. Compared to vehicle-treated animals, oral DMF treatment could not halt or retard disease progression in the mice. DISCUSSION Our results do not favour the use of DMF as treatment for AD. While our results stand in contrast to previous findings in other models, they emphasize the importance of animal model selection and suggest further studies to elucidate the mechanisms leading to conflicting results.
Collapse
Affiliation(s)
- Luisa Möhle
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway.
| | - Mirjam Brackhan
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway; LIED, University of Lübeck, Lübeck, Germany
| | - Pablo Bascuñana
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
| | - Jens Pahnke
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway; LIED, University of Lübeck, Lübeck, Germany; Department of Pharmacology, Faculty of Medicine, University of Latvia, Rīga, Latvia.
| |
Collapse
|
24
|
Dimethyl Fumarate, an Approved Multiple Sclerosis Treatment, Reduces Brain Oxidative Stress in SIV-Infected Rhesus Macaques: Potential Therapeutic Repurposing for HIV Neuroprotection. Antioxidants (Basel) 2021; 10:antiox10030416. [PMID: 33803289 PMCID: PMC7998206 DOI: 10.3390/antiox10030416] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
Dimethyl fumarate (DMF), an antioxidant/anti-inflammatory drug approved for the treatment of multiple sclerosis, induces antioxidant enzymes, in part through transcriptional upregulation. We hypothesized that DMF administration to simian immunodeficiency virus (SIV)-infected rhesus macaques would induce antioxidant enzyme expression and reduce oxidative injury and inflammation throughout the brain. Nine SIV-infected, CD8+-T-lymphocyte-depleted rhesus macaques were studied. Five received oral DMF prior to the SIV infection and through to the necropsy day. Protein expression was analyzed in 11 brain regions, as well as the thymus, liver, and spleen, using Western blot and immunohistochemistry for antioxidant, inflammatory, and neuronal proteins. Additionally, oxidative stress was determined in brain sections using immunohistochemistry (8-OHdG, 3NT) and optical redox imaging of oxidized flavoproteins containing flavin adenine dinucleotide (Fp) and reduced nicotinamide adenine dinucleotide (NADH). The DMF treatment was associated with no changes in virus replication; higher expressions of the antioxidant enzymes NQO1, GPX1, and HO-1 in the brain and PRDX1 and HO-2 in the spleen; lower levels of 8-OHdG and 3NT; a lower optical redox ratio. The DMF treatment was also associated with increased expressions of cell-adhesion molecules (VCAM-1, ICAM-1) and no changes in HLA-DR, CD68, GFAP, NFL, or synaptic proteins. The concordantly increased brain antioxidant enzyme expressions and reduced oxidative stress in DMF-treated SIV-infected macaques suggest that DMF could limit oxidative stress throughout the brain through effective induction of the endogenous antioxidant response. We propose that DMF could potentially induce neuroprotective brain responses in persons living with HIV.
Collapse
|
25
|
Certo M, Tsai CH, Pucino V, Ho PC, Mauro C. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol 2021; 21:151-161. [PMID: 32839570 DOI: 10.1038/s41577-020-0406-2] [Citation(s) in RCA: 351] [Impact Index Per Article: 117.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2020] [Indexed: 12/15/2022]
Abstract
The microenvironment in cancerous tissues is immunosuppressive and pro-tumorigenic, whereas the microenvironment of tissues affected by chronic inflammatory disease is pro-inflammatory and anti-resolution. Despite these opposing immunological states, the metabolic states in the tissue microenvironments of cancer and inflammatory diseases are similar: both are hypoxic, show elevated levels of lactate and other metabolic by-products and have low levels of nutrients. In this Review, we describe how the bioavailability of lactate differs in the microenvironments of tumours and inflammatory diseases compared with normal tissues, thus contributing to the establishment of specific immunological states in disease. A clear understanding of the metabolic signature of tumours and inflammatory diseases will enable therapeutic intervention aimed at resetting the bioavailability of metabolites and correcting the dysregulated immunological state, triggering beneficial cytotoxic, inflammatory responses in tumours and immunosuppressive responses in chronic inflammation.
Collapse
Affiliation(s)
- Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Chin-Hsien Tsai
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Valentina Pucino
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Ping-Chih Ho
- Department of Oncology UNIL CHUV, Ludwig Institute for Cancer Research, Lausanne, Switzerland.
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
26
|
Aqel SI, Yang X, Kraus EE, Song J, Farinas MF, Zhao EY, Pei W, Lovett-Racke AE, Racke MK, Li C, Yang Y. A STAT3 inhibitor ameliorates CNS autoimmunity by restoring Teff:Treg balance. JCI Insight 2021; 6:142376. [PMID: 33411696 PMCID: PMC7934926 DOI: 10.1172/jci.insight.142376] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Reestablishing an appropriate balance between T effector cells (Teff) and Tregs is essential for correcting autoimmunity. Multiple sclerosis (MS) is an immune-mediated chronic CNS disease characterized by neuroinflammation, demyelination, and neuronal degeneration, in which the Teff:Treg balance is skewed toward pathogenic Teffs Th1 and Th17 cells. STAT3 is a key regulator of Teff:Treg balance. Using the structure-based design, we have developed a potentially novel small-molecule prodrug LLL12b that specifically inhibits STAT3 and suppresses Th17 differentiation and expansion. Moreover, LLL12b regulates the fate decision between Th17 and Tregs in an inflammatory environment, shifting Th17:Treg balance toward Tregs and favoring the resolution of inflammation. Therapeutic administration of LLL12b after disease onset significantly suppresses disease progression in adoptively transferred, chronic, and relapsing-remitting experimental autoimmune encephalomyelitis. Disease relapses were also significantly suppressed by LLL12b given during the remission phase. Additionally, LLL12b shifts Th17:Treg balance of CD4+ T cells from MS patients toward Tregs and increases Teff sensitivity to Treg-mediated suppression. These data suggest that selective inhibition of STAT3 by the small molecule LLL12b recalibrates the effector and regulatory arms of CD4+ T responses, representing a potentially clinically translatable therapeutic strategy for MS.
Collapse
Affiliation(s)
- Saba I Aqel
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Xiaozhi Yang
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Emma E Kraus
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Jinhua Song
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Marissa F Farinas
- Neuroscience program, College of Arts and Sciences, OSU, Columbus, Ohio, USA
| | - Erin Y Zhao
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Wei Pei
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, OSU Wexner Medical Center, Columbus, Ohio, USA
| | - Michael K Racke
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA.,Quest Diagnostics, Secaucus, New Jersey, USA
| | - Chenglong Li
- Division of Medicinal Chemistry, College of Pharmacy, OSU, Columbus, Ohio, USA.,Department of Medicinal Chemistry, University of Florida, Gainesville, Florida, USA
| | - Yuhong Yang
- Department of Neurology, Ohio State University (OSU) Wexner Medical Center, Columbus, Ohio, USA.,Department of Microbial Infection and Immunity, OSU Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
27
|
Yadav SK, Ito N, Soin D, Ito K, Dhib-Jalbut S. Dimethyl Fumarate Suppresses Demyelination and Axonal Loss through Reduction in Pro-Inflammatory Macrophage-Induced Reactive Astrocytes and Complement C3 Deposition. J Clin Med 2021; 10:jcm10040857. [PMID: 33669652 PMCID: PMC7922578 DOI: 10.3390/jcm10040857] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/06/2021] [Accepted: 02/14/2021] [Indexed: 01/24/2023] Open
Abstract
Dimethyl fumarate (DMF) is an oral agent for relapsing-remitting multiple sclerosis (RRMS). In this study, we investigated the therapeutic mechanism of DMF using experimental autoimmune encephalomyelitis (EAE). DMF treatment decreased the proliferation of T cells and the production of IL-17A and GM-CSF. DMF treatment also decreased the development and/or infiltration of macrophages in the central nervous system (CNS), and reduced the ratio of iNOS+ pro-inflammatory macrophage versus Ym1+ immunomodulatory macrophages. Furthermore, DMF treatment suppressed the deposition of complement C3 (C3) and development of reactive C3+ astrocytes. The decrease in iNOS+ macrophages, C3+astrocytes, and C3 deposition in the CNS resulted in the reduction in demyelination and axonal loss. This study suggests that the beneficial effects of DMF involve the suppression of iNOS+ pro-inflammatory macrophages, C3+ astrocytes, and deposition of C3 in the CNS.
Collapse
Affiliation(s)
- Sudhir K. Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (N.I.); (D.S.)
| | - Naoko Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (N.I.); (D.S.)
| | - Devika Soin
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (N.I.); (D.S.)
- Touro College of Osteopathic Medicine, Middletown, NY 10940, USA
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (N.I.); (D.S.)
- Correspondence: (K.I.); (S.D.-J.); Tel.: +1-732-235-5482 (K.I.); +1-732-235-7335 (S.D.-J.)
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (N.I.); (D.S.)
- Department of Neurology, New Jersey Medical School, Newark, NJ 07101, USA
- Correspondence: (K.I.); (S.D.-J.); Tel.: +1-732-235-5482 (K.I.); +1-732-235-7335 (S.D.-J.)
| |
Collapse
|
28
|
Goess C, Terrillon S, Mayo M, Bousquet P, Wallace C, Hart M, Mathieu S, Twomey R, Donnelly-Roberts D, Namovic M, Jung P, Hu M, Richardson P, Esbenshade T, Cuff CA. NRF2 activator A-1396076 ameliorates inflammation in autoimmune disease models by inhibiting antigen dependent T cell activation. J Transl Autoimmun 2021; 4:100079. [PMID: 33490940 PMCID: PMC7809192 DOI: 10.1016/j.jtauto.2020.100079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 10/26/2022] Open
Abstract
Nuclear factor (erythroid-derived 2) like 2 (NRF2) is a nuclear transcription factor activated in response to oxidative stress that induces a gene program that dampens inflammation and can limit cell damage that perpetuates the inflammatory response. We have identified A-1396076, a potent and selective NRF2 activator with demonstrated KEAP1 binding and modulation of cellular NRF2 mediated effects. In vivo administration of A-1396076 inhibits inflammation across several rodent models of autoimmunity when administered at or before the time of antigen challenge while also inducing NRF2 modulated gene transcription in the liver of the animals. It was not effective when administered after the time of antigen challenge or in a T cell independent model of arthritis induced by passive transfer of anti-collagen antibodies. A-1396076 inhibited antigen dependent T cell activation as measured by IFN-γ production in an ex vivo re-stimulation assay and following anti-CD3 challenge of MOG-sensitized mice. A-1396076 reduced costimulatory molecule expression on dendritic cells in the lungs of OVA LPS challenged mice suggesting that the mechanism of T cell inhibition was mediated at least partially by interfering with antigen presentation. These data suggest that NRF2 activation may be an effective strategy to dampen inflammation for treatment of autoimmune disease.
Collapse
Affiliation(s)
- Christian Goess
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Sonia Terrillon
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Martha Mayo
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Peter Bousquet
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Craig Wallace
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Michelle Hart
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Suzanne Mathieu
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Rachel Twomey
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | | | - Marian Namovic
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Paul Jung
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Min Hu
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Paul Richardson
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Tim Esbenshade
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL, 60064, USA
| | - Carolyn A Cuff
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| |
Collapse
|
29
|
Islam SMT, Won J, Kim J, Qiao F, Singh AK, Khan M, Singh I. Detoxification of Reactive Aldehydes by Alda-1 Treatment Ameliorates Experimental Autoimmune Encephalomyelitis in Mice. Neuroscience 2021; 458:31-42. [PMID: 33493617 DOI: 10.1016/j.neuroscience.2021.01.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/12/2020] [Accepted: 01/12/2021] [Indexed: 11/28/2022]
Abstract
Reactive aldehydes are generated as a toxic end-product of lipid peroxidation under inflammatory oxidative stress condition which is a well-established phenomenon in the pathogenesis of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Alda-1, a selective agonist of mitochondrial aldehyde dehydrogenase 2 (ALDH2), is known to detoxify the reactive aldehydes. In this study, we investigated the effect of Alda-1 on CNS myelin pathology associated with reactive aldehydes and mitochondrial/peroxisomal dysfunctions in a mouse model of EAE. Daily treatment of EAE mice with Alda-1, starting at the peak of disease, ameliorated the clinical manifestation of disease along with the improvement of motor functions. Accordingly, Alda-1 treatment improved demyelination and neuroaxonal degeneration in EAE mice. EAE mice had increased levels of reactive aldehyde species, such as 4-hydroxynonenal (4-HNE), malondialdehyde (MDA), and acrolein (ACL) in the spinal cords and these levels were significantly reduced in Alda-1-treated EAE mice. Furthermore, Alda-1 treatment improved the loss of mitochondrial (OXPHOS) and peroxisomal (PMP70 and catalase) proteins as well as mitochondrial/peroxisomal proliferation factors (PGC-1α and PPARs) in the spinal cords of EAE mice. Taken together, this study demonstrates the therapeutic efficacy of ALDH2-agonist Alda-1 in the abatement of EAE disease through the detoxification of reactive aldehydes, thus suggesting Alda-1 as a potential therapeutic intervention for MS.
Collapse
Affiliation(s)
- S M Touhidul Islam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Judong Kim
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Fei Qiao
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| |
Collapse
|
30
|
Banik A, Ghosh K, Pal S, Halder SK, Ghosh C, Mondal KC. Biofortification of multi-grain substrates by probiotic yeast. FOOD BIOTECHNOL 2020. [DOI: 10.1080/08905436.2020.1833913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Abhijit Banik
- Department of Microbiology, Vidyasagar University, Midnapore, India
- Department of Human Physiology with Community Health, Vidyasagar University, Midnapore, India
| | - Kuntal Ghosh
- Department of Biological Sciences, Midnapore City College, Bhadutala, Paschim Medinipur, Midnapore, India
| | - Shilpee Pal
- Bioinformatics Infrastructure Facility Centre, Department of Microbiology, Vidyasagar University, Midnapore, India
| | | | - Chandradipa Ghosh
- Department of Human Physiology with Community Health, Vidyasagar University, Midnapore, India
| | | |
Collapse
|
31
|
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: 50] [Impact Index Per Article: 12.5] [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
|
32
|
Unni S, Deshmukh P, Krishnappa G, Kommu P, Padmanabhan B. Structural insights into the multiple binding modes of Dimethyl Fumarate (DMF) and its analogs to the Kelch domain of Keap1. FEBS J 2020; 288:1599-1613. [PMID: 32672401 DOI: 10.1111/febs.15485] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 11/29/2022]
Abstract
The activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) transcription function has been implicated in the protection of neurodegenerative diseases. The cytoplasmic protein, Kelch-like ECH-associated protein 1 (Keap1), negatively regulates Nrf2. The Keap1-Nrf2 pathway is a potential therapeutic target for tackling free-radical damage. Dimethyl fumarate (DMF) is currently an approved drug for the treatment of relapsing multiple sclerosis. Recent studies showed that DMF modifies the reactive cysteines in the BTB domain of Keap1 and thus activates Nrf2 transcription function. Intriguingly, our crystal structure studies revealed that DMF also binds to the β-propeller domain (Keap1-DC) of Keap1. The crystal structure of the complex, refined to 1.54 Å resolution, revealed unexpected features: DMF binds (a) to the Nrf2-binding site (bottom region of Keap1-DC, site 1) with moderate interaction, and (b) to the top region of Keap1-DC, near to the blade II (site 2). The specificity of the binding 'site 2' was found to be unique to blade II of the β-propeller domain. The newly identified 'site 2' region in Keap1-DC may have a different functional role to regulate Nrf2. Moreover, the crystal structures of Keap1-DC in complex with the DMF analogs, including monoethyl fumarate, fumarate, and itaconate, also exhibited similar binding modes with Keap1-DC. Binding studies confirmed that DMF binds, in a nanomolar range, to the Keap1-DC region as well as the BTB domain of Keap1. Furthermore, the competitive binding assay in the presence of the Nrf2 peptide affirmed the direct binding of DMF at the Nrf2-binding region of Keap1-DC. Overall, our studies suggest that the drug molecule, DMF, binds at multiple sites of Keap1 and thus potentially activates Nrf2 function through covalent as well as the noncovalent mode of action, to combat oxidative stress. DATABASE: Structural data are available in RCSB-protein data bank database(s) under the accession numbers 6LRZ, 7C60, and 7C5E.
Collapse
Affiliation(s)
- Sruthi Unni
- Department of Biophysics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Prashant Deshmukh
- Department of Biophysics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Gopinatha Krishnappa
- Department of Biophysics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Padmini Kommu
- Department of Biophysics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - Balasundaram Padmanabhan
- Department of Biophysics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| |
Collapse
|
33
|
Scuderi SA, Ardizzone A, Paterniti I, Esposito E, Campolo M. Antioxidant and Anti-inflammatory Effect of Nrf2 Inducer Dimethyl Fumarate in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:antiox9070630. [PMID: 32708926 PMCID: PMC7402174 DOI: 10.3390/antiox9070630] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (NDs) represents debilitating conditions characterized by degeneration of neuronal cells in specific brain areas, causing disability and death in patients. In the pathophysiology of NDs, oxidative stress, apoptosis and neuroinflammation have a key role, as demonstrated by in vivo and in vitro models. Therefore, the use of molecules with antioxidant and anti-inflammatory activities represents a possible strategy for the treatment of NDs. Many studies demonstrated the beneficial effects of fumaric acid esters (FAEs) to counteract neuroinflammation and oxidative stress. Among these molecules, dimethyl fumarate (DMF) showed a valid therapeutic approach to slow down neurodegeneration and relieve symptoms in patients with NDs. DMF is a methyl ester of fumaric acid and acts as modulator of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway as well as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) translocation. Therefore, this review aims to examine the potential beneficial effects of DMF to counteract oxidative stress and inflammation in patients with NDs.
Collapse
|
34
|
Kempe PRG, Chiarotto GB, Barraviera B, Ferreira RS, de Oliveira ALR. Neuroprotection and immunomodulation by dimethyl fumarate and a heterologous fibrin biopolymer after ventral root avulsion and reimplantation. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190093. [PMID: 32518556 PMCID: PMC7250131 DOI: 10.1590/1678-9199-jvatitd-2019-0093] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background: Ventral root avulsion (VRA) is an experimental approach in which there is an abrupt separation of the motor roots from the surface of the spinal cord. As a result, most of the axotomized motoneurons degenerate by the second week after injury, and the significant loss of synapses and increased glial reaction triggers a chronic inflammatory state. Pharmacological treatment associated with root reimplantation is thought to overcome the degenerative effects of VRA. Therefore, treatment with dimethyl fumarate (DMF), a drug with neuroprotective and immunomodulatory effects, in combination with a heterologous fibrin sealant/biopolymer (FS), a biological glue, may improve the regenerative response. Methods: Adult female Lewis rats were subjected to VRA of L4-L6 roots followed by reimplantation and daily treatment with DMF for four weeks. Survival times were evaluated 1, 4 or 12 weeks after surgery. Neuronal survival assessed by Nissl staining, glial reactivity (anti-GFAP for astrocytes and anti-Iba-1 for microglia) and synapse preservation (anti-VGLUT1 for glutamatergic inputs and anti-GAD65 for GABAergic inputs) evaluated by immunofluorescence, gene expression (pro- and anti-inflammatory molecules) and motor function recovery were measured. Results: Treatment with DMF at a dose of 15 mg/kg was found to be neuroprotective and immunomodulatory because it preserved motoneurons and synapses and decreased astrogliosis and microglial reactions, as well as downregulated the expression of pro-inflammatory gene transcripts. Conclusion: The pharmacological benefit was further enhanced when associated with root reimplantation with FS, in which animals recovered at least 50% of motor function, showing the efficacy of employing multiple regenerative approaches following spinal cord root injury.
Collapse
Affiliation(s)
- Paula R G Kempe
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Benedito Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | |
Collapse
|
35
|
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
|
36
|
das Neves SP, Santos G, Barros C, Pereira DR, Ferreira R, Mota C, Monteiro S, Fernandes A, Marques F, Cerqueira JJ. Enhanced cognitive performance in experimental autoimmune encephalomyelitis mice treated with dimethyl fumarate after the appearance of disease symptoms. J Neuroimmunol 2020; 340:577163. [PMID: 31982706 DOI: 10.1016/j.jneuroim.2020.577163] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/02/2020] [Accepted: 01/10/2020] [Indexed: 12/23/2022]
Abstract
In multiple sclerosis (MS), cognitive dysfunction is common but difficult to treat. We analyzed the impact of dimethyl fumarate, an MS drug with neuroprotective properties, in spatial memory performance in a mouse model of MS and looked for structural correlates in the hippocampus. Treated mice presented better cognitive performance which was not associated with structural hippocampal damage but with decreased demyelination in the fimbria. Dimethyl fumarate, even if initiated after hindlimb paralysis, ameliorated memory deficits in the MS mouse model due, at least in part, to its positive impact in the demyelination of the main hippocampal output pathway.
Collapse
Affiliation(s)
- Sofia P das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Gisela Santos
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Catarina Barros
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Diana Rodrigues Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Mota
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adelaide Fernandes
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João José Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 2CA, Clinical Academic Center - Braga, Braga, Portugal.
| |
Collapse
|
37
|
Lynch MA. Can the emerging field of immunometabolism provide insights into neuroinflammation? Prog Neurobiol 2019; 184:101719. [PMID: 31704314 DOI: 10.1016/j.pneurobio.2019.101719] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/18/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
In the past few years it has become increasingly clear that an understanding of the interaction between metabolism and immune function can provide an insight into cellular responses to challenges. Significant progress has been made in terms of how macrophages are metabolically re-programmed in response to inflammatory stimuli but, to date, little emphasis has been placed on evaluating equivalent changes in microglia. The need to make progress is driven by the fact that, while microglial activation and the cell's ability to adopt an inflammatory phenotype is necessary to fulfil the neuroprotective function of the cell, persistent activation of microglia and the associated neuroinflammation is at the heart of several neurodegenerative diseases. Understanding the metabolic changes that accompany microglial responses may broaden our perspective on how dysfunction might arise and be tempered. This review will evaluate the current literature that addresses the interplay between inflammation and metabolic reprogramming in microglia, reflecting on the parallels that exist with macrophages. It will consider the changes that take place with age including those that have been reported in neurons and astrocytes with the development of non-invasive imaging techniques, and reflect on the literature that is currently available relating to metabolic reprogramming of microglia with age and in neurodegeneration. Finally it will consider the possibility that manipulating microglial metabolism may provide a valuable approach to modulating neuroinflammation.
Collapse
Affiliation(s)
- Marina A Lynch
- Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
38
|
Pitarokoili K, Bachir H, Sgodzai M, Grüter T, Haupeltshofer S, Duscha A, Pedreiturria X, Motte J, Gold R. Induction of Regulatory Properties in the Intestinal Immune System by Dimethyl Fumarate in Lewis Rat Experimental Autoimmune Neuritis. Front Immunol 2019; 10:2132. [PMID: 31552056 PMCID: PMC6746892 DOI: 10.3389/fimmu.2019.02132] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 08/23/2019] [Indexed: 12/22/2022] Open
Abstract
Objective: Dimethyl fumarate (DMF) exerts immunomodulatory and neuroprotective effects in the animal model of experimental autoimmune neuritis (EAN) in the Lewis rat. DMF has been shown to modulate gut microbiota in veterinary medicine, however the effects of oral DMF on the gut-associated lymphoid tissue (GALT) remain unknown. Methods: Lewis rats were treated orally twice daily with DMF up to day 10 after immunization with immunogenic P2 peptide. Histological, flow cytometric and RT-PCR analyses of the GALT (intraepithelial layer, lamina propria, and Peyer patches) in duodenum, jejunum, and ileum were performed ex vivo. Moreover, cell transfer experiments were used to examine the protective effects of GALT regulatory T cells of the Peyer patches. Results: In the upper layers of duodenum, DMF induced a reduction of the toll-like receptor 4 (TLR4) mRNA expression. This was combined by a decrease of the pro-inflammatory lamina propria IFN-γ mRNA expression. In the ileum, we detected an immunoregulatory phenotype characterized by an increase of FoxP3 mRNA expression and of the nuclear factor (erythroid-derived-2)- like 2 (Nrf2) downstream molecule heme oxygenase-1 (HO-1) mRNA. Finally, CD4+ CD25+ regulatory T cells were increased in the Peyer patches. In vivo, the protective effect of these regulatory cells was verified by cell transfer into recipient EAN rats. Conclusions: Our results identified a novel immunomodulatory effect of DMF through the different regions and layers of the small intestine, which led to an increase of regulatory T cells, exerting a protective role in experimental neuritis.
Collapse
Affiliation(s)
- Kalliopi Pitarokoili
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Hussein Bachir
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Melissa Sgodzai
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Grüter
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Steffen Haupeltshofer
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Duscha
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Xiomara Pedreiturria
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jeremias Motte
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
39
|
Tatara AM, Watson E, Albert ND, Kontoyiannis PD, Kontoyiannis DP, Mikos AG. A murine model of cutaneous aspergillosis for evaluation of biomaterials-based local delivery therapies. J Biomed Mater Res A 2019; 107:1867-1874. [PMID: 30882993 PMCID: PMC6626589 DOI: 10.1002/jbm.a.36671] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/28/2019] [Accepted: 02/21/2019] [Indexed: 01/10/2023]
Abstract
Cutaneous fungal infection is a challenging condition to treat that primarily afflicts immunocompromised patients. Local antifungal therapy may permit the delivery of high concentrations of antifungals directly to wounds while minimizing systemic toxicities. However, the field currently lacks suitable in vivo models. Therefore, a large cutaneous wound was created in immunosuppressed mice and inoculated with Aspergillus fumigatus. We fabricated biodegradable polymer microparticles (MPs) that were capable of locally delivering antifungal and characterized in vitro release kinetics. We compared wound bed size, fungal burden, and histological presence of fungi in mice treated with antifungal-loaded MPs. Mice with a cutaneous defect but no infection, mice with infected cutaneous defect but no treatment, and infected mice treated with blank MPs were used as controls. Infection of large wounds inhibited healing and resulted in tissue invasion in an inoculum-dependent manner. MPs were capable of releasing antifungals at concentrations above A. fumigatus Minimum Inhibitory Concentration (MIC) for at least 6 days. Wounds treated with MPs had significantly decreased size compared with no treatment (64.2% vs. 19.4% wound reduction, p = 0.002) and were not significantly different from uninfected controls (64.2% vs. 58.1%, p = 0.497). This murine model may serve to better understand cutaneous fungal infection and evaluate local biomaterials-based therapies. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1867-1874, 2019.
Collapse
Affiliation(s)
- Alexander M. Tatara
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emma Watson
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Nathaniel D. Albert
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Dimitrios P. Kontoyiannis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
40
|
Ahmadi-Beni R, Najafi A, Savar SM, Mohebbi N, Khoshnevisan A. Role of dimethyl fumarate in the treatment of glioblastoma multiforme: A review article. IRANIAN JOURNAL OF NEUROLOGY 2019; 18:127-133. [PMID: 31749934 PMCID: PMC6858600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/16/2019] [Indexed: 10/28/2022]
Abstract
Glioblastoma multiforme (GBM), the most frequent malignant and aggressive primary brain tumor, is characterized by genetically unstable heterogeneous cells, diffused growth pattern, microvascular proliferation, and resistance to chemotherapy. Extensive investigations are being carried out to identify the molecular origin of resistance to chemo- and radio-therapy in GBM and find novel targets for therapy to improve overall survival rate. Dimethyl fumarate (DMF) has been shown to be a safe drug with limited short and long-term side effects, and fumaric acid esters (FAEs), including DMF, present both anti-oxidative and anti-inflammatory activity in different cell types and tissues. DMF has also anti-tumoral and neuroprotective effects and so it could be repurposed in the treatment of this invasive tumor in the future. Here, we have reviewed DMF pharmacokinetics and different mechanisms by which DMF could have therapeutic effects on GBM.
Collapse
Affiliation(s)
- Reza Ahmadi-Beni
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Najafi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Niayesh Mohebbi
- Department of Clinical Pharmacy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Khoshnevisan
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Shaikh NS, Iyer JP, Munot YS, Mukhopadhyay PP, Raje AA, Nagaraj R, Jamdar V, Gavhane R, Lohote M, Sherkar P, Bala M, Petla R, Meru A, Umrani D, Rouduri S, Joshi S, Reddy S, Kandikere V, Bhuniya D, Kulkarni B, Mookhtiar KA. Discovery and pharmacological evaluation of indole derivatives as potent and selective RORγt inverse agonist for multiple autoimmune conditions. Bioorg Med Chem Lett 2019; 29:2208-2217. [PMID: 31272795 DOI: 10.1016/j.bmcl.2019.06.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/13/2019] [Accepted: 06/22/2019] [Indexed: 12/30/2022]
Abstract
Targeting nuclear receptor RORγ is recognized to be beneficial in multiple autoimmune disorders. We disclosed new indole analogues as potent RORγ inverse agonists. RO-2 as one of the potent and orally bioavailable compounds was evaluated in various models of autoimmune disorder. It showed potent suppression of downstream markers of RORγt activity in murine and human primary cells, ex vivo PD assay and in multiple animal models of autoimmune diseases. The results indicate the potential of these indole analogues as orally bioavailable small molecule inverse agonists of RORγt, efficacious in various Th17 driven models of autoimmune disorders.
Collapse
Affiliation(s)
- Nadim S Shaikh
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India.
| | - Jitesh P Iyer
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Yogesh S Munot
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Partha P Mukhopadhyay
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Amol A Raje
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Ranganayaki Nagaraj
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Vijay Jamdar
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Ravindra Gavhane
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Mahendra Lohote
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Prasad Sherkar
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Madhu Bala
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Rajkanth Petla
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Ashwinkumar Meru
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Dhananjay Umrani
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Sreekanth Rouduri
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Sachin Joshi
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Satyanarayan Reddy
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Vishwottam Kandikere
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Debnath Bhuniya
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Bheemashankar Kulkarni
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| | - Kasim A Mookhtiar
- Drug Discovery Facility - Pune, Advinus Therapeutics Limited, Head Office: Block No. 21 & 22, Phase II, Peenya Industrial Area, Bangalore 560058, India
| |
Collapse
|
42
|
Nally FK, De Santi C, McCoy CE. Nanomodulation of Macrophages in Multiple Sclerosis. Cells 2019; 8:cells8060543. [PMID: 31195710 PMCID: PMC6628349 DOI: 10.3390/cells8060543] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Multiple Sclerosis (MS) is a chronic demyelinating autoimmune disease primarily affecting young adults. Despite an unclear causal factor, symptoms and pathology arise from the infiltration of peripheral immune cells across the blood brain barrier. Accounting for the largest fraction of this infiltrate, macrophages are functionally heterogeneous innate immune cells capable of adopting either a pro or an anti-inflammatory phenotype, a phenomenon dependent upon cytokine milieu in the CNS. This functional plasticity is of key relevance in MS, where the pro-inflammatory state dominates the early stage, instructing demyelination and axonal loss while the later anti-inflammatory state holds a key role in promoting tissue repair and regeneration in later remission. This review highlights a potential therapeutic benefit of modulating macrophage polarisation to harness the anti-inflammatory and reparative state in MS. Here, we outline the role of macrophages in MS and look at the role of current FDA approved therapeutics in macrophage polarisation. Moreover, we explore the potential of particulate carriers as a novel strategy to manipulate polarisation states in macrophages, whilst examining how optimising macrophage uptake via nanoparticle size and functionalisation could offer a novel therapeutic approach for MS.
Collapse
Affiliation(s)
- Frances K Nally
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Chiara De Santi
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Claire E McCoy
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| |
Collapse
|
43
|
Novel CMKLR1 Inhibitors for Application in Demyelinating Disease. Sci Rep 2019; 9:7178. [PMID: 31073181 PMCID: PMC6509344 DOI: 10.1038/s41598-019-43428-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/23/2019] [Indexed: 01/25/2023] Open
Abstract
Small molecules that disrupt leukocyte trafficking have proven effective in treating patients with multiple sclerosis (MS). We previously reported that chemerin receptor chemokine-like receptor 1 (CMKLR1) is required for maximal clinical and histological experimental autoimmune encephalomyelitis (EAE); and identified CMKLR1 small molecule antagonist 2-(α-naphthoyl) ethyltrimethylammonium iodide (α-NETA) that significantly suppressed disease onset in vivo. Here we directly compared α-NETA versus FDA-approved MS drug Tecfidera for clinical efficacy in EAE; characterized key safety/toxicity parameters for α-NETA; identified structure-activity relationships among α-NETA domains and CMKLR1 inhibition; and evaluated improved α-NETA analogs for in vivo efficacy. α-NETA proved safe and superior to Tecfidera in suppressing clinical EAE. In addition, we discovered structurally differentiated α-NETA analogs (primarily ortho- or para-methoxy substitutions) with significantly improved target potency in vitro and improved efficacy in vivo. These findings suggest that α-NETA-based CMKLR1 inhibitors may prove safe and effective in treating demyelinating diseases and potentially other autoimmune disorders.
Collapse
|
44
|
SFX-01 reduces residual disability after experimental autoimmune encephalomyelitis. Mult Scler Relat Disord 2019; 30:257-261. [DOI: 10.1016/j.msard.2019.02.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/01/2018] [Accepted: 02/25/2019] [Indexed: 11/22/2022]
|
45
|
Ranea-Robles P, Launay N, Ruiz M, Calingasan NY, Dumont M, Naudí A, Portero-Otín M, Pamplona R, Ferrer I, Beal MF, Fourcade S, Pujol A. Aberrant regulation of the GSK-3β/NRF2 axis unveils a novel therapy for adrenoleukodystrophy. EMBO Mol Med 2019; 10:emmm.201708604. [PMID: 29997171 PMCID: PMC6079538 DOI: 10.15252/emmm.201708604] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The nuclear factor erythroid 2‐like 2 (NRF2) is the master regulator of endogenous antioxidant responses. Oxidative damage is a shared and early‐appearing feature in X‐linked adrenoleukodystrophy (X‐ALD) patients and the mouse model (Abcd1 null mouse). This rare neurometabolic disease is caused by the loss of function of the peroxisomal transporter ABCD1, leading to an accumulation of very long‐chain fatty acids and the induction of reactive oxygen species of mitochondrial origin. Here, we identify an impaired NRF2 response caused by aberrant activity of GSK‐3β. We find that GSK‐3β inhibitors can significantly reactivate the blunted NRF2 response in patients’ fibroblasts. In the mouse models (Abcd1− and Abcd1−/Abcd2−/− mice), oral administration of dimethyl fumarate (DMF/BG12/Tecfidera), an NRF2 activator in use for multiple sclerosis, normalized (i) mitochondrial depletion, (ii) bioenergetic failure, (iii) oxidative damage, and (iv) inflammation, highlighting an intricate cross‐talk governing energetic and redox homeostasis in X‐ALD. Importantly, DMF halted axonal degeneration and locomotor disability suggesting that therapies activating NRF2 hold therapeutic potential for X‐ALD and other axonopathies with impaired GSK‐3β/NRF2 axis.
Collapse
Affiliation(s)
- Pablo Ranea-Robles
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat Barcelona, Spain.,CIBERER U759, Center for Biomedical Research on Rare Diseases ISCIII, Barcelona, Spain
| | - Nathalie Launay
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat Barcelona, Spain.,CIBERER U759, Center for Biomedical Research on Rare Diseases ISCIII, Barcelona, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat Barcelona, Spain.,CIBERER U759, Center for Biomedical Research on Rare Diseases ISCIII, Barcelona, Spain
| | - Noel Ylagan Calingasan
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Magali Dumont
- UMR S 1127, Inserm, U1127, CNRS, UMR 7225, Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, UPMC Université Paris 06, Paris, France
| | - Alba Naudí
- Experimental Medicine Department, University of Lleida-IRB Lleida, Lleida, Spain
| | - Manuel Portero-Otín
- Experimental Medicine Department, University of Lleida-IRB Lleida, Lleida, Spain
| | - Reinald Pamplona
- Experimental Medicine Department, University of Lleida-IRB Lleida, Lleida, Spain
| | - Isidre Ferrer
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, L'Hospitalet de Llobregat Barcelona, Spain.,Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,IDIBELL-Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
| | - M Flint Beal
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat Barcelona, Spain .,CIBERER U759, Center for Biomedical Research on Rare Diseases ISCIII, Barcelona, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat Barcelona, Spain .,CIBERER U759, Center for Biomedical Research on Rare Diseases ISCIII, Barcelona, Spain.,Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
46
|
Pouzol L, Piali L, Bernard CC, Martinic MM, Steiner B, Clozel M. Therapeutic Potential of Ponesimod Alone and in Combination with Dimethyl Fumarate in Experimental Models of Multiple Sclerosis. INNOVATIONS IN CLINICAL NEUROSCIENCE 2019; 16:22-30. [PMID: 31214480 PMCID: PMC6538399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Background: Despite the recent approval of new oral therapies for the treatment of multiple sclerosis (MS), a significant percentage of patients are still not free from disease activity. In view of the complex pathogenesis and the relapsing and progressive nature of MS, combination therapy, a classical approach to treat many chronic diseases, could improve disease control over monotherapy. Ponesimod, a selective and rapidly reversible sphingosine-1-phosphate receptor Type 1 (S1P1) modulator, currently in Phase III clinical trial stage in relapsing MS (RMS), and dimethyl fumarate (DMF) would potentially be an ideal combination due to their differing mechanisms of action and oral administration. Objective: The goal of the study was to evaluate the therapeutic effect of ponesimod monotherapy and investigate the potential additive, or synergistic, activity of ponesimod-DMF combination therapy in experimental autoimmune encephalomyelitis (EAE) animal models of MS. Methods: Efficacy was evaluated in the myelin oligodendrocyte glycoprotein (MOG)-induced EAE model in C57BL/6 mice (ponesimod monotherapy) and in the myelin basic protein (MBP)-induced EAE model in Lewis rats (monotherapies and combination therapy). The principal readout was the clinical score assessing paralysis. Additional readouts, such as histopathology, survival, and disease prevalence, were generated in parallel when applicable. Results: Ponesimod monotherapy in the mouse EAE model showed significant efficacy in both preventative and therapeutic settings. In the rat EAE model, ponesimod demonstrated significant dose-dependent efficacy on clinical scores, while DMF showed only modest activity. Combination therapy synergistically reduced the severity and prevalence of disease. Only the combination treatment of ponesimod and DMF fully suppressed clinical disease activity by the end of the study. Conclusion: The results support the potential therapeutic benefits of combining ponesimod with DMF to improve disease activity control in patients with MS. Additionally, the results suggest that combining ponesimod with other oral agents that have different mechanisms of action might also be therapeutically beneficial to patients with MS.
Collapse
Affiliation(s)
- Laetitia Pouzol
- Drs. Pouzol and Clozel were with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and are presently with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Piali was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and is presently with Hoffmann la Roche in Basel, Switzerland
- Dr. Bernard is with Monash University, Faculty of Medicine, Nursing & Health Sciences in Melbourne, Australia
- Dr. Martinic is with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Steiner was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research
| | - Luca Piali
- Drs. Pouzol and Clozel were with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and are presently with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Piali was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and is presently with Hoffmann la Roche in Basel, Switzerland
- Dr. Bernard is with Monash University, Faculty of Medicine, Nursing & Health Sciences in Melbourne, Australia
- Dr. Martinic is with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Steiner was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research
| | - Claude Ca Bernard
- Drs. Pouzol and Clozel were with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and are presently with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Piali was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and is presently with Hoffmann la Roche in Basel, Switzerland
- Dr. Bernard is with Monash University, Faculty of Medicine, Nursing & Health Sciences in Melbourne, Australia
- Dr. Martinic is with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Steiner was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research
| | - Marianne M Martinic
- Drs. Pouzol and Clozel were with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and are presently with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Piali was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and is presently with Hoffmann la Roche in Basel, Switzerland
- Dr. Bernard is with Monash University, Faculty of Medicine, Nursing & Health Sciences in Melbourne, Australia
- Dr. Martinic is with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Steiner was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research
| | - Beat Steiner
- Drs. Pouzol and Clozel were with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and are presently with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Piali was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and is presently with Hoffmann la Roche in Basel, Switzerland
- Dr. Bernard is with Monash University, Faculty of Medicine, Nursing & Health Sciences in Melbourne, Australia
- Dr. Martinic is with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Steiner was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research
| | - Martine Clozel
- Drs. Pouzol and Clozel were with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and are presently with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Piali was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research, and is presently with Hoffmann la Roche in Basel, Switzerland
- Dr. Bernard is with Monash University, Faculty of Medicine, Nursing & Health Sciences in Melbourne, Australia
- Dr. Martinic is with Idorsia Pharmaceuticals Ltd. in Allschwil, Switzerland
- Dr. Steiner was with Actelion Pharmaceuticals Ltd. in Allschwil, Switzerland, at the time of this research
| |
Collapse
|
47
|
Yadav SK, Soin D, Ito K, Dhib-Jalbut S. Insight into the mechanism of action of dimethyl fumarate in multiple sclerosis. J Mol Med (Berl) 2019; 97:463-472. [PMID: 30820593 DOI: 10.1007/s00109-019-01761-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/26/2022]
Abstract
Dimethyl fumarate (DMF) is an oral, disease-modifying agent for the treatment of relapsing-remitting multiple sclerosis (RRMS). However, details regarding its mode of action are still emerging. It is believed that the mode of action of DMF involves both nuclear factor erythroid-derived 2-related factor (Nrf2)-dependent and independent pathways, which lead to an anti-inflammatory immune response due to type II myeloid cell and Th2 cell differentiation and neuroprotection. In this review, we will focus on the molecular and signaling effects of DMF that lead to changes in peripheral immune cell composition and function, alteration in CNS cell-specific functions, and effect on the blood-brain barrier.
Collapse
Affiliation(s)
- Sudhir Kumar Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Devika Soin
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
48
|
Deleu D, Mesraoua B, Canibaño B, Melikyan G, Al Hail H, El-Sheikh L, Ali M, Al Hussein H, Ibrahim F, Hanssens Y. Oral disease-modifying therapies for multiple sclerosis in the Middle Eastern and North African (MENA) region: an overview. Curr Med Res Opin 2019; 35:249-260. [PMID: 29764226 DOI: 10.1080/03007995.2018.1476334] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
BACKGROUND The introduction of new disease-modifying therapies (DMTs) for remitting-relapsing multiple sclerosis (RRMS) has considerably transformed the landscape of therapeutic opportunities for this chronic disabling disease. Unlike injectable drugs, oral DMTs promote patient satisfaction and increase therapeutic adherence. REVIEW This article reviews the salient features about the mode of action, efficacy, safety, and tolerability profile of approved oral DMTs in RRMS, and reviews their place in clinical algorithms in the Middle East and North Africa (MENA) region. A systematic review was conducted using a comprehensive search of MEDLINE, PubMed, Cochrane Database of Systematic Reviews (period January 1, 1995-January 31, 2018). Additional searches of the American Academy of Neurology and European Committee for Treatment and Research in Multiple Sclerosis abstracts from 2012-2017 were performed, in addition to searches of the Food and Drug Administration and European Medicines Agency websites, to obtain relevant safety information on these DMTs. CONCLUSIONS Four oral DMTs: fingolimod, teriflunomide, dimethyl fumarate, and cladribine have been approved by the regulatory agencies. Based on the number needed to treat (NNT), the potential role of these DMTs in the management of active and highly active or rapidly evolving RRMS is assessed. Finally, the place of the oral DMTs in clinical algorithms in the MENA region is reviewed.
Collapse
Affiliation(s)
- Dirk Deleu
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Boulenouar Mesraoua
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Beatriz Canibaño
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Gayane Melikyan
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Hassan Al Hail
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Lubna El-Sheikh
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Musab Ali
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Hassan Al Hussein
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Faiza Ibrahim
- a Department of Neurology , Neuroscience Institute, Hamad Medical Corporation , Doha , State of Qatar
| | - Yolande Hanssens
- b Department of Clinical Services Unit , Corporate Pharmacy, Hamad Medical Corporation , Doha , State of Qatar
| |
Collapse
|
49
|
Staurengo-Ferrari L, Badaro-Garcia S, Hohmann MSN, Manchope MF, Zaninelli TH, Casagrande R, Verri WA. Contribution of Nrf2 Modulation to the Mechanism of Action of Analgesic and Anti-inflammatory Drugs in Pre-clinical and Clinical Stages. Front Pharmacol 2019; 9:1536. [PMID: 30687097 PMCID: PMC6337248 DOI: 10.3389/fphar.2018.01536] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
Despite the progress that has occurred in recent years in the development of therapies to treat painful and inflammatory diseases, there is still a need for effective and potent analgesics and anti-inflammatory drugs. It has long been known that several types of antioxidants also possess analgesic and anti-inflammatory properties, indicating a strong relationship between inflammation and oxidative stress. Understanding the underlying mechanisms of action of anti-inflammatory and analgesic drugs, as well as essential targets in disease physiopathology, is essential to the development of novel therapeutic strategies. The Nuclear factor-2 erythroid related factor-2 (Nrf2) is a transcription factor that regulates cellular redox status through endogenous antioxidant systems with simultaneous anti-inflammatory activity. This review summarizes the molecular mechanisms and pharmacological actions screened that link analgesic, anti-inflammatory, natural products, and other therapies to Nrf2 as a regulatory system based on emerging evidences from experimental disease models and new clinical trial data.
Collapse
Affiliation(s)
- Larissa Staurengo-Ferrari
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Stephanie Badaro-Garcia
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Miriam S. N. Hohmann
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Marília F. Manchope
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Tiago H. Zaninelli
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Rubia Casagrande
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Estadual de Londrina, Londrina, Brazil
| | - Waldiceu A. Verri
- Departamento de Patologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
50
|
Singh N, Saha L, Kumari P, Singh J, Bhatia A, Banerjee D, Chakrabarti A. Effect of dimethyl fumarate on neuroinflammation and apoptosis in pentylenetetrazol kindling model in rats. Brain Res Bull 2019; 144:233-245. [DOI: 10.1016/j.brainresbull.2018.11.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/05/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
|